phospholamban: a crucial regulator of cardiac

Adrenaline and other β-AGONISTS initiate .... been defined: Ca2+ and ATP bind independently, forming ..... an important determinant of β-adrenergic-agonist.
3MB taille 5 téléchargements 308 vues
REVIEWS

PHOSPHOLAMBAN: A CRUCIAL REGULATOR OF CARDIAC CONTRACTILITY David H. MacLennan* and Evangelia G. Kranias ‡ Heart failure is a major cause of death and disability. Impairments in blood circulation that accompany heart failure can be traced, in part, to alterations in the activity of the sarcoplasmic reticulum Ca2+ pump that are induced by its interactions with phospholamban, a reversible inhibitor. If phospholamban becomes superinhibitory or chronically inhibitory, contractility is diminished, inducing dilated cardiomyopathy in mice and humans. In mice, phospholamban seems to encumber an otherwise healthy heart, but humans with a phospholamban-null genotype develop early-onset dilated cardiomyopathy. C A LC I U M CARDIAC RESERVE

The maximum percentage that the cardiac output can increase above normal — the ability of the heart to adjust rapidly to demands placed on it. β-AGONIST

A molecule that activates β-adrenergic receptors.

*Banting and Best Department of Medical Research, University of Toronto, Charles H. Best Institute, 112 College Street, Toronto, Ontario M5G 1L6, Canada. ‡ Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA. Correspondence to D.H.M. e-mail: david.maclennan@ utoronto.ca doi:10.1038/nrm1151

566

Cardiac function is regulated on a beat-to-beat basis through the sympathetic nervous system. When the demand arises, the heart can respond to stress and increase blood flow to peripheral tissues within seconds. This is thanks to the large CARDIAC RESERVE in humans; the slow heart beat rate and submaximal contractility at rest are increased markedly after the release of adrenaline into the blood1. Adrenaline and other β-AGONISTS initiate an important signal-transduction pathway in the heart by binding to and activating β-ADRENERGIC RECEPTORS in the cell membrane (FIG. 1). The signal proceeds through Gs proteins to stimulate the formation of cyclic AMP by adenylate cyclase2. Elevations in cAMP concentration activate cAMP-dependent protein kinase (PKA), which then phosphorylates and alters the function of a few cardiac proteins that have key effects on the overall cardiac function. Prominent among these proteins is phospholamban (PLN), a small, reversibly phosphorylated, transmembrane protein that is located in the cardiac SARCOPLASMIC RETICULUM (SR), which, depending on its phosphorylation state, binds to and regulates the activity of a Ca2+ pump, the SARCO(ENDO)PLASMIC RETICULUM Ca -ATPASE SERCA2a. The trigger for cardiac contraction is the elevation of the Ca2+ concentration in the cytoplasm of the muscle cell (BOX 1), which is mediated by Ca2+-release channels 2+

| JULY 2003 | VOLUME 4

(RYANODINE RECEPTORS; RyRs) that tap the Ca2+ store in the lumen of the SR, or plasma-membrane Ca 2+ channels (DIHYDROPYRIDINE RECEPTORS; DHPRs) that tap the high concentrations of Ca2+ in the extracellular space. The trigger for relaxation is the lowering of the cytosolic Ca2+ concentration by the combined activity of SERCA2a, PLASMA-MEMBRANE Ca -ATPASES (PMCAs) and Na /Ca EXCHANGERS (NCXs) that replenish the SR and extracellular Ca2+ stores3. In humans, the activity of SERCA2a determines the rate of removal of >70% of cytosolic Ca 2+, thereby determining the rate of relaxation of the heart, and influences cardiac contractility by determining the size of the lumenal Ca2+ store that is available for release in the next beat. In its dephosphorylated state, PLN binds to SERCA2a at resting Ca2+ concentrations and inhibits Ca2+ pump activity: phosphorylation of PLN alters the PLN–SERCA2a interaction, relieving Ca2+-pump inhibition and enhancing relaxation rates (LUSITROPIC effects) and contractility (INOTROPIC effects)4. In this review, we discuss advances in the understanding of the molecular aspects of the interaction between PLN and SERCA2a that have been derived from extensive mutagenesis and structural modelling studies5. We describe gain- and loss-of-inhibitoryfunction PLN mutants, which led to the investigation 2+

+

2+

www.nature.com/reviews/molcellbio

© 2003 Nature Publishing Group

REVIEWS β-adrenergic receptor

β-ADRENERGIC STIMULATION

The ligand- or agonistdependent activation of β-adrenergic receptors and subsequent signalling events.

DHPR

NCX

PMCA Ca2+

1 Ca2+

Adenylate cyclase

SARCOPLASMIC RETICULUM

(SR). An organellar membrane system that encases each myofibril within a muscle cell. Its essential components are a Ca2+-ATPase (Ca2+ pump), lumenal Ca2+-sequestering proteins and a Ca2+-release channel.

Gsα 3 Na+

Ca2+ Ca2+

Myofibril

β-adrenergic receptor kinase

+ + + +

RyR Junctin/triadin

SARCO(ENDO)PLASMIC RETICULUM Ca2+-ATPASE

(SERCA). A pump that is located in sarcoplasmic or endoplasmic reticulum membranes that couples ATP hydrolysis to the transport of Ca2+ from cytosolic to lumenal spaces.

P

C-protein

P

Troponin I

cAMP

DIHYDROPYRIDINE RECEPTOR

(DHPR). A slow, or L-type, voltage-dependent Ca2+-entry channel that is located in the plasma membrane. DHPRs require a membrane potential that is greater than –30 mV for activation, and they are commonly found in neurons, neuroendocrine cells and muscle cells. PLASMA-MEMBRANE Ca2+-ATPASE

(PMCA). A plasma-membrane pump that couples ATP hydrolysis to the transport of Ca2+ from cytosolic to extracellular spaces.

