scholarly journals Altered contractility and [Ca2+]i homeostasis in phospholemman-deficient murine myocytes: role of Na+/Ca2+ exchange

2006 ◽  
Vol 291 (5) ◽  
pp. H2199-H2209 ◽  
Author(s):  
Amy L. Tucker ◽  
Jianliang Song ◽  
Xue-Qian Zhang ◽  
JuFang Wang ◽  
Belinda A. Ahlers ◽  
...  

Phospholemman (PLM) regulates contractility and Ca2+ 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+/Ca2+ exchanger, sarcoplasmic reticulum (SR) Ca2+-ATPase, α1-subunit of Na+-K+-ATPase, and calsequestrin levels in PLM-null myocytes. At 5 mM extracellular Ca2+ concentration ([Ca2+]o), contraction and cytosolic [Ca2+] ([Ca2+]i) transient amplitudes and SR Ca2+ contents in PLM-null myocytes were significantly ( P < 0.0004) higher than wild-type myocytes, whereas the converse was true at 0.6 mM [Ca2+]o. This pattern of contractile and [Ca2+]i transient abnormalities in PLM-null myocytes mimics that observed in adult rat myocytes overexpressing the cardiac Na+/Ca2+ exchanger. Indeed, we have previously reported that Na+/Ca2+ 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 Ca2+ 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 Ca2+ fluxes, likely by modulating Na+/Ca2+ exchange activity.

2002 ◽  
Vol 93 (4) ◽  
pp. 1310-1317 ◽  
Author(s):  
Xue-Qian Zhang ◽  
Jianliang Song ◽  
Lois L. Carl ◽  
Weixing Shi ◽  
Anwer Qureshi ◽  
...  

The effects of 6–8 wk of high-intensity sprint training (HIST) on rat myocyte contractility and intracellular Ca2+ concentration ([Ca2+]i) transients were investigated. Compared with sedentary (Sed) myocytes, HIST induced a modest (5%) but significant ( P < 0.0005) increase in cell length with no changes in cell width. In addition, the percentage of myosin heavy chain α-isoenzyme increased significantly ( P < 0.02) from 0.566 ± 0.077% in Sed rats to 0.871 ± 0.006% in HIST rats. At all three (0.6, 1.8, and 5 mM) extracellular Ca2+concentrations ([Ca2+]o) examined, maximal shortening amplitudes and maximal shortening velocities were significantly ( P < 0.0001) lower and half-times of relaxation were significantly ( P < 0.005) longer in HIST myocytes. HIST myocytes had significantly ( P < 0.0001) higher diastolic [Ca2+]i levels. Compared with Sed myocytes, systolic [Ca2+]ilevels in HIST myocytes were higher at 0.6 mM [Ca2+]o, similar at 1.8 mM [Ca2+]o, and lower at 5 mM [Ca2+]o. The amplitudes of [Ca2+]i transients were significantly ( P < 0.0001) lower in HIST myocytes. Half-times of [Ca2+]i transient decline, an estimate of sarcoplasmic reticulum (SR) Ca2+ uptake activity, were not different between Sed and HIST myocytes. Compared with Sed hearts, Western blots demonstrated a significant ( P < 0.03) threefold decrease in Na+/Ca2+ exchanger, but SR Ca2+-ATPase and calsequestrin protein levels were unchanged in HIST hearts. We conclude that HIST effected diminished myocyte contractile function and [Ca2+]itransient amplitudes under the conditions studied. We speculate that downregulation of Na+/Ca2+ exchanger may partly account for the decreased contractility in HIST myocytes.


2017 ◽  
Vol 313 (4) ◽  
pp. H810-H827 ◽  
Author(s):  
C. M. Kofron ◽  
T. Y. Kim ◽  
M. E. King ◽  
A. Xie ◽  
F. Feng ◽  
...  

