Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sanda Despa is active.

Publication


Featured researches published by Sanda Despa.


Circulation | 2002

Intracellular Na+ Concentration Is Elevated in Heart Failure But Na/K Pump Function Is Unchanged

Sanda Despa; Mohammed A. Islam; Christopher R. Weber; Steven M. Pogwizd; Donald M. Bers

Background—Intracellular sodium concentration ([Na+]i) modulates cardiac contractile and electrical activity through Na/Ca exchange (NCX). Upregulation of NCX in heart failure (HF) may magnify the functional impact of altered [Na+]i. Methods and Results—We measured [Na+]i by using sodium binding benzofuran isophthalate in control and HF rabbit ventricular myocytes (HF induced by aortic insufficiency and constriction). Resting [Na+]i was 9.7±0.7 versus 6.6±0.5 mmol/L in HF versus control. In both cases, [Na+]i increased by ≈2 mmol/L when myocytes were stimulated (0.5 to 3 Hz). To identify the mechanisms responsible for [Na+]i elevation in HF, we measured the [Na+]i dependence of Na/K pump–mediated Na+ extrusion. There was no difference in Vmax (8.3±0.7 versus 8.0±0.8 mmol/L/min) or Km (9.2±1.0 versus 9.9±0.8 mmol/L in HF and control, respectively). Therefore, at measured [Na+]i levels, the Na/K pump rate is actually higher in HF. However, resting Na+ influx was twice as high in HF versus control (2.3±0.3 versus 1.1±0.2 mmol/L/min), primarily the result of a tetrodotoxin-sensitive pathway. Conclusions—Myocyte [Na+]i is elevated in HF as a result of higher diastolic Na+ influx (with unaltered Na/K-ATPase characteristics). In HF, the combined increased [Na+]i, decreased Ca2+ transient, and prolonged action potential all profoundly affect cellular Ca2+ regulation, promoting greater Ca2+ influx through NCX during action potentials. Notably, the elevated [Na+]i may be critical in limiting the contractile dysfunction observed in HF.


Cardiovascular Research | 2003

Intracellular Na+ regulation in cardiac myocytes

Donald M. Bers; William H. Barry; Sanda Despa

Intracellular [Na+] ([Na+]i) is regulated in cardiac myocytes by a balance of Na+ influx and efflux mechanisms. In the normal cell there is a large steady state electrochemical gradient favoring Na+ influx. This potential energy is used by numerous transport mechanisms, including Na+ channels and transporters which couple Na+ influx to either co- or counter-transport of other ions and solutes. Six sarcolemmal Na+ influx pathways are discussed in relatively quantitative terms: Na+ channels, Na+/Ca2+ exchange, Na+/H+ exchange, Na+/Mg2+ exchange, Na+/HCO3- cotransport and Na+/K+/2Cl- cotransport. Under normal conditions Na+/Ca2+ exchange and Na+ channels are the dominant Na+ influx pathways, but other transporters may become increasingly important during altered conditions (e.g. acidosis or cell volume stress). Mitochondria also exhibit Na+/Ca2+ antiporter and Na+/H+ exchange activity that are important in mitochondrial function. These coupled fluxes of Na+ with Ca2+, H+ and HCO3- make the detailed understanding of [Na+]i regulation pivotal to the understanding of both cardiac excitation-contraction coupling and pH regulation. The Na+/K+-ATPase is the main route for Na+ extrusion from cells and [Na+]i is a primary regulator under physiological conditions. [Na+]i is higher in rat than rabbit ventricular myocytes and the reason appears to be higher Na+ influx in rat with a consequent rise in Na+/K+-ATPase activity (rather than lower Na+/K+-ATPase function in rat). This has direct functional consequences. There may also be subcellular [Na+]i gradients locally in ventricular myocytes and this may also have important functional implications. Thus, the balance of Na+ fluxes in heart cells may be complex, but myocyte Na+ regulation is functionally important and merits focused attention as in this issue.