Ca2+

Ca2+

ATP

SERCA2a

RYANODINE RECEPTOR

(RyR). A Ca2+-release channel that is located in the membrane of the sarcoplasmic and the endoplasmic reticulum that is regulated by protein–protein interactions with the dihydropyridine receptor and by a series of ligands, including Ca2+ itself.

Ca2+–calsequestrin

PKA

Ca2+

PLN P

PKA [Ca2+] Systolic 1 µM Diastolic 0.1 µM SR 500 µM Extracellular 2–3 mM

AKAP PP1

RgI SR

Mitochondria

Figure 1 | Interactions between cardiac signalling pathways. The heart provides an example of how two signalling pathways that are involved in elevating the levels of two intracellular second messengers, cyclic AMP (cAMP) and Ca2+, can interact physiologically (the two pathways are shown by the blue and red arrows, respectively). In response to depolarization, Ca2+ enters the cytoplasm through Ca2+ channels in the plasma membrane (dihydropyridine receptors; DHPRs). This ‘trigger’ Ca2+ then binds to the Ca2+-release channels (ryanodine receptors; RyRs) to stimulate Ca2+ release from the sarcoplasmic reticulum (SR). After activating muscle contraction by binding to troponin C in the thin filament, Ca2+ is removed from the myoplasm by plasma-membrane Ca2+-ATPases (PMCAs) or Na+/Ca2+ exchangers (NCXs), which are located in the plasma membrane, or by the sarco(endo)plasmic reticulum Ca2+-ATPase SERCA2a, which is located in the SR. As SERCA2a activity accounts for the removal of >70% of myoplasmic Ca2+ in humans, it determines both the rate of Ca2+ removal (and, consequently, the rate of cardiac muscle relaxation) and the size of the Ca2+ store (which affects cardiac contractility in the subsequent beat). SERCA2a activity is regulated by its interaction with phospholamban (PLN), which is a target for phosphorylation by protein kinase A (PKA) through the second signalling pathway — the β-adrenergic-receptor pathway (see main text). In its dephosphorylated form, PLN is an inhibitor of SERCA2a, but, when phosphorylated by PKA (or Ca2+/CaM kinase), PLN dissociates from SERCA2a, activating this Ca2+ pump. As a result, the rate of cardiac relaxation is increased and, on subsequent beats, contractility is increased in proportion to the elevation in the size of the SR Ca2+ store and the resulting increase in Ca2+ release from the SR. PLN is dephosphorylated by a protein phosphatase (PP1), which terminates the stimulation phase. AKAP, A-kinase anchoring protein; [Ca2+], Ca2+ concentration; Rgl, regulatory binding subunit A.

Na+/Ca2+ EXCHANGER

(NCX). A plasma-membrane enzyme that exchanges three moles of Na+ for one mole of Ca2+ either inward or outward, depending on ionic gradients across the membrane. LUSITROPIC

Affecting cardiac relaxation. INOTROPIC

Affecting the force of cardiac contractions. CARDIOMYOPATHY

A disease of the heart muscle. VMAX

The maximal rate of enzymatic

of mutations in the PLN gene as a potential cause of 6 CARDIOMYOPATHY . We also discuss studies that were carried out in transgenic mice that overexpress superinhibitory forms of PLN and that have confirmed this potential7–9. The creation and analysis of PLN-null mice10 and of mice that overexpress PLN 11 have enabled the elucidation of the remarkable role of PLN in the regulation of the kinetics of cardiac contractility12. The fact that ablation of PLN or suppression of the inhibitory function of PLN can intervene to prevent the progression of dilated cardiomyopathy in wellcharacterized animal models opens the door to the investigation of the diverse pathways that lead to endstage heart failure and to potential therapeutic interventions13–15. Finally, we discuss very recent data,

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY

which show not only that mutations in PLN can cause dilated cardiomyopathy in humans, but also introduce new principles16,17. We learn that humans, unlike mice, require PLN and that dilated cardiomyopathy can result from chronic inhibition of SERCA2a by the prevention of phosphorylation of PLN by PKA. Phospholamban–SERCA interactions

One of the most useful assays for the interaction between PLN and SERCA molecules is the measurement of the Ca2+ dependence of Ca2+ transport in isolated SR vesicles18. By varying the Ca2+ concentration over four orders of magnitude and calculating the rate of ATP-dependent Ca2+ transport relative to the maximal rate observed at 10 µM Ca2+ (V ), the apparent MAX

VOLUME 4 | JULY 2003 | 5 6 7

© 2003 Nature Publishing Group

REVIEWS

Box 1 | Regulation and dysregulation of the Ca2+ transient During the evolution of phosphate- and organic-acid-based metabolic processes, it became important that cytosolic Ca2+ concentrations be maintained below ~10 µM; and so the need for Ca2+ pumps that lower resting Ca2+ concentrations to 4 at Θ=0.00)16. The effects of the PLN Arg9Cys mutation were characterized by expression in heterologous cell culture, by the creation of a transgenic mouse and by analysis of cardiac tissue obtained from an explant. In all cases, the level of PLN phosphorylation was reduced markedly. In heterologous cell culture, the Arg9Cys mutant PLN had reduced inhibitory properties in the homozygous state, but, when expressed in the heterozygous state, did not function in a dominant fashion to prevent the inhibition of SERCA2a by wild-type PLN. The key effect of the mutation was enhancement of the affinity of Arg9Cys mutant PLN for PKA. In attempting to phosphorylate mutant PLN, PKA becomes trapped in a stabilized mutant PLN–PKA complex and can no longer dissociate to phosphorylate wild-type PLN molecules. The effect seems to be local and restricted to the SR, perhaps because a specific fraction of PKA is associated with