Cardiac fibroblasts (CFs) are known to regulate cardiomyocyte (CM) function in vivo and in two-dimensional in vitro cultures. This study examined the effect of CF activation on the regulation of CM electrical activity in a three-dimensional (3-D) microtissue environment. Using a scaffold-free 3-D platform with interspersed neonatal rat ventricular CMs and CFs, Gq-mediated signaling was selectively enhanced in CFs by Gαq adenoviral infection before coseeding with CMs in nonadhesive hydrogels. After 3 days, the microtissues were analyzed by signaling assay, histological staining, quantitative PCR, Western blots, optical mapping with voltage- or Ca2+-sensitive dyes, and microelectrode recordings of CF resting membrane potential (RMPCF). Enhanced Gq signaling in CFs increased microtissue size and profibrotic and prohypertrophic markers. Expression of constitutively active Gαq in CFs prolonged CM action potential duration (by 33%) and rise time (by 31%), prolonged Ca2+ transient duration (by 98%) and rise time (by 65%), and caused abnormal electrical activity based on depolarization-induced automaticity. Constitutive Gq activation in CFs also depolarized RMPCF from –33 to −20 mV and increased connexin 43 and connexin 45 expression. Computational modeling confers that elevated RMPCF and increased cell-cell coupling between CMs and CFs in a 3-D environment could lead to automaticity. In conclusion, our data demonstrate that CF activation alone is capable of altering action potential and Ca2+ transient characteristics of CMs, leading to proarrhythmic electrical activity. Our results also emphasize the importance of a 3-D environment where cell-cell interactions are prevalent, underscoring that CF activation in 3-D tissue plays a significant role in modulating CM electrophysiology and arrhythmias. NEW & NOTEWORTHY In a three-dimensional microtissue model, which lowers baseline activation of cardiac fibroblasts but enables cell-cell, paracrine, and cell-extracellular matrix interactions, we demonstrate that selective cardiac fibroblast activation by enhanced Gq signaling, a pathophysiological trigger in the diseased heart, modulates cardiomyocyte electrical activity, leading to proarrhythmogenic automaticity.


2014 ◽  
Vol 307 (4) ◽  
pp. C338-C348 ◽  
Author(s):  
Hongguang Wei ◽  
J.-P. Jin

We previously reported a point mutation substituting Cys for Arg111 in the highly conserved troponin T (TnT)-contacting helix of cardiac troponin I (cTnI) in wild turkey hearts (Biesiadecki et al. J Biol Chem 279: 13825–13832, 2004). This dominantly negative TnI-TnT interface mutation decreases the binding affinity of cTnI for TnT, impairs diastolic function, and blunts the β-adrenergic response of cardiac muscle (Wei et al. J Biol Chem 285: 27806–27816, 2010). Here we further investigate cellular phenotypes of transgenic mouse cardiomyocytes expressing the equivalent mutation cTnI-K118C. Functional studies were performed on single adult cardiomyocytes after recovery in short-term culture from isolation stress. The amplitude of contraction and the velocities of shortening and relengthening were lower in cTnI-K118C cardiomyocytes than wild-type controls. The intracellular Ca2+ transient was slower in cTnI-K118C cardiomyocytes than wild-type cells. cTnI-K118C cardiomyocytes also showed a weaker β-adrenergic response. The resting length of cTnI-K118C cardiomyocytes was significantly greater than that of age-matched wild-type cells, with no difference in cell width. The resting sarcomere was not longer, but slightly shorter, in cTnI-K118C cardiomyocytes than wild-type cells, indicating longitudinal addition of sarcomeres. More tri- and quadrinuclei cardiomyocytes were found in TnI-K118C than wild-type hearts, suggesting increased nuclear divisions. Whole-genome mRNA array and Western blots detected an increased expression of leukemia inhibitory factor receptor-β in the hearts of 2-mo-old cTnI-K118C mice, suggesting a signaling pathway responsible for the potent effect of cTnI-K118C mutation on early remodeling in cardiomyocytes.


2001 ◽  
Vol 90 (4) ◽  
pp. 1327-1333 ◽  
Author(s):  
Korinne N. Jew ◽  
M. Charlotte Olsson ◽  
Eric A. Mokelke ◽  
Bradley M. Palmer ◽  
Russell L. Moore

The effect of endurance run training on outward K+ currents with rapidly inactivating ( I to) and sustained or slowly inactivating ( I sus) characteristics was examined in left ventricular (LV) cardiocytes isolated from sedentary (Sed) and treadmill-trained (Tr) female Sprague-Dawley rats. Isolated LV cardiocytes were used in whole cell patch-clamp studies to characterize whole cell I to and I sus. Peak I to was greatest in cells isolated from the Tr group. When I to was corrected for cell capacitance to yield a current density, most, but not all, of the Sed vs. Tr differences in I to magnitude were eliminated. Regardless of how I to was expressed (e.g., I to or I todensity), the time required to achieve a peak value was markedly shortened in the cardiocytes isolated from the Tr group. Training elicited a reduction in I sus density. Action potential characteristics were determined in Sed and Tr cardiocytes in primary culture. Training did not affect resting membrane potential, whereas peak membrane potential was reduced and time to peak membrane potential was prolonged in the Tr group. In addition, time to 50% repolarization was significantly increased in cells from the Tr group. Collectively, these data indicate that I to and I sus characteristics are altered by training in isolated LV cardiocytes. These alterations in I to and I sus may be responsible, at least in part, for the training-induced alterations in action potential configuration in cardiocytes in primary culture.


1993 ◽  
Vol 265 (2) ◽  
pp. H747-H754 ◽  
Author(s):  
O. Ellingsen ◽  
A. J. Davidoff ◽  
S. K. Prasad ◽  
H. J. Berger ◽  
J. P. Springhorn ◽  
...  

We studied primary short-term cultures of adult rat ventricular myocytes in defined medium to determine whether phenotype and electromechanical function are maintained in rod-shaped, quiescent cells. Although > 80% of the myocytes retained their rod-shaped in vivo morphology for up to 72 h, contractile function as measured by cell edge motion declined 30-50% from 6 to 24 h, paralleling a 68% shortening of action potential duration. From 24 to 72 h, contractility remained unchanged. Ca2+ channel current density increased 55% after 24-48 h and then returned to the level of freshly isolated cells (9 +/- 1 pA/pF, mean +/- SE). Resting membrane potential (-71 +/- 1 mV) and action potential overshoot (34 +/- 3 mV) did not change. The ratio of alpha- to beta-myosin heavy chain mRNA and the level of cardiac alpha-actin mRNA were maintained for 8 days. Thus quiescent adult rat ventricular myocytes in defined medium undergo extensive phenotypic adaptation within 72 h of isolation, despite maintenance of a rod-shaped morphology and stable levels of contractile protein mRNA, which may limit their suitability for electrophysiological and contractile function studies.


2018 ◽  
Vol 243 (8) ◽  
pp. 708-714
Author(s):  
Hong Cao ◽  
Xin Wang ◽  
Shaozheng Ying ◽  
Congxin Huang