Circulation Research | 2005

Phospholemman-Phosphorylation Mediates the β-Adrenergic Effects on Na/K Pump Function in Cardiac Myocytes

Sanda Despa; Julie Bossuyt; Fei Han; Kenneth S. Ginsburg; Li Guo Jia; Howard Kutchai; Amy L. Tucker; Donald M. Bers

Cardiac sympathetic stimulation activates β-adrenergic (β-AR) receptors and protein kinase A (PKA) phosphorylation of proteins involved in myocyte Ca regulation. The Na/K-ATPase (NKA) is essential in regulating intracellular [Na] ([Na]i), which in turn affects [Ca]i via Na/Ca exchange. However, how PKA modifies NKA function is unknown. Phospholemman (PLM), a member of the FXYD family of proteins that interact with NKA in various tissues, is a major PKA substrate in heart. Here we tested the hypothesis that PLM phosphorylation is responsible for the PKA effects on cardiac NKA function using wild-type (WT) and PLM knockout (PLM-KO) mice. We measured NKA-mediated [Na]i decline and current (IPump) to assess β-AR effects on NKA function in isolated myocytes. In WT myocytes, 1 &mgr;mol/L isoproterenol (ISO) increased PLM phosphorylation and stimulated NKA activity mainly by increasing its affinity for internal Na (Km decreased from 18.8±1.4 to 13.6±1.5 mmol/L), with no significant effect on the maximum pump rate. This led to a significant decrease in resting [Na]i (from 12.5±1.8 to 10.5±1.4 mmol/L). In PLM-KO mice under control conditions Km (14.2±1.5 mmol/L) was lower than in WT, but comparable to that for WT in the presence of ISO. Furthermore, ISO had no significant effect on NKA function in PLM-KO mice. ATPase activity in sarcolemmal vesicles also showed a lower Km(Na) in PLM-KO versus WT (12.9±0.9 versus 16.2±1.5). Thus, PLM inhibits NKA activity by decreasing its [Na]i affinity, and this inhibitory effect is relieved by PKA activation. We conclude that PLM modulates the NKA function in a manner similar to the way phospholamban affects the related SR Ca-ATPase (inhibition of transport substrate affinity, that is relieved by phosphorylation).


Annals of the New York Academy of Sciences | 2006

Regulation of Ca2+ and Na+ in Normal and Failing Cardiac Myocytes

Donald M. Bers; Sanda Despa; Julie Bossuyt

Abstract:  Ca2+ in cardiac myocytes regulates contractility and relaxation, and Ca2+ and Na +regulation are linked via Na+/Ca2+ exchange (NCX). Heart failure (HF) is accompanied by contractile dysfunction and arrhythmias, both of which may be due to altered cellular Ca2+ handling. Smaller Ca2+ transient and sarcoplasmic reticulum (SR) Ca2+ content cause systolic dysfunction in HF. The reduced SR Ca2+ content is due to: (a) reduced SR Ca2+‐ATPase function (which also contributes to diastolic dysfunction), (b) increased expression and function of NCX (which competes with SR Ca2+‐ATPase during relaxation, but preserves diastolic function), and (c) enhanced diastolic SR Ca2+ leak. Relative contributions of these may vary with HF etiology and stage. Triggered arrhythmias (e.g., delayed afterdepolarizations [DADs]) are prominent in HF. DADs are due to spontaneous SR Ca2+ release and consequent activation of transient inward NCX current, which in HF allows DADs to more readily trigger arrhythmogenic action potentials. Thus NCX and Na+ are critical in systolic and diastolic function and arrhythmias. [Na+]i is elevated in HF, which may limit SR unloading and provide some Ca2+ influx during the HF action potential, thus limiting the depression of systolic function. High [Na+]i in HF is due to enhanced Na+ influx. Cellular Na+/K+‐ATPase (NKA) function appears unaltered, despite reduced NKA expression. This dichotomy led us to test NKA regulation by phospholemman (PLM). We find that PLM regulates NKA in a manner analogous to phospholamban regulation of SR Ca2+‐ATPase (i.e., inhibition that is relieved by PLM phosphorylation). We measured intermolecular FRET between PLM and NKA, which is reduced upon PLM phosphorylation. The lower expression level of more phosphorylated PLM in HF may explain the above dichotomy. Thus, altered Ca2+ and Na+ handling contributes to altered contractile function and arrhythmogenesis in HF.