www.nature.com/reviews/molcellbio

© 2003 Nature Publishing Group

REVIEWS

Contraction

DHPR

1 Ca2+

RyR

NCX

SR Myofibrils

3 Na+ 1 Ca2+

DHPR

SERCA

NCX

PLN 3 Na+

Mouse Human

Relaxation

Figure 5 | Differences in the regulation of Ca2+ transients between mice and humans. With heart beat rates of up to 800 beats per min, mice are normally at two thirds of the theoretical maximum and the sarcoplasmic reticulum (SR) Ca2+ store is large at normal heart rates. Mice cycle 90% of their cardiac Ca2+ through the SR and only 10% through extracellular spaces in a very short temporal cycle. Humans, with heart beat rates ranging between 60 and 180 beats per min, have a larger cardiac reserve. A major part of this reserve is the potential for a gain in SR Ca2+ content. Normally humans cycle only 70% of their Ca2+ through the SR in a longer temporal cycle. The red arrows show the cycle in humans, including the involvement of the dihydropyridine receptor (DHPR) and Na+/Ca2+ exchanger (NCX), and the blue arrows show the cycle in mice, again including the DHPR and NCX. The red dots represent Ca2+ ions. For a general explanation of cardiac Ca2+ signalling and, specifically, the roles of ryanodine receptors (RyRs), phospholamban (PLN) and sarco(endo)plasmic Ca2+-ATPase (SERCA), see FIG. 1.

the SR through A-kinase anchoring proteins (AKAPs)98. These results explain the dominant effects of the mutation. Affected individuals must go through life with chronically inhibited SERCA2a and can never draw on their full cardiac reserve. The long-term effects of the chronic, specific inhibition of PLN phosphorylation on Ca2+ transients are clearly sufficiently deleterious to cause the onset of dilated cardiomyopathy in humans in their teenage years. However, analysis of the transgenic expression of a PLN Ser16Ala, Thr17Ala double mutant in mice provided different results. This mutant PLN could not be phosphorylated, but produced the same shift in Ca2+ affinity as unphosphorylated, wild-type PLN. The measures of basal contractility and Ca 2+ transients were similar in wild-type and mutant myocytes, but isoproterenol did not increase the rate of Ca2+ removal in the mutant. Increased L-type Ca 2+ current (ICa) density, with unaltered characteristics, was the principal compensation in mutant myocytes. This ICa modulation might compensate in part for the loss in SERCA2a responsiveness and thereby partially normalize β-adrenergic inotropy in Ser16Ala, Thr17Ala double-mutant mice, allowing them to survive without cardiomyopathy73.

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY

A second human PLN mutation, Leu39stop, was discovered in two large Greek families17. The heterozygous inheritance of the Leu39stop mutation in one family led to left ventricular hypertrophy in one third of the older affected family members, without diminished contractile performance. However, the inheritance of two copies of the mutant PLN gene led to dilated cardiomyopathy and heart failure in two teenage siblings. In heterologous expression studies, the Leu39stop mutant protein was unstable or misrouted to other membranes and no protein was detected in the endoplasmic reticulum of these cells or in a cardiac explant from one of the affected individuals. As a result, there was no effect of the mutant protein, in either the homozygous or heterozygous state, on the Ca2+ affinity of SERCA2a. Accordingly, these two homozygous mutant individuals can be considered to be equivalent to a PLN-null genotype with a phenotype of dilated cardiomyopathy. So, in contrast to the benefits of PLN ablation in mouse, humans that lack PLN develop lethal cardiomyopathy. A caveat in these studies is that the number of affected individuals is very low and the lod score for linkage of the mutation to the disease is low. So, how can this discrepancy between the cardiac phenotypes in mice and humans be explained? In contrast to humans, mice have relatively little cardiac reserve and differ in the balance of myocyte Ca2+ fluxes (FIG. 5). With heart beat rates ranging up to 800 beats per min, mice are normally at two thirds of the theoretical maximum and the SR Ca2+ store is nearly full at normal heart rates. Humans, with heart beat rates ranging between 60 and 180 beats per min, have a large cardiac reserve1. A major part of this reserve is the potential for a gain in SR Ca2+ content99. If defective PLN regulation limits the normal gain in SR Ca2+ content with increasing heart rate, cardiac reserve will be compromised, as observed in human heart failure99. So, PLN modulation might be of more paramount importance in humans, who have a life span that is 40 times that of a mouse. Conclusion

PLN is an important regulator of the kinetics of cardiac Ca2+ transients and of contractility, and is a key determinant of β-adrenergic stimulation of the heart. Significant advances have been made in understanding the structure and dynamics of the interaction of PLN with SERCA2a. And the generation of mouse models with altered PLN expression levels or activity has allowed a thorough understanding of the ‘physiological brake’ that is provided by PLN in vivo. Although the mouse and human myocardia differ in excitation–contraction coupling and in the isoforms of contractile proteins that they express in different developmental stages, studies of mice form the basis for comparative studies in humans. They also form the basis for potential therapeutic approaches in humans through disruption of PLN inhibition of SERCA2a. Depressed SR Ca 2+ cycling is a common feature of the depressed contractility that is seen in hypertrophied and failing myocardia, so that interference

VOLUME 4 | JULY 2003 | 5 7 5

© 2003 Nature Publishing Group

REVIEWS with the PLN–SERCA2a interaction through a ‘molecular inotrope’ might rescue contractile dysfunction and prevent transcription-based remodelling of the heart, thereby preventing progression to heart failure. It is evident, however, that elimination of PLN activity might not benefit all forms of heart failure, especially those in which depressed SR Ca2+ cycling is not a primary event.