AMP-activated protein kinase-α2 is the main catalytic subunit of the heart, which is mainly located in cardiac myocytes. The effect of AMP-activated protein kinase-α2 on the cardiac electrophysiology is barely studied. From the previous study, it is possible that AMP-activated protein kinase-α2 may have some effect on the electrophysiology of the heart. To prove the hypothesis, we used the AMP-activated protein kinase-α2 knockout (AMPKα2−/−) mice to estimate the electrophysiological characteristics of AMPKα2−/− mice and try to find the mechanism between them. We used AMP-activated protein kinase-α2 gene knockout (AMPKα2−/−) mice and control wild-type mice as the experimental animals. In the experiment, we measured the monophasic action potential duration and test the inducibility to ventricular arrhythmia in isolated mice heart with and without β-adrenoceptor antagonist metoprolol. Meanwhile, plasma concentration of catecholamine was collected. We found that AMPKα2−/− significantly shortened 90% repolarization of monophasic action potential (MAP) (MAPD90) than wild-type (47.4 ± 2.6 ms vs. 55.5 ± 2.4 ms, n = 10, P < 0.05) and were more vulnerable to be induced to ventricular arrhythmias (70% (7/10) vs. 10% (1/10), P < 0.05), accompanied by the higher concentration of catecholamine (epinephrine: 1.75 ± 0.18 nmol/L vs. 0.68 ± 0.10 nmol/L n = 10, P < 0.05; norepinephrine: 9.56 ± 0.71 nmol/L vs. 2.52 ± 0.31 nmol/L n = 10, P < 0.05). The shortening of MAPD90 and increased inducibility to ventricular arrhythmias of AMPKα2−/− could almost be abolished when perfusion with β-adrenoceptor antagonist metoprolol. It indicated that the β-adrenoceptor activation resulting from catecholamine release was mainly responsible for the relating changes of electrophysiology of AMPKα2−/−. It had great clinical significance, as in patients who had problem with AMP-activated protein kinase-α2 gene, we might use β-adrenoceptor antagonists as the prevention of arrhythmias in future. Impact statement As far as we know, this is the first time the role of AMP-activated protein kinase-α2 (AMPKα2) on the cardiac electrophysiology is explored, and we found that the β-adrenoceptor activation resulting from catecholamine release was mainly responsible for the changes of electrophysiology related to the absence of AMPKα2. This has great clinical significance, as in patients who have problems with AMPKα2 gene, we may use β-adrenoceptor antagonists for the prevention of arrhythmias in future.


2015 ◽  
Vol 146 (6) ◽  
pp. 509-525 ◽  
Author(s):  
Tarek Ammar ◽  
Wei Lin ◽  
Amanda Higgins ◽  
Lawrence J. Hayward ◽  
Jean-Marc Renaud

The diaphragm muscle of hyperkalemic periodic paralysis (HyperKPP) patients and of the M1592V HyperKPP mouse model rarely suffers from the myotonic and paralytic symptoms that occur in limb muscles. Enigmatically, HyperKPP diaphragm expresses the mutant NaV1.4 channel and, more importantly, has an abnormally high Na+ influx similar to that in extensor digitorum longus (EDL) and soleus, two hindlimb muscles suffering from the robust HyperKPP abnormalities. The objective was to uncover the physiological mechanisms that render HyperKPP diaphragm asymptomatic. A first mechanism involves efficient maintenance of resting membrane polarization in HyperKPP diaphragm at various extracellular K+ concentrations compared with larger membrane depolarizations in HyperKPP EDL and soleus. The improved resting membrane potential (EM) results from significantly increased Na+ K+ pump electrogenic activity, and not from an increased protein content. Action potential amplitude was greater in HyperKPP diaphragm than in HyperKPP soleus and EDL, providing a second mechanism for the asymptomatic behavior of the HyperKPP diaphragm. One suggested mechanism for the greater action potential amplitude is lower intracellular Na+ concentration because of greater Na+ K+ pump activity, allowing better Na+ current during the action potential depolarization phase. Finally, HyperKPP diaphragm had a greater capacity to generate force at depolarized EM compared with wild-type diaphragm. Action potential amplitude was not different between wild-type and HyperKPP diaphragm. There was also no evidence for an increased activity of the Na+–Ca2+ exchanger working in the reverse mode in the HyperKPP diaphragm compared with the wild-type diaphragm. So, a third mechanism remains to be elucidated to fully understand how HyperKPP diaphragm generates more force compared with wild type. Although the mechanism for the greater force at depolarized resting EM remains to be determined, this study provides support for the modulation of the Na+ K+ pump as a component of therapy to alleviate weakness in HyperKPP.