The Journal of Physiology | 2002

Intracellular [Na^+] and Na^+ pump rate in rat and rabbit ventricular myocytes

Sanda Despa; Mohammed A. Islam; Steven M. Pogwizd; Donald M. Bers

Intracellular [Na+] ([Na+]i) is centrally involved in regulation of cardiac Ca2+ and contractility via Na+‐Ca2+ exchange (NCX) and Na+‐H+ exchange (NHX). Previous work has indicated that [Na+]i is higher in rat than rabbit ventricular myocytes. This has major functional consequences, but the reason for the higher [Na+]i in rat is unknown. Here, resting [Na+]i was measured using the fluorescent indicator SBFI, with both traditional calibration and a novel null‐point method (which circumvents many limitations of prior methods). In rabbit, resting [Na+]i was 4.5 ± 0.4 mm (traditional calibration) and 4.4 mm (null‐point). Resting [Na+]i in rat was significantly higher using both the traditional calibration (11.1 ± 0.7 mm) and the null‐point approach (11.2 mm). The rate of Na+ transport by the Na+ pump was measured as a function of [Na+]i in intact cells. Rat cells exhibited a higher Vmax than rabbit (7.7 ± 1.1 vs. 4.0 ± 0.5 mm min−1) and a higher Km (10.2 ± 1.2 vs. 7.5 ± 1.1 mm). This results in little difference in pump activity for a given [Na+]i below 10 mm, but at measured resting [Na+]i levels the pump‐mediated Na+ efflux is much higher in rat. Thus, Na+ pump rate cannot explain the higher [Na+]i in rat. Resting Na+ influx rate was two to four times higher in rat, and this accounts for the higher resting [Na+]i. Using tetrodotoxin, HOE‐642 and Ni2+ to block Na+ channels, NHX and NCX, respectively, we found that all three pathways may contribute to the higher resting Na+ influx in rat (albeit differentially). We conclude that resting [Na+]i is higher in rat than in rabbit, that this is caused by higher resting Na+ influx in rat and that a higher Na+,K+‐ATPase pumping rate in rat is a consequence of the higher [Na+]i.


Biophysical Journal | 2003

Na/Ca exchange and Na/K-ATPase function are equally concentrated in transverse tubules of rat ventricular myocytes.

Sanda Despa; F. Brette; C. H. Orchard; Donald M. Bers

Formamide-induced detubulation of rat ventricular myocytes was used to investigate the functional distribution of the Na/Ca exchanger (NCX) and Na/K-ATPase between the t-tubules and external sarcolemma. Detubulation resulted in a 32% decrease in cell capacitance, whereas cell volume was unchanged. Thus, the surface-to-volume ratio was used to assess the success of detubulation. NCX current (I(NCX)) and Na/K pump current (I(pump)) were recorded using whole-cell patch clamp, as Cd-sensitive and K-activated currents, respectively. Both inward and outward I(NCX) density was significantly reduced by approximately 40% in detubulated cells. I(NCX) density at 0 mV decreased from 0.19 +/- 0.03 to 0.10 +/- 0.03 pA/pF upon detubulation. I(pump) density was also lower in detubulated myocytes over the range of voltages (-50 to +100 mV) and internal [Na] ([Na](i)) investigated (7-22 mM). At [Na](i) = 10 mM and -20 mV, I(pump) density was reduced by 39% in detubulated myocytes (0.28 +/- 0.02 vs. 0.17 +/- 0.03 pA/pF), but the apparent K(m) for [Na](i) was unchanged (16.9 +/- 0.4 vs. 17.0 +/- 0.3 mM). These results indicate that although thet-tubules represent only approximately 32% of the total sarcolemma, they contribute approximately 60% to the total I(NCX) and I(pump). Thus, the functional density of NCX and Na/K pump in the t-tubules is 3-3.5-fold higher than in the external sarcolemma.