1.

2. 3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

576

Kass, D. A., Hare, J. M. & Georgakopoulos, D. Murine cardiac function: a cautionary tail. Circ. Res. 82, 519–522 (1998). Levitzki, A. From epinephrine to cyclic AMP. Science 241, 800–806 (1988). Bers, D. M. Calcium and cardiac rhythms: physiological and pathophysiological. Circ. Res. 90, 14–17 (2002). Simmerman, H. K. & Jones, L. R. Phospholamban: protein structure, mechanism of action, and role in cardiac function. Physiol. Rev. 78, 921–947 (1998). Toyoshima, C. et al. Modeling of the inhibitory interaction of phospholamban with the Ca2+ ATPase. Proc. Natl Acad. Sci USA 100, 467–472 (2003). Structural modelling shows that grooves in the transmembrane and cytosolic domains of SERCA1a provide high-affinity binding sites for PLN in the E2, but not the E1.2Ca2+ conformation. Kimura, Y., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Phospholamban inhibitory function is activated by depolymerization. J. Biol. Chem. 272, 15061–15064 (1997). Scanning mutagenesis of the PLN transmembrane helix shows two faces: one interacts with SERCA, so that mutants remain pentameric but lose inhibitory function; the other interacts with PLN, so that mutants become monomeric and gain inhibitory function. It is deduced that the PLN monomer is the active inhibitory species. Zhai, J. et al. Cardiac-specific overexpression of a superinhibitory pentameric phospholamban mutant enhances inhibition of cardiac function in vivo. J. Biol. Chem. 275, 10538–10544 (2000). Superinhibition of SERCA2a by a PLN mutant results in dilated cardiomyopathy in a mouse model. Zvaritch, E. et al. The transgenic expression of highly inhibitory monomeric forms of phospholamban in mouse heart impairs cardiac contractility. J. Biol. Chem. 275, 14985–14991 (2000). Haghighi, K. et al. Superinhibition of sarcoplasmic reticulum function by phospholamban induces cardiac contractile failure. J. Biol. Chem. 276, 24145–24152 (2001). Luo, W. et al. Targeted ablation of the phospholamban gene is associated with markedly enhanced myocardial contractility and loss of β-agonist stimulation. Circ. Res. 75, 401–409 (1994). The first clear evidence of the important role of PLN in the regulation of cardiac contractility in a whole animal. Kadambi, V. J. et al. Cardiac-specific overexpression of phospholamban alters calcium kinetics and resultant cardiomyocyte mechanics in transgenic mice. J. Clin. Invest. 97, 533–539 (1996). Only about 40% of SERCA2a molecules are inhibited by PLN under normal conditions, even though PLN is in molar excess over SERCA2a. Koss, K. L. & Kranias, E. G. Phospholamban: a prominent regulator of myocardial contractility. Circ. Res. 79, 1059–1063 (1996). Minamisawa, S. et al. Chronic phospholamban–sarcoplasmic reticulum calcium ATPase interaction is the critical calcium cycling defect in dilated cardiomyopathy. Cell 99, 313–322 (1999). PLN ablation can reverse heart failure in a cardiomyopathic mouse model that is induced by a mutation in a structural protein. He, H. et al. Effects of mutant and antisense RNA of phospholamban on SR Ca2+-ATPase activity and cardiac myocyte contractility. Circulation 100, 974–980 (1999). Hoshijima, M. et al. Chronic suppression of heart-failure progression by a pseudophosphorylated mutant of phospholamban via in vivo cardiac rAAV gene delivery. Nature Med. 8, 864–871 (2002). Schmitt, J. P. et al. Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Science 299, 1410–1413 (2003).

17.

18.

19. 20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

Further structural studies of the PLN–SERCA2a complex might provide further clues as to the mechanisms that underlie PLN regulation and provide new insights into ways to design drugs that would disrupt this interaction selectively. The characterization of other deleterious PLN mutants in the human population will further elucidate the functional role of PLN in cardiac physiology and its effects on predisposition to heart disease.

A human PLN mutation that is associated with heart failure in man inhibits PKA, leading to a lack of PLN phosphorylation and chronic inhibition of SERCA2a. Haghighi, K. et al. Human phospholamban null results in lethal dilated cardiomyopathy: critical difference between mouse and man. J. Clin. Invest. 111, 869–876 (2003). In contrast to the beneficial effects of PLN ablation in mice, the absence of PLN in man might be associated with heart failure. Kirchberger, M. A., Tada, M., Repke, D. I. & Katz, A. M. Cyclic adenosine 3′,5′-monophosphate-dependent protein kinase stimulation of calcium uptake by canine cardiac microsomes. J. Mol. Cell. Cardiol. 4, 673–680 (1972). Katz, A. M. Discovery of phospholamban. A personal history. Ann. NY Acad. Sci. 853, 9–19 (1998). Tada, M., Kirchberger, M. A. & Katz, A. M. Phosphorylation of a 22,000-dalton component of the cardiac sarcoplasmic reticulum by adenosine 3′:5′-monophosphate-dependent protein kinase. J. Biol. Chem. 250, 2640–2647 (1975). Identification of PLN as a crucial player in β-adrenergic stimulation of the heart. La Raia, P. J. & Morkin, E. Adenosine 3′,5′-monophosphate dependent membrane phosphorylation: a possible mechanism for the control of microsomal calcium transport in heart muscle. Circ. Res. 35, 298–306 (1974). Cantilina, T., Sagara, Y., Inesi, G. & Jones, L. R. Comparative studies of cardiac and skeletal sarcoplasmic reticulum ATPases. Effect of a phospholamban antibody on enzyme activation by Ca2+. J. Biol. Chem. 268, 17018–17025 (1993). Hughes, G., Starling, A. P., Sharma, R. P., East, J. M. & Lee, A. G. An investigation of the mechanism of inhibition of the Ca2+-ATPase by phospholamban. Biochem. J. 318, 973–979 (1996). Kranias, E. G. & Solaro, R. J. Phosphorylation of troponin I and phospholamban during catecholamine stimulation of rabbit heart. Nature 298, 182–184 (1982). PLN is phosphorylated when elevated levels of catecholamines stimulate contractility of intact hearts. Wegener, A. D., Simmerman, H. K., Lindemann, J. P. & Jones, L. R. Phospholamban phosphorylation in intact ventricles. Phosphorylation of serine 16 and threonine 17 in response to β-adrenergic stimulation. J. Biol. Chem. 264, 11468–11474 (1989). Talosi, L., Edes, I. & Kranias, E. G. Intracellular mechanisms mediating reversal of β-adrenergic stimulation in intact beating hearts. Am. J. Physiol. 264, H791–H797 (1993). Lindemann, J. P., Jones, L. R., Hathaway, D. R., Henry, B. G. & Watanabe, A. M. β-adrenergic stimulation of phospholamban phosphorylation and Ca2+-ATPase activity in guinea pig ventricles. J. Biol. Chem. 258, 464–471 (1983). Garvey, J. L., Kranias, E. G. & Solaro, R. J. Phosphorylation of C-protein, troponin I and phospholamban in isolated rabbit hearts. Biochem. J. 249, 709–714 (1988). Mundina-Weilenmann, C., Vittone, L., Ortale, M., de Cingolani, G. C. & Mattiazzi, A. Immunodetection of phosphorylation sites gives new insights into the mechanisms underlying phospholamban phosphorylation in the intact heart. J. Biol. Chem. 271, 33561–33567 (1996). Fujii, J. et al. Complete complementary DNA-derived amino acid sequence of canine cardiac phospholamban. J. Clin. Invest. 79, 301–304 (1987). Odermatt, A. et al. Characterization of the gene encoding human sarcolipin (SLN), a proteolipid associated with SERCA1: absence of structural mutations in five patients with Brody disease. Genomics 45, 541–553 (1997). Odermatt, A. et al. Sarcolipin regulates the activity of SERCA1, the fast-twitch skeletal muscle sarcoplasmic reticulum Ca2+-ATPase. J. Biol. Chem. 273, 12360–12369 (1998). Mortishire-Smith, R. J. et al. Solution structure of the cytoplasmic domain of phopholamban: phosphorylation leads to a local perturbation in secondary structure. Biochemistry 34, 7603–7613 (1995).

| JULY 2003 | VOLUME 4

34. Pollesello, P., Annila, A. & Ovaska, M. Structure of the 1–36 amino-terminal fragment of human phospholamban by nuclear magnetic resonance and modeling of the phospholamban pentamer. Biophys. J. 76, 1784–1795 (1999). 35. Lamberth, S. et al. NMR structure of phospholamban. Helvetica Chimica Acta 83, 2141–2152 (2000). 36. Mascioni, A., Karim, C., Barany, G., Thomas, D. D. & Veglia, G. Structure and orientation of sarcolipin in lipid environments. Biochemistry 41, 475–482 (2002). 37. James, P., Inui, M., Tada, M., Chiesi, M. & Carafoli, E. Nature and site of phospholamban regulation of the Ca2+ pump of sarcoplasmic reticulum. Nature 342, 90–92 (1989). 38. Toyoshima, C. & Nomura, H. Structural changes in the calcium pump accompanying the dissociation of calcium. Nature 418, 605–611 (2002). 39. Toyofuku, T., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Identification of regions in the Ca2+-ATPase of sarcoplasmic reticulum that affect functional association with phospholamban. J. Biol. Chem. 268, 2809–2815 (1993). 40. Asahi, M., Kimura, Y., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Transmembrane helix m6 in sarco(endo)plasmic reticulum Ca2+-ATPase forms a functional interaction site with phospholamban. Evidence for physical interactions at other sites. J. Biol. Chem. 274, 32855–32862 (1999). 41. Toyofuku, T., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Amino acids Lys-Asp-Asp-Lys-Pro-Val402 in the Ca2+-ATPase of cardiac sarcoplasmic reticulum are critical for functional association with phospholamban. J. Biol. Chem. 269, 22929–22932 (1994). 42. Toyofuku, T., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Amino acids Glu2 to Ile18 in the cytoplasmic domain of phospholamban are essential for functional association with the Ca2+-ATPase of sarcoplasmic reticulum. J. Biol. Chem. 269, 3088–3094 (1994). 43. Kimura, Y., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Phospholamban regulates the Ca2+-ATPase through intramembrane interactions. J. Biol. Chem. 271, 21726–21731 (1996). 44. Arkin, I. T. et al. Structural organization of the pentameric transmembrane α-helices of phospholamban, a cardiac ion channel. EMBO J. 13, 4757–4764 (1994). 45. Simmerman, H. K., Kobayashi, Y. M., Autry, J. M. & Jones, L. R. A leucine zipper stabilizes the pentameric membrane domain of phospholamban and forms a coiledcoil pore structure. J. Biol. Chem. 271, 5941–5946 (1996). 46. Adams, P. D., Arkin, I. T., Engelman, D. M. & Brunger, A. T. Computational searching and mutagenesis suggest a structure for the pentameric transmembrane domain of phospholamban. Nature Struct. Biol. 2, 154–162 (1995). 47. Autry, J. M. & Jones, L. R. Functional co-expression of the canine cardiac Ca2+ pump and phospholamban in Spodoptera frugiperda (Sf21) cells reveals new insights on ATPase regulation. J. Biol. Chem. 272, 15872–15880 (1997). 48. Kimura, Y., Asahi, M., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Phospholamban domain Ib mutations influence functional interactions with the Ca2+-ATPase isoform of cardiac sarcoplasmic reticulum. J. Biol. Chem. 273, 14238–14241 (1998). 49. MacLennan, D. H., Rice, W. J. & Green, N. M. The mechanism of Ca2+ transport by sarco(endo)plasmic reticulum Ca2+-ATPases. J. Biol. Chem. 272, 28815–28818 (1997). 50. Toyoshima, C., Nakasako, M., Nomura, H. & Ogawa, H. Crystal structure of the calcium pump of sarcoplasmic reticulum at 2.6 Å resolution. Nature 405, 647–655 (2000). 51. Knighton, D. R. et al. Structure of a peptide inhibitor bound to the catalytic subunit of cyclic adenosine monophosphatedependent protein kinase. Science 253, 414–420 (1991). 52. Hutter, M. C. et al. A structural model of the complex formed by phospholamban and the calcium pump of sarcoplasmic reticulum obtained by molecular mechanics. Chembiochem. 3, 1200–1208 (2002).

www.nature.com/reviews/molcellbio

© 2003 Nature Publishing Group

REVIEWS 53. Asahi, M., McKenna, E., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Physical interactions between phospholamban and sarco(endo)plasmic reticulum Ca2+-ATPases are dissociated by elevated Ca2+, but not by phospholamban phosphorylation, vanadate, or thapsigargin, and are enhanced by ATP. J. Biol. Chem. 275, 15034–15038 (2000). 54. Luo, W. et al. Phospholamban gene dosage effects in the mammalian heart. Circ. Res. 78, 839–847 (1996). 55. Wolska, B. M., Stojanovic, M. O., Luo, W., Kranias, E. G. & Solaro, R. J. Effect of ablation of phospholamban on dynamics of cardiac myocyte contraction and intracellular Ca2+. Am. J. Physiol. 271, C391–C397 (1996). 56. Li, L., Chu, G., Kranias, E. G. & Bers, D. M. Cardiac myocyte calcium transport in phospholamban knockout mouse: relaxation and endogenous CaMKII effects. Am. J. Physiol. 274, H1335–H1347 (1998). 57. Lorenz, J. N. & Kranias, E. G. Regulatory effects of phospholamban on cardiac function in intact mice. Am. J. Physiol. 273, H2826–H2831 (1997). 58. Kiss, E. et al. β-adrenergic regulation of cAMP and protein phosphorylation in phospholamban-knockout mouse hearts. Am. J. Physiol. 272, H785–H790 (1997). 59. Santana, L. F., Gomez, A. M., Kranias, E. G. & Lederer, W. J. Amount of calcium in the sarcoplasmic reticulum: influence on excitation–contraction coupling in heart muscle. Heart Vessels Suppl. 12, 44–49 (1997). 60. Masaki, H., Sato, Y., Luo, W., Kranias, E. G. & Yatani, A. Phospholamban deficiency alters inactivation kinetics of L-type Ca2+ channels in mouse ventricular myocytes. Am. J. Physiol. 272, H606–H612 (1997). 61. Chu, G. et al. Compensatory mechanisms associated with the hyperdynamic function of phospholamban-deficient mouse hearts. Circ. Res. 79, 1064–1076. (1996). 62. Desai, K. H., Schauble, E., Luo, W., Kranias, E. & Bernstein, D. Phospholamban deficiency does not compromise exercise capacity. Am. J. Physiol. 276, H1172–H1177 (1999). 63. Slack, J. P. et al. The enhanced contractility of the phospholamban-deficient mouse heart persists with aging. J. Mol. Cell. Cardiol. 33, 1031–1040 (2001). 64. Kiriazis, H. et al. Hypertrophy and functional alterations in hyperdynamic phospholamban-knockout mouse hearts under chronic aortic stenosis. Cardiovasc. Res. 53, 372–381 (2002). 65. Cross, H. R., Kranias, E. G., Murphy, E. & Steenbergen, C. Ablation of PLB exacerbates ischemic injury to a lesser extent in female than male mice: protective role of NO. Am. J. Physiol. Heart Circ. Physiol. 284, H683–H690 (2003). 66. Lalli, J., Harrer, J. M., Luo, W., Kranias, E. G. & Paul, R. J. Targeted ablation of the phospholamban gene is associated with a marked decrease in sensitivity in aortic smooth muscle. Circ. Res. 80, 506–513 (1997). 67. Slack, J. P., Grupp, I. L., Luo, W. & Kranias, E. G. Phospholamban ablation enhances relaxation in the murine soleus. Am. J. Physiol. 273, C1–C6 (1997). 68. Nobe, K., Sutliff, R. L., Kranias, E. G. & Paul, R. J. Phospholamban regulation of bladder contractility: evidence from gene-altered mouse models. J. Physiol. 535, 867–878 (2001). 69. Sutliff, R. L., Hoying, J. B., Kadambi, V. J., Kranias, E. G. & Paul, R. J. Phospholamban is present in endothelial cells and modulates endothelium-dependent relaxation. Evidence from phospholamban gene-ablated mice. Circ. Res. 84, 360–364 (1999). 70. Dash, R. et al. Interactions between phospholamban and β-adrenergic drive may lead to cardiomyopathy and early mortality. Circulation 103, 889–896 (2001). 71. Dash, R., Frank, K. F., Carr, A. N., Moravec, C. S. & Kranias, E. G. Gender influences on sarcoplasmic reticulum Ca2+-handling in failing human myocardium. J. Mol. Cell. Cardiol. 33, 1345–1353 (2001). 72. Colyer, J. & Wang, J. H. Dependence of cardiac sarcoplasmic reticulum calcium pump activity on the phosphorylation status of phospholamban. J. Biol. Chem. 266, 17486–17493 (1991). 73. Brittsan, A. G., Carr, A. N., Schmidt, A. G. & Kranias, E. G. Maximal inhibition of SERCA2 Ca2+ affinity by phospholamban in transgenic hearts overexpressing a non-phosphorylatable form of phospholamban. J. Biol. Chem. 275, 12129–12135 (2000). 74. Schmidt, A. G. et al. Structural and functional implications of the phospholamban hinge domain: impaired SR Ca2+ uptake as a primary cause of heart failure. Cardiovasc. Res. 56, 248–259 (2002). 75. Luo, W. et al. Transgenic approaches to define the functional role of dual site phospholamban phosphorylation. J. Biol. Chem. 273, 4734–4739 (1998).

76. Chu, G. et al. A single site (Ser16) phosphorylation in phospholamban is sufficient in mediating its maximal cardiac responses to β-agonists. J. Biol. Chem. 275, 38938–38943 (2000). 77. Vittone, L., Mundina-Weilenmann, C., Said, M., Ferrero, P. & Mattiazzi, A. Time course and mechanisms of phosphorylation of phospholamban residues in ischemiareperfused rat hearts. Dissociation of phospholamban phosphorylation pathways. J. Mol. Cell. Cardiol. 34, 39–50 (2002). 78. Hagemann, D. et al. Frequency-encoding Thr17 phospholamban phosphorylation is independent of Ser16 phosphorylation in cardiac myocytes. J. Biol. Chem. 275, 22532–22536 (2000). 79. Beuckelmann, D. J., Nabauer, M. & Erdmann, E. Intracellular calcium handling in isolated ventricular myocytes from patients with terminal heart failure. Circulation 85, 1046–1055 (1992). 80. Dipla, K., Mattiello, J. A., Margulies, K. B., Jeevanandam, V. & Houser, S. R. The sarcoplasmic reticulum and the Na+/Ca2+ exchanger both contribute to the Ca2+ transient of failing human ventricular myocytes. Circ. Res. 84, 435–444 (1999). 81. Hasenfuss, G. et al. Relation between myocardial function and expression of sarcoplasmic reticulum Ca2+-ATPase in failing and nonfailing human myocardium. Circ. Res. 75, 434–442 (1994). 82. Schwinger, R. H. et al. Unchanged protein levels of SERCA II and phospholamban but reduced Ca2+ uptake and Ca2+-ATPase activity of cardiac sarcoplasmic reticulum from dilated cardiomyopathy patients compared with patients with nonfailing hearts. Circulation 92, 3220–3228 (1995). 83. Frank, K., Bolck, B., Bavendiek, U. & Schwinger, R. H. Frequency dependent force generation correlates with sarcoplasmic calcium ATPase activity in human myocardium. Basic Res. Cardiol. 93, 405–411 (1998). 84. Movsesian, M. A., Karimi, M., Green, K. & Jones, L. R. Ca2+-transporting ATPase, phospholamban, and calsequestrin levels in nonfailing and failing human myocardium. Circulation 90, 653–657 (1994). 85. Meyer, M. et al. Alterations of sarcoplasmic reticulum proteins in failing human dilated cardiomyopathy. Circulation 92, 778–784 (1995). 86. Schwinger, R. H. et al. Reduced Ca2+-sensitivity of SERCA2a in failing human myocardium due to reduced serine-16 phospholamban phosphorylation. J. Mol. Cell. Cardiol. 31, 479–491 (1999). 87. Giordano, F. J. et al. Adenovirus-mediated gene transfer reconstitutes depressed sarcoplasmic reticulum Ca2+-ATPase levels and shortens prolonged cardiac myocyte Ca2+ transients. Circulation 96, 400–403 (1997). 88. del Monte, F. et al. Restoration of contractile function in isolated cardiomyocytes from failing human hearts by gene transfer of SERCA2a. Circulation 100, 2308–2311 (1999). 89. Miyamoto, M. I. et al. Adenoviral gene transfer of SERCA2a improves left-ventricular function in aortic-banded rats in transition to heart failure. Proc. Natl Acad. Sci. USA 97, 793–798 (2000). 90. del Monte, F. et al. Improvement in survival and cardiac metabolism after gene transfer of sarcoplasmic reticulum Ca2+-ATPase in a rat model of heart failure. Circulation 104, 1424–1429 (2001). 91. He, H. et al. Overexpression of the rat sarcoplasmic reticulum Ca2+ ATPase gene in the heart of transgenic mice accelerates calcium transients and cardiac relaxation. J. Clin. Invest. 100, 380–389 (1997). 92. Baker, D. L. et al. Targeted overexpression of the sarcoplasmic reticulum Ca2+-ATPase increases cardiac contractility in transgenic mouse hearts. Circ. Res. 83, 1205–1214 (1998). 93. Eizema, K. et al. Adenovirus-based phospholamban antisense expression as a novel approach to improve cardiac contractile dysfunction: comparison of a constitutive viral versus an endothelin-1-responsive cardiac promoter. Circulation 101, 2193–2199 (2000). 94. del Monte, F., Harding, S. E., Dec, G. W., Gwathmey, J. K. & Hajjar, R. J. Targeting phospholamban by gene transfer in human heart failure. Circulation 105, 904–907 (2002). 95. Sato, Y. et al. Rescue of contractile parameters and myocyte hypertrophy in calsequestrin overexpressing myocardium by phospholamban ablation. J. Biol. Chem. 276, 9392–9399 (2001). 96. Freeman, K. et al. Alterations in cardiac adrenergic signaling and calcium cycling differentially affect the progression of cardiomyopathy. J. Clin. Invest. 107, 967–974 (2001). 97. Song, Q. et al. Rescue of cardiomyocyte dysfunction by phospholamban ablation does not prevent ventricular failure in genetic hypertrophy. J. Clin. Invest. 111, 859–867 (2003).

NATURE REVIEWS | MOLECUL AR CELL BIOLOGY

98. Fink, M. A. et al. AKAP-mediated targeting of protein kinase a regulates contractility in cardiac myocytes. Circ. Res. 88, 291–297 (2001). 99. Pieske, B. et al. Functional effects of endothelin and regulation of endothelin receptors in isolated human nonfailing and failing myocardium. Circulation 99, 1802–1809 (1999). 100. MacLennan, D. H. & Reithmeier, R. A. Ion tamers. Nature Struct. Biol. 5, 409–411 (1998). 101. Zot, A. S. & Potter, J. D. Structural aspects of troponin–tropomyosin regulation of skeletal muscle contraction. Annu. Rev. Biophys. Biophys. Chem. 16, 535–559 (1987). 102. MacLennan, D. H. Ca2+ signalling and muscle disease. Eur. J. Biochem. 267, 5291–5297 (2000). 103. Loke, J. & MacLennan, D. H. Malignant hyperthermia and central core disease: disorders of Ca2+ release channels. Am. J. Med. 104, 470–486 (1998). 104. Priori, S. G. et al. Mutations in the cardiac ryanodine receptor gene (hRyR2) underlie catecholaminergic polymorphic ventricular tachycardia. Circulation 103, 196–200 (2001). 105. Laitinen, P. J. et al. Mutations of the cardiac ryanodine receptor (RyR2) gene in familial polymorphic ventricular tachycardia. Circulation 103, 485–490 (2001). 106. Postma, A. V. et al. Absence of calsequestrin 2 causes severe forms of catecholaminergic polymorphic ventricular tachycardia. Circ. Res. 91, e21–e26 (2002). 107. Sakuntabhai, A. et al. Mutations in ATP2A2, encoding a Ca2+ pump, cause Darier disease. Nature Genet. 21, 271–277 (1999). 108. Wawrzynow, A. et al. Sarcolipin, the ‘proteolipid’ of skeletal muscle sarcoplasmic reticulum, is a unique, amphipathic, 31-residue peptide. Arch. Biochem. Biophys. 298, 620–623 (1992). 109. Gayan-Ramirez, G., Vanzeir, L., Wuytack, F. & Decramer, M. Corticosteroids decrease mRNA levels of SERCA pumps, whereas they increase sarcolipin mRNA in the rat diaphragm. J. Physiol. 524, 387–397 (2000). 110. Minamisawa, S. et al. Atrial-specific expression of sarcolipin is regulated during development and hypertrophic remodeling. J. Biol. Chem. 278, 9570–9575 (2003). 111. Tupling, A. R., Asahi, M. & MacLennan, D. H. Sarcolipin overexpression in rat slow twitch muscle inhibits sarcoplasmic reticulum Ca2+ uptake and impairs contractile function. J. Biol. Chem. 277, 44740–44746 (2002). 112. Asahi, M., Kurzydlowski, K., Tada, M. & MacLennan, D. H. Sarcolipin inhibits polymerization of phospholamban to induce superinhibition of sarco(endo)plasmic reticulum Ca2+-ATPases (SERCAs). J. Biol. Chem. 277, 26725–26728 (2002). 113. Asahi, M. et al. Sarcolipin regulates sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA1a) by binding to transmembrane helices alone or in association with phospholamban. Proc. Natl Acad. Sci. USA 100, 5040–5045 (2003). 114. de Meis, L. & Vianna, A. L. Energy interconversion by the Ca2+-dependent ATPase of the sarcoplasmic reticulum. Annu. Rev. Biochem. 48, 275–293 (1979). 115. Stokes, D. L. & Green, N. M. Structure and function of the calcium pump. Annu. Rev. Biophys. Biomol. Struct. 32, 445–468 (2003). 116. Cornea, R. L., Jones, L. R., Autry, J. M. & Thomas, D. D. Mutation and phosphorylation change the oligomeric structure of phospholamban in lipid bilayers. Biochemistry 36, 2960–2967 (1997).

Acknowledgements We are grateful to D. Bers, A. Gramolini, K. Haghighi, G. Inesi and C. Toyoshima for their helpful comments on this manuscript. The original studies described in this review were supported by grants to D.H.M. from the Heart and Stroke Foundation of Ontario, the Canadian Institutes for Health Research and the Canadian Genetic Diseases Network of Centres of Excellence, and to E.G.K. by grants from the National Institutes of Health (USA).

Online links DATABASES The following terms in this article are linked online to: LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/ AKAPs | ATP2A1 | ATP2A2 | Ca2+/CaM kinase | CACNA1S | CASQ2 | PKA | PMCAs | RYR1 | RYR2 Swiss-Prot: http://www.expasy.ch/ calmodulin | calsequestrin | MKP-1 | PLN | SERCA1a | SERCA2a | SLN Access to this interactive links box is free online.

VOLUME 4 | JULY 2003 | 5 7 7

© 2003 Nature Publishing Group