2012 ◽  
Vol 111 (suppl_1) ◽  
Author(s):  
Toru Akaike ◽  
Gang Lu ◽  
Yibin Wang ◽  
Hongmei Ruan

Regulation of sarcoplasmic reticulum (SR) calcium-ATPase (SERCA) activity is critical for calcium homeostasis in cardiomyocytes, and has a major impact on contractility and cellular viability of cardiomyocytes. The key regulators for SERCA activity include protein kinases, cAMP dependent protein kinase A and calcium/calmodulin dependent protein kinase II, and protein phosphatase 1. In this report, we have discovered that protein phosphatase 2Ce (PP2Ce) is a novel serine/threonine protein phosphatase specifically targeted to SR membrane in cardiomyocytes. PP2Ce was detected to interact with phosphlamban in heart. Recombinant PP2Ce protein showed a potent and specific activity towards the calcium/calmodulin dependent protein kinase II dependent phospholamban phosphorylation at threonin 17 site with no significant activity to cAMP dependent protein kinase A dependent phospholamban phosphorylation at serine 16 site. Expression of PP2Ce blunted β-adrenergic stimulated increase of phospholamban phosphorylation without affecting phosphorylation of ryanodine recepter or troponin I. PP2Ce expression reduced β-adrenergic stimulated intracellular calcium transient in isolated adult rabbit ventricular myocytes, and promoted hydrogen peroxide induced cell death in cultured neonatal rat ventricular myocytes. Transgenic mice with cardiac specific expression of PP2Ce showed no significant basal phenotype. However, in isolated perfusion heart preparation, β-adrenergic stimulated contractility was significant reduced in PP2Ce transgenic hearts comparing to wild type controls. Furthermore, we observed significantly larger infarct sizes and more impaired functional recovery following global ischemia/reperfusion injury in the transgenic hearts comparing to wild type controls. Therefore, PP2Ce is a novel component of SR calcium regulatory network that has a potentially important role in cell death regulation and cardiac contractility.


2011 ◽  
Vol 300 (3) ◽  
pp. H859-H868 ◽  
Author(s):  
JuFang Wang ◽  
Erhe Gao ◽  
Joseph Rabinowitz ◽  
Jianliang Song ◽  
Xue-Qian Zhang ◽  
...  

Phospholemman (PLM), when phosphorylated at serine 68, relieves its inhibition on Na+-K+-ATPase but inhibits Na+/Ca2+ 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 α1- and α2-subunits of Na+-K+-ATPase, NCX1, and sarco(endo)plasmic reticulum Ca2+-ATPase between KO-GFP and KO-S68E LV homogenates. Compared with KO-GFP myocytes, Na+/Ca2+ 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 (APD90) was shortened in KO-S68E myocytes. Isoproterenol (Iso; 1 μM) increased APD90 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 [Ca2+]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+/Ca2+ exchanger.


1995 ◽  
Vol 268 (2) ◽  
pp. H926-H934 ◽  
Author(s):  
E. A. Aiello ◽  
M. P. Walsh ◽  
W. C. Cole

The effect of adenosine 3',5'-cyclic monophosphate-dependent protein kinase (PKA) activity on 4-aminopyridine (4-AP)-sensitive delayed rectifier current (IdK) in isolated rabbit portal vein smooth muscle cells was studied via whole cell voltage clamp (20–22 degrees C). A threefold increase in 4-AP-sensitive (5 mM) IdK was recorded after gaining cell access during dialysis with 5 mM intracellular ATP and internal Ca2+ buffered to a low level with 5 mM ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid. Dialysis with the nonhydrolyzable ATP analogue 5'-adenylylimidodiphosphate (5 mM) or the specific peptide inhibitor of PKA (PKI; 10 microM) reduced current runup by 50 and 70%, respectively. Delayed dialysis with PKI reversed runup and inhibited IdK to below initial levels. Forskolin (1 microM) caused a reversible increase in IdK, which was inhibited by 4-AP (5 mM). Isoproterenol (1 microM) reversibly enhanced IdK; the increase was sensitive to propranolol (2 microM) and 4-AP (5 mM) and was prevented by dialysis with PKI (10 microM). IdK was enhanced over the entire voltage range of activation and associated with a negative shift in reversal potential of net whole cell current, consistent with hyperpolarization of resting membrane potential. The data provide the first evidence for a signal transduction mechanism involving beta-adrenoceptors, adenylate cyclase, and a phosphotransferase reaction mediated by PKA for the regulation of delayed rectifier K+ channels in vascular smooth muscle.


Sign in / Sign up

Export Citation Format

Share Document