Circulation Research | 2006

Phospholemman Phosphorylation Mediates the Protein Kinase C–Dependent Effects on Na+/K+ Pump Function in Cardiac Myocytes

Fei Han; Julie Bossuyt; Sanda Despa; Amy L. Tucker; Donald M. Bers

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 (Ipump) (voltage clamp). In wild-type mouse myocytes, PDBu increased PLM phosphorylation at Ser63 and Ser68, Ipump (both at 10 and 100 mmol/L Na+ in the pipette solution) and maximal NKA-mediated Na+ extrusion rate (Vmax) from 7.9±1.1 to 12.7±1.9 mmol·L−1 per minute without altering NKA affinity for internal Na+ (K0.5). In PLM knockout mice, PDBu had no effect on either Vmax or K0.5. After pretreatment with isoproterenol (ISO) (1 &mgr;mol/L), PDBu still increased the NKA Vmax 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 Vmax and K0.5, respectively), and (3) PDBu and ISO combine to activate NKA in wild-type to the level found in the PLM knockout mouse.


Biophysical Journal | 2003

Na/K Pump Current and [Na]i in Rabbit Ventricular Myocytes: Local [Na]i Depletion and Na Buffering

Sanda Despa; Donald M. Bers

Na/K pump current (I(pump)) and intracellular Na concentration ([Na](i)) were measured simultaneously in voltage-clamped rabbit ventricular myocytes, under conditions where [Na](i) is controlled mainly by membrane transport. Upon abrupt pump reactivation (after 10-12 min blockade), I(pump) decays in two phases. Initially, I(pump) declines with little [Na](i) change, whereas the second phase is accompanied by [Na](i) decline. Initial I(pump) sag was still present at external [K] = 15 mM, but prevented by [Na](i) approximately 100 mM. Initial I(pump) sag might be explained by subsarcolemmal [Na](i) ([Na](SL)) depletion produced by rapid Na extrusion and I(pump). Brief episodes of pump blockade allowed [Na](SL) repletion, since peak postblockade I(pump) exceeded I(pump) at the end of previous activation (without appreciably altered global [Na](i)). The apparent K(m) for [Na](i) was higher for continuous I(pump) activation than peak I(pump) (14.1 +/- 0.2 vs. 11.2 +/- 0.2 mM), whereas that based on d[Na](i)/dt matched peak I(pump) (11.6 +/- 0.3 mM). [Na](SL) depletion (vs. [Na](i)) could be as high as 3 mM for [Na](i) approximately 18-20 mM. A simple diffusion model indicates that such [Na](SL) depletion requires a Na diffusion coefficient 10(3)- to 10(4)-fold below that expected in bulk cytoplasm (although this could be subsarcolemmal only). I(pump) integrals and [Na](i) decline were used to estimate intracellular Na buffering, which is slight (1.39 +/- 0.09).


Cardiovascular Research | 2012

Na+/K+-ATPase α2-isoform preferentially modulates Ca2+ transients and sarcoplasmic reticulum Ca2+ release in cardiac myocytes

Sanda Despa; Jerry B. Lingrel; Donald M. Bers

AIMS Na(+)/K(+)-ATPase (NKA) is essential in regulating [Na(+)](i), and thus cardiac myocyte Ca(2+) and contractility via Na(+)/Ca(2+) exchange. Different NKA-α subunit isoforms are present in the heart and may differ functionally, depending on specific membrane localization. In smooth muscle and astrocytes, NKA-α2 is located at the junctions with the endo(sarco)plasmic reticulum, where they could regulate local [Na(+)], and indirectly junctional cleft [Ca(2+)]. Whether this model holds for cardiac myocytes is unclear. METHODS AND RESULTS The ouabain-resistant NKA-α1 cannot be selectively blocked to assess its effect. To overcome this, we used mice in which NKA-α1 is ouabain sensitive and NKA-α2 is ouabain resistant (SWAP mice). We measured the effect of ouabain at low concentration on [Na(+)](i), Ca(2+) transients, and the fractional sarcoplasmic reticulum (SR) Ca(2+) release in cardiac myocytes from wild-type (WT; NKA-α2 inhibition) and SWAP mice (selective NKA-α1 block). At baseline, Na(+) and Ca(2+) regulations are similar in WT and SWAP mice. For equal levels of total NKA inhibition (~25%), ouabain significantly increased Ca(2+) transients (from ΔF/F(0)= 1.5 ± 0.1 to 1.8 ± 0.1), and fractional SR Ca(2+) release (from 24 ± 3 to 29 ± 3%) in WT (NKA-α2 block) but not in SWAP myocytes (NKA-α1 block). This occurred despite a similar and modest increase in [Na(+)](i) (~2 mM) in both groups. The effect in WT mice was mediated specifically by NKA-α2 inhibition because at a similar concentration ouabain had no effect in transgenic mice where both NKA-α1 and NKA-α2 are ouabain resistant. CONCLUSION NKA-α2 has a more prominent role (vs. NKA-α1) in modulating cardiac myocyte SR Ca(2+) release.


Journal of Biological Chemistry | 2006

Phospholemman Phosphorylation Alters Its Fluorescence Resonance Energy Transfer with the Na/K-ATPase Pump

Julie Bossuyt; Sanda Despa; Jody L. Martin; Donald M. Bers

Phospholemman (PLM) or FXYD1 is a major cardiac myocyte phosphorylation target upon adrenergic stimulation. Prior immunoprecipitation and functional studies suggest that phospholemman associates with the Na/K-pump (NKA) and mediates adrenergic Na/K-pump regulation. Here, we tested whether the NKA-PLM interaction is close enough to allow fluorescence resonance energy transfer (FRET) between cyan and yellow fluorescent (CFP/YFP) fusion proteins of Na/K pump and phospholemman and whether phospholemman phosphorylation alters such FRET. Co-expressed NKA-CFP and PLM-YFP in HEK293 cells co-localized in the plasma membrane and exhibited robust FRET. Selective acceptor photobleach increased donor fluorescence (FCFP) by 21.5 ± 4.1% (n = 13), an effect nearly abolished when co-expressing excess phospholemman lacking YFP. Activation of protein kinase C or A progressively and reversibly decreased FRET assessed by either the fluorescence ratio (FYFP/FCFP) or the enhancement of donor fluorescence after acceptor bleach. After protein kinase C activation, forskolin did not further reduce FRET, but after forskolin pretreatment, protein kinase C could still reduce FRET. This agreed with phospholemman phosphorylation measurements: by protein kinase C at both Ser-63 and Ser-68, but by protein kinase A only at Ser-68. Expression of PLM-YFP and PLM-CFP resulted in even stronger FRET than for NKA-PLM (FCFP increased by 37 ± 1% upon YFP photobleach), and this FRET was enhanced by phospholemman phosphorylation, consistent with phospholemman multimerization. Co-expressed PLM-CFP and Na/Ca exchange-YFP were highly membrane co-localized, but FRET was undetectable. We conclude that phospholemman and Na/K-pump are in very close proximity (FRET occurs) and that phospholemman phosphorylation alters the interaction of Na/K-pump and phospholemman.

Collaboration


Dive into the Sanda Despa's collaboration.

Top Co-Authors

Avatar

Donald M. Bers

University of California

View shared research outputs
Top Co-Authors

Avatar

Florin Despa

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie Bossuyt

University of California

View shared research outputs
Top Co-Authors

Avatar

Heinrich Taegtmeyer

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Peter J. Havel

University of California

View shared research outputs
Top Co-Authors

Avatar

Marcel Ameloot

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Le Chen

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge