Abstract
Existing therapies to improve heart function target β-adrenergic receptor (β-AR) signaling and Ca2+ handling and often lead to adverse outcomes. This underscores an unmet need for positive inotropes that improve heart function without any adverse effects. The GTPase Ras associated with diabetes (RAD) regulates L-type Ca2+ channel (LTCC) current (ICa,L). Global RAD-knockout mice (gRAD−/−) have elevated Ca2+ handling and increased cardiac hypertrophy, but RAD is expressed also in noncardiac tissues, suggesting the possibility that pathological remodeling is due also to noncardiac effects. Here, we engineered a myocardial-restricted inducible RAD-knockout mouse (RADΔ/Δ). Using an array of methods and techniques, including single-cell electrophysiological and calcium transient recordings, echocardiography, and radiotelemetry monitoring, we found that RAD deficiency results in a sustained increase of inotropy without structural or functional remodeling of the heart. ICa,L was significantly increased, with RAD loss conferring a β-AR–modulated phenotype on basal ICa,L. Cardiomyocytes from RADΔ/Δ hearts exhibited enhanced cytosolic Ca2+ handling, increased contractile function, elevated sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2a) expression, and faster lusitropy. These results argue that myocardial RAD ablation promotes a beneficial elevation in Ca2+ dynamics, which would obviate a need for increased β-AR signaling to improve cardiac function.
Keywords: calcium channel, cardiomyocyte, GTPase, heart failure, transgenic mice, gene knockout, positive inotrope, RGK GTPase
Introduction
There is an unfulfilled need to identify inotropic agents for the heart (1). However, long-term use of inotropic agents is associated with adverse outcomes in patients (2, 3) that may be related to Ca2+-homeostasis- or protein kinase A-induced maladaptive remodeling (4, 5). The L-type Ca2+ channel complex (LTCC)3 provides trigger Ca2+ for Ca2+-induced Ca2+-release and serves as an upstream control point for modulating the inotropic response. The LTCC complex contains a pore-forming α-subunit (CaV1.2 in the ventricular myocardium) and auxiliary subunits including CaVβ2, α2δ, and calmodulin (6). In addition to established contributions to channel trafficking and gating, CaVβ subunits mediate RGK (Rem, Rad, Rem2, and Gem/Kir) GTPase inhibition of LTCC function in muscle, neuronal, and endocrine cells (7–12). RAD (Ras associated with diabetes) GTPase, was originally identified as a gene up-regulated in the skeletal muscle of a subset of patients with type 2 diabetics (13), and is the founding member of the RGK subfamily of Ras-related small GTPases (14). In heterologous expression systems, RAD shares the common RGK protein property of binding CaVβ subunits and profoundly inhibiting CaV1/CaV2 channel trafficking and activity, with exogenous RAD overexpression potently inhibiting ICa,L in adult and embryonic ventricular myocytes (7, 8, 11, 15, 16). In vivo, RAD deficiency promotes positive inotropic effects (17, 18). In contrast, genetic deletion of the Rem GTPase displays no evidence for inotropic effects secondary to only modest increases of ICa,L (15). RAD is abundantly expressed in the heart, and our findings, as well as others showed that RAD protein levels fall in patients with end-stage nonischemic heart failure (heart failure with reduced ejection fraction, HFrEF) (17), and in a mouse model of cardiac hypertrophy (19), suggesting that RAD down-regulation may be an integral signaling component in myocardial adaptation.
A growing literature indicates that RAD serves as an endogenous inhibitor of myocardial LTCC activity and attenuates β-AR signaling (18, 20, 21). Whole body RAD deletion (gRAD−/−) results in progressive structural changes to the heart wall and increased myocyte size (21), increased up-regulation of the canonical fetal gene program associated with cardiac hypertrophy (21), and cardiac fibrosis (22). In keeping with these molecular changes, RAD-deficient mice are reported to be more susceptible to cardiac hypertrophy (19). These results are in agreement with earlier studies of genetically modified mouse models designed to augment trigger Ca2+. Pore-forming CaV1.2 transgenic mice exhibit increased ICa,L and develop heart failure within about 8 months (23). CaVβ2 overexpressing mice show a more rapid progression to pathological cardiac hypertrophy and death (24). The conclusion from these early studies is that increased ICa,L promotes pathological cardiac hypertrophy.
RAD protein is expressed in myocardial and nonheart tissues including, but not limited to, vascular smooth muscle (25) and in cells in the bone marrow (26). Thus the complex cardiac phenotype observed in gRAD−/− mice may include noncardiac influences on cardiac remodeling that are superimposed over direct positive inotropic effect arising from increased trigger Ca2+-current in the myocardium. The purpose of this study is address the key question: are pro-adaptive, positive-inotropic effects of RAD-deletion obscured by the loss of RAD in noncardiac cell types? A key hypothesis maintains that RAD deletion, by targeting LTCC downstream of β-AR signaling provides a gain of systolic function. Therefore, we engineered an inducible, myocardial-restricted RAD knockout mouse (RADΔ/Δ). Our results show that RADΔ/Δ mice have enhanced heart function into senescence without evidence for structural remodeling. These major new findings challenge the dogmatic assumption that increased myocardial Ca2+ necessarily promotes pathology, and suggests that the heart pathology observed in gRAD−/− mice are the result of noncardiac influences of RAD loss.
Results
Development of conditional cardiac-restricted RAD-deficient transgenic mice
Standard gene targeting of RRAD in the mouse results in a complex phenotype, including cardiac remodeling (21) and fibrosis (22), with increased cardiac output (18). To examine the role of RAD signaling selectively in cardiomyocytes, we used conditional gene targeting with the Cre-loxP system. To achieve cardiomyocyte-specific deletion, CRISPR/Cas9 techniques were used to genetically target RRAD, introducing loxP (flox: fl) recombination sites flanking exons 3 and 4 (RADfl/fl; Fig. 1A). These exons encode a large portion of the RAD GTPase core that directs both guanine nucleotide binding and hydrolysis, and the C-terminal membrane targeting motif required for LTCC regulation (27) such that any transcribed mRNA that is translated following recombination would produce a truncated protein lacking biological activity. Successful introduction of loxP sites was confirmed by genomic PCR (Fig. 1B). To permit cardiomyocyte-specific RAD deletion, RADfl/fl mice were crossed with a mouse line expressing a tamoxifen-inducible Cre recombinase under control of the α-myosin heavy chain promoter (αMHC-CreERT2) (28). RAD is abundantly expressed in the heart and other tissues (8, 13, 25, 26, 29), and administration of tamoxifen to RADfl/fl-MHC mice (Fig. 1A) resulted in RAD deletion from the myocardium (RADΔ/Δ) but not the spleen (Fig. 1C). Disruption of RAD expression in the heart was confirmed by qRT-PCR (Fig. 1D). RADfl/fl without αMHC-CreERT2 (RADfl/fl) mice served as littermate controls, with RAD expression unaffected following tamoxifen challenge (Fig. 1, C and D). Unless stated, all RADΔ/Δ and RADfl/fl mice were subjected to analysis >2 weeks following tamoxifen exposure.
Figure 1.
Myocardial deletion of RAD. A, graphic representation of the RRAD conditional targeting strategy. Flanking exons 3 and 4 of RRAD with loxP sites generated the floxed allele (fl). Cre-mediated recombination of the loxP sites results in the cKO allele (Δ). The approximate locations of the PCR primers used for genotyping (a and b) are shown. B, genotyping PCR of tail genomic DNA yielded the following genotypes: RRADwt (lane 1), RRADw/fl (lane 2), and RRADfl/fl (lane 3). Amplicons were generated by primers a and b as shown in A. C, Western blot analysis for RAD in protein lysates from wildtype (WT), global RAD knockout (gRADKO), RADfl/fl, and RADΔ/Δ (2 weeks post-tamoxifen treatment) from total heart (top panel) and spleen (bottom panel). Note that RAD expression is retained in the spleen of RADΔ/Δ mice following tamoxifen treatment. D, qRT-PCR analysis in mRNA samples from RRADfl/fl and RRADΔ/Δ hearts. MW, 100 bp DNA ladder. E, quantification by Western blotting of RAD levels in the heart 4 weeks post-tamoxifen treatment (n = 7); ***, p = 0.001.
Cardiac-restricted RAD-deficient mice (RADΔ/Δ) show improved function with no changes in heart dimensions
Global constitutive RAD knockout mice (gRAD−/−) showed increased ICa,L (18) along with hypertrophic fetal gene program expression, structural remodeling (19), and fibrosis (22). By contrast, ANF and RCAN1, markers of the fetal gene program, were not altered in RADΔ/Δ (Fig. 2A). There was also no difference in the ratio of heart to body weight (Fig. 2B) and RADΔ/Δ shows no significant difference in fibrosis compared with RADfl/fl (Fig. 2, C and D). This is in stark contrast to gRAD−/− hearts, in which RAD deficiency has been found to increase connective tissue growth factor (CTGF) expression, leading to greater extracellular matrix production and basal cardiac fibrosis (22). Importantly, cardiomyocyte-restricted RAD deletion failed to increase CTGF expression (Fig. 2, E and F). Global RADKO mice experience hypertension (50), and this may be a significant driver of the cardiac remodeling observed in these mice, as gRAD−/− hearts have likely undergone adaptive alterations secondary to pressure overload. Importantly, RADΔ/Δ showed no change in aortic pressure (Fig. 2G). Taken together, these data demonstrate that myocardial-restricted RAD deficiency improves cardiac output without instigating structural remodeling.
Figure 2.
Cardiomyocyte-restricted RAD deletion does not induce markers of myocardial pathology. A, qRT-PCR for ANF and RCAN1 mRNA expression levels. RADfl/fl (n = 6) and RADΔ/Δ (n = 5) as shown. B, heart weight was not significantly different between RADΔ/Δ and RADfl/fl. C, representative pictures of PicroSirius Red staining with fibrosis quantification in D. RADfl/fl (n = 5) and RADΔ/Δ (n = 7) are shown. E, Western blot analysis with quantification in F demonstrates no difference in CTGF expression. Lanes were run on the same gel, but noncontiguous lanes are marked by the black line. Radfl/fl (n = 7) and RADΔ/Δ (n = 9) are shown. **, p = 0.008. G, aortic pressure recordings, 4 continuous days from baseline (prior to tamoxifen), and from the same mice 2 weeks after tamoxifen administration (RADΔ/Δ). Data are average of 3 mice. Data were fitted to a sine wave with a fixed cycle length of 24 h. Active (nighttime) and resting (daytime) blood pressures were 129 ± 2 mm Hg (baseline), 132 ± 5 mm Hg (0.5 months); and 118 ± 2 mm Hg (baseline), 122 ± 6 (0.5 months), respectively.
To test the effect of myocardial-restricted RAD deficiency on heart function we longitudinally followed RADΔ/Δ mice from induced knockout in early adulthood for an additional 15 months (Fig. 3A). Ejection fraction (EF) significantly increased 7 days after tamoxifen administration and remained increased continuously for 15 months (Fig. 3B). There was no difference in heart chamber size (Fig. 3C) or in heart wall thickness (Fig. 3, D and E). Cardiomyocyte-restricted RAD deletion effects on in vivo heart function were not different between males and females (Fig. 3, F–H). EF has a shallow dependence on heart rate (HR) that is preserved in RADΔ/Δ. Regression analysis supports elevated EF in RADΔ/Δ across the range of HR measured (Fig. 3I).
Figure 3.
Myocardial RAD deletion results in a rapid, stable gain of cardiac function without pathological structural remodeling in vivo. A, representative M-mode short-axis echocardiography of female mice 1-week through 15 months following tamoxifen injection. Scale bars of depth: RADfl/fl baseline = 0.69 mm; RADfl/fl 15 months = 0.67 mm; RADΔ/Δ baseline = 0.69 mm; RADΔ/Δ 15 months = 0.76 mm; all images have a duration of 1 s. B, ejection fraction; C, left ventricular inner dimension; D, left ventricular anterior; and E, posterior wall thickness. Dimensions (C–E) are shown in diastole; RADfl/fl (n = 7) and RADΔ/Δ (n = 17; by genetic modification, p = 0.0002; F = 20; by time p = 0.04; F = 2; by gene × time, p = 0.04; F = 2). F–H, echocardiography of male and female mice 3 months after tamoxifen treatment. F, ejection fraction. Sidak's multiple comparison test showed RADΔ/Δ increased in EF (p < 10−3 males, p < 10−4 females; by genetic modification, p < 10−4, F = 49; by sex, p < 0.01; F = 8; no interaction). RADfl/fl 2-way ANOVA shows female gender contributed 8% to variance suggesting more sensitivity than males to RADΔ/Δ (p < 0.06). G, anterior wall thickness and H, posterior wall thickness during diastole was not different by RADΔ/Δ or by male versus female. I, linear regression of EF as a function of HR shows HR differences do not account for elevated EF in RADΔ/Δ. The dashed lines show 95% confidence limits. The slope is not different between RADΔ/Δ and RADfl/fl; elevation of EF is significantly different (p < 10−4). **, p = 0.006; ****, p < 10−4.
RADΔ/Δ modification of voltage-gated calcium current, ICa,L
A straightforward explanation for enhanced heart function would be increased ICa,L in RADΔ/Δ. Previous studies of whole body, constitutive RAD knockout showed that LTCC conductance increased, voltage dependence shifted negatively, and the predicted ICaL window increased (18); however, constitutive global RAD knockout hearts displayed structural remodeling. Thus, it is possible that changes in gRAD−/− ICa,L were secondary to myocardial remodeling. To interrogate the contribution of RAD in structurally unaltered hearts, we measured ICa,L in cardiomyocytes from mature RADΔ/Δ hearts. Fig. 4A shows representative families of ICa,L traces from RADfl/fl and RADΔ/Δ for evaluating current/voltage (I(V)) relationships, and Fig. 4B shows representative current traces generated by the steady-state availability voltage protocol. These representative traces highlight the increased current density and accelerated kinetics of RADΔ/Δ. The I(V) curves for ICa,L show that RAD loss results in a greater current density compared with RADfl/fl myocytes (Fig. 4C). RAD deficiency results in greater maximal conductance (Fig. 4, D and E; maximal conductance: RADΔ/Δ = 254 ± 19 pS/pF, n = 15; RADfl/fl = 144 ± 12 pS/pF, n = 18; p < 10−4). The normalized conductance voltage curves superimposed on the steady-state inactivation curves (Fig. 4F) highlight the selective shift of steady-state activation with RAD ablation; activation midpoint is shifted toward more negative membrane potential (Fig. 4, F and G; RADΔ/Δ = −18.3 ± 1.0 mV, n = 15; RADfl/fl = −8.1 ± 1.9 mV, n = 18; p < 10−4). Steady-state availability shows no significant difference of midpoint between RADfl/fl and RADΔ/Δ (RADΔ/Δ = −24.2 ± 1.0 mV, n = 9; RADfl/fl = −22.1 ± 1.7 mV, n = 11; p = 0.11). There is no sexual dimorphism in the effect of RAD deletion on ICa,L (data not shown). The increase in ICa,L could stem from an increase in CaV1.2. To test this, we performed qRT-PCR for CaV1.2 from RADfl/fl and RADΔ/Δ hearts and found CaV1.2 mRNA did not change (Fig. 4H). CaV1.2 protein levels from RADΔ/Δ hearts was also not significantly different from that of RADfl/fl (Fig. 4I). Taken together these data show that myocardial-restricted RAD deficiency is sufficient to increase ICa,L by modulating LTCC function.
Figure 4.
Myocardial RAD deletion results in increased calcium current (ICa,L). A, representative family of ICa,L traces, Vtest ranging from −75 to +45 mV in 10-mV increments, with voltage protocol schematic shown above. B, representative family of ICa,L traces from Vpre ranging from −50 to 0 mV in 10-mV increments for determining steady-state availability, with voltage protocol schematic shown above. Scale bars: 500 pA and 200 ms. Scale bars common to panels A and B. C, current/voltage curve shows that ICa,L density is increased in cardiomyocytes from RADΔ/Δ compared with RADfl/fl. D, conductance transform of the current/voltage curve demonstrates higher maximal conductance in RADΔ/Δ compared with RADfl/fl with quantification shown in E. F, conductance-voltage curve normalized to maximal conductance illustrates that RADΔ/Δ cardiomyocytes have shifted the activation midpoint. Smooth curves are Boltzmann distribution fitted to data. Steady-state availability was not different. Note that voltage for the availability curve is the value for the 500-ms pre-pulse potential step (see schematic in A). G, activation midpoint is significantly negative-shifted in RADΔ/Δ. Current data are from RADfl/fl (n = 7 mice, n = 18 cells) and RADΔ/Δ (n = 7 mice, n = 17 cells); ****, p < 10−4. H, qRT-PCR for CaV1.2 showed no change in expression. Data were displayed from RADfl/fl (5 mice) and RADΔ/Δ (5 mice). I, Western blot analysis demonstrates no difference in CaV1.2 expression between RADfl/fl and RADΔ/Δ. Lanes were run on the same gel, but noncontiguous lanes are marked by the black line; total protein stained with Coomassie Blue and quantification are shown on the right. Western blotting data from RADfl/fl (7 mice) and RADΔ/Δ (9 mice).
RADΔ/Δ ICa,L decay is biphasic with a prominent fast and slow decay phase compared with monophasic decaying RADfl/fl (Fig. 4A; Fig. S1). At −5 mV the fast and slow decaying amplitudes of RADΔ/Δ ICa,L are roughly similar (Fig. S1); the slow-decay component is significantly slower than RADfl/fl decay. The fast ICa,L decay is >10-fold faster than the monotonic ICa,L time constant observed in RADfl/fl (Fig. S1). In summary, myocardial RAD-reduction results in larger, but faster decaying ICa,L.
RADΔ/Δ enhances cellular calcium homeostasis
ICa,L provides trigger Ca2+ for myocardial Ca2+-induced Ca2+ release. Whole-cell cytosolic Ca2+ dynamics are significantly larger in amplitude for RADΔ/Δ compared with RADfl/fl (Fig. 5, A and B), and the upstroke velocity of the cytosolic Ca2+-transient is accelerated in RADΔ/Δ (Fig. 5C). To assess Ca2+ re-uptake we measured the time constant of the Ca2+ transient decay (τ). The value of the mean τ of RADΔ/Δ was faster than that for RADfl/fl but did not reach statistical significance using ratiometric imaging of fura2 in cardiomyocytes (Fig. 5D). We augmented ratiometric imaging with a separate series of experiments using fluo4 for high speed 2-dimensional cytosol-restricted Ca2+ dynamics. A benefit of fluo4 imaging is the elimination of contaminating, slower nuclear Ca2+ signals. Two-dimensional cytosolic Ca2+ decay was statistically significantly faster in RADΔ/Δ than RADfl/fl (Fig. 5E; RADΔ/Δ = 0.07 ± 0.003, n = 67, RADfl/fl = 0.10 ± 0.005, n = 69; p < 10−4). To probe mechanisms of re-uptake we assessed SERCA2a protein levels (Fig. 5F). RADΔ/Δ hearts expressed elevated SERCA2a protein levels (p < 0.05) but unchanged phospholamban (PLN) protein levels (see below). Taken together these data suggest that faster ICa,L trigger (Fig. 4) and increased SERCA2a expression conspire to contribute to accelerate Ca2+ transient dynamics following RAD knockout.
Figure 5.
RADΔ/Δ enhances cellular calcium handling. A, representative calcium transients from isolated ventricular cardiomyocytes loaded with fura2-AM, RADfl/fl (top) and RADΔ/Δ (bottom) paced at 1 Hz. B, amplitude of the transients from RADΔ/Δ are higher than in RADfl/fl. C, the velocity at which calcium enters the cytosol (upstroke of the transient) is faster in RADΔ/Δ than in RADfl/fl. D and E, calcium transient decay is faster in RADΔ/Δ than in RADfl/fl. Measurements in E used fluo4-AM and high speed 2-dimensional imaging. Measurements in A–D used fura2-AM, RADfl/fl (n = 10 mice, n = 38), RADΔ/Δ (n = 8 mice, n = 49). For E, RADfl/fl (n = 10 mice, n = 69) and RADΔ/Δ (n = 8 mice, n = 67). F, Western blot analysis demonstrates increased expression of SERCA2a, quantification is shown on the right (p = 0.035). Lanes were run on the same gel, but noncontiguous lanes are marked by the black line. Below, total protein stained with Coomasie Blue. RADfl/fl (7 mice) and RADΔ/Δ (8 mice) are shown. *, p = 0.014; **, p = 0.008; ****, p < 10−4.
Sarcomere dynamics is an important determinant of myocardial function (30). We therefore measured sarcomere length and dynamics (Fig. 6A) simultaneously with calcium transients. Elevated calcium dynamics (Fig. 5) accompanying RAD deficiency coincided with unchanged resting sarcomere length (Fig. 6, A and B), increased fractional shortening (Fig. 6C; RADΔ/Δ = 16.1 ± 0.9, n = 24, RADfl/fl = 9.9 ± 1.0, n = 15; p < 10−4), and the rate at which the cell shortened (Fig. 6D; RADΔ/Δ = −4.8 ± 0.4, n = 24; RADfl/fl = −2.8 ± 0.3, n = 15; p = 0.0002). Overall, these results suggest an increase in contractility in RADΔ/Δ compared with RADfl/fl, consistent with cellular dynamics driving improved heart function with RADΔ/Δ.
Figure 6.
RADΔ/Δ enhances sarcomere shortening and increases the tension-integral. A, representative sarcomere length traces of RADfl/fl and RADΔ/Δ paced at 1 Hz. B, diastolic sarcomere length was not different between RADfl/fl and RADΔ/Δ. C, sarcomere fractional shortening was higher in RADΔ/Δ than in RADfl/fl. D, the velocity of shortening was faster in RADΔ/Δ than in RADfl/fl. E, integral of sarcomere length is larger for RADΔ/Δ than in RADfl/fl. RADfl/fl (n = 7 mice, n = 15 cells) and RADΔ/Δ (n = 8 mice, n = 24 cells) are shown. ***, p < 0.001; ****, p < 10−4.
Davis et al. (31) introduced a predictive model of cardiac growth based on integrated tension measured from mean twitch tensions or sarcomere shortening. A negative score of this tension-integral model predicts dilated cardiomyopathy; a positive score trends toward hypertrophic remodeling. Using this model, we calculated the index score for our RADΔ/Δ model to predict the direction and intensity of hypertrophy. The RADΔ/Δ tension index score is +0.03 relative to RADfl/fl (Fig. 6E), predicting only a modest tendency toward concentric hypertrophy. This provides novel evidence that increasing trigger calcium does not necessarily promote significant cardiac hypertrophy and pathological remodeling.
Basal ICa,L in RADΔ/Δ cardiomyocytes reflects a modulated ICa,L
The increase of ICa,L in response to β-adrenergic stimulation is atop a constellation of effectors that enhances cardiac output in the fight or flight response. We therefore next tested the effect of acute isoproterenol (ISO) treatment on ICa,L and calcium dynamics. RADfl/fl ventricular cardiomyocyte ICa,L conductance was significantly increased (Fig. 7, A and Bi, Fig. S2); however, ISO had no detectable effect on RADΔ/Δ ICa,L (Fig. 7, A and Bii, Fig. S2) resulting in ISO-treated RADfl/fl conductance rising to a level that was indistinguishable from that of unstimulated RADΔ/Δ cardiomyocytes (Fig. S2). Comparison of the voltage-dependence of activation of ICa,L reveals that the acute ISO effect in RADfl/fl, whereas shifted negative, remained more positive than that for RADΔ/Δ (Fig. 7, D and E, Fig. S2). Acute ISO had no detectable effect on ICa,L kinetics (data not shown). Two summary conclusions are apparent: 1) RADΔ/Δ ICa,L is maximally modulated; and 2) RADfl/fl–modulated ICa,L remains at a more positive activation midpoint potential than that for RADΔ/Δ. We conclude that basal RADΔ/Δ phenocopies β-AR–modulated RADfl/fl ICa,L.
Figure 7.
ISO does not alter RADΔ/Δ ICa,L. A, representative ICa,L traces at Vtest of −5 mV before (black line) and after treatment with ISO (colored line; blue for Radfl/fl and red for RADΔ/Δ). Scale bars: 500 pA and 200 ms. B, current/voltage curves superimposing basal and acute 1 μm ISO treatment for RADfl/fl (B, i) and RADΔ/Δ (B, ii). C, fractional change of maximal conductance between baseline and ISO response of RADfl/fl (mean = 32.68%) and RADΔ/Δ (mean = 0.84%). D, conductance-voltage curve normalized to maximal conductance for ISO-treated cells demonstrates RADΔ/Δ cardiomyocytes have negative shifted activation midpoint relative to RADfl/fl in ISO. Smooth curves are Boltzmann distribution fitted to data. Steady-state availability was not different. E, difference between activation midpoints between baseline and ISO response of RADfl/fl (mean = −4.47) and RADΔ/Δ (mean = −0.19). RADfl/fl (n = 4 mice) and RADΔ/Δ (n = 4 mice) are shown. *. p = 0.02.
RADΔ/Δ basal heart function is elevated in vivo, yet retains a partial ISO response
Given that RAD deficiency results in ICa,L resembling β-AR augmentation we next performed an echocardiography ISO stress test in RADfl/fl and RADΔ/Δ mice at 1 and 12 months after tamoxifen administration. Acute in vivo ISO resulted in a significant gain of EF in RADfl/fl and in RADΔ/Δ mice at 1-month post-tamoxifen treatment (Fig. 8). The ISO effect was preserved at 1 year (16-month-old mice). We conclude that myocardial-restricted RAD deletion provides stable inotropic support, without globally disrupting myocardial β-AR responsiveness.
Figure 8.
β-AR–mediated responsiveness retained following RAD deletion in vivo. Echocardiography of mice scanned at baseline (filled symbols) and after injection of isoproterenol (open symbol with connecting line). Ejection fraction significantly increased with addition of isoproterenol in both RADfl/fl and RADΔ/Δ at 1- and 12-months after tamoxifen administration. RADfl/fl (n = 7) and RADΔ/Δ (n = 17) are shown. ****, p < 10−4.
RADΔ/Δ retains β-adrenergic receptor modulation of Ca2+ and sarcomere dynamics
To explore the source of ISO responsiveness in RAD knockout cardiomyocytes we next interrogated Ca2+ and sarcomere dynamical response to ISO. Acute ISO enhanced calcium dynamics in RADΔ/Δ and RADfl/fl ventricular cardiomyocytes (Fig. 9A). For cytosolic Ca2+ handling, RADfl/fl ISO response demonstrated an increase in amplitude, a faster upstroke velocity, and a faster rate of calcium reuptake compared with basal conditions, as expected (Fig. 9, B–D, Fig. S3). RADΔ/Δ ISO response also demonstrated an increase in amplitude, faster upstroke velocity, and a faster rate of Ca2+-reuptake. Compared with RADfl/fl with ISO, RADΔ/Δ with ISO had a relatively larger amplitude (RADΔ/Δ = 2.6 ± 0.1, n = 49; RADfl/fl = 2.0 ± 0.2, n = 37; p = 0.008), faster upstroke velocity (RADΔ/Δ = 63 ± 3.0, n = 49; RADfl/fl = 47 ± 5.0, n = 37; p = 0.009), and faster rate of calcium reuptake (RADΔ/Δ = 0.11 ± 0.003, n = 49; RADfl/fl = 0.13 ± 0.006, n = 38; p = 0.05; Fig. S3). To further test for active β-AR signaling in RADΔ/Δ we assessed PLN-serine 16 phosphorylation status. At baseline, PLN Ser-16P and Thr-17P relative to total PLN protein were not different between RADΔ/Δ and RADfl/fl (Fig. 9E). Acute ISO stimulation significantly increase phosphorylation of PLN–Ser-16P protein from RADΔ/Δ hearts (Fig. 9F) providing a basis for accelerated Ca2+-reuptake.
Figure 9.
Cardiomyocyte cytosolic Ca2+ handling responds to β-AR–mediated activation in RADΔ/Δ. A, representative calcium transients from RADfl/fl and RADΔ/Δ cardiomyocytes paced at 1 Hz in 1 μm ISO. B, fractional change (ISO/baseline) of Ca2+ transient amplitude. C, upstroke velocity, and D, rate of decay. RADfl/fl (n = 10 mice, n = 38 cells) and RADΔ/Δ (n = 8 mice, n = 49 cells) are shown. E, Western blot analysis of total and phosphorylated PLN, RAD, and calsequestrin (15% SDS-PAGE). Lower panel, quantification shows no significant effect of myocardial Rad deletion. RADfl/fl (n = 6 mice) and RADΔ/Δ (n = 9 mice) are shown. F, acute ISO increased PLN Ser-16P in RADΔ/Δ (4–20% SDS-PAGE). RADΔ/Δ (n = 5 mice) is shown. For all panels: *, p < 0.05, **. p = 0.005; ****, p < 10−4.
To test for preservation of acute β-AR signaling to contractile function we measured sarcomere dynamics in response to isoproterenol (Fig. 10A). Fractional shortening (Fig. 10B, Fig. S4) and sarcomere relaxation was accelerated in RADΔ/Δ (Fig. 10C, Fig. S4). Acute ISO elevated the tension-integral index in RADfl/fl approaching the level observed for RADΔ/Δ (Fig. 10, D and E). To further investigate contractile mechanisms underlying the response to ISO, we assessed the phosphorylation status of sarcomeric proteins that modulate contractility (32, 33). Acute ISO significantly increased phosphorylation of cMyBPC and TnI (Fig. 10F). These data support the conclusion that RADΔ/Δ does not exhibit de-sensitized myocardial β-AR signaling. The reduction of RAD imposes a chronic modulated ICa,L while preserving β-AR modulation of critical targets, such as sarcoplasmic reticulum Ca2+-reuptake and contractile apparatus proteins.
Figure 10.
Sarcomere dynamics and proteins respond to β-AR–mediated activation in RADΔ/Δ. A, representative sarcomere traces of RADfl/fl and RADΔ/Δ paced at 1 Hz after treatment with 1 μm ISO. B, fractional change (ISO/baseline) in fractional shortening, and C, relaxation velocity. ISO significantly increases shortening and relaxation velocity in RADΔ/Δ. D, integral of sarcomere length is larger in RadΔ/Δ than in RADfl/fl. E, ISO treatment increased the tension-integral index in RADfl/fl but not in RADΔ/Δ. RADfl/fl (n = 3 mice, n = 8 cells) and RADΔ/Δ (n = 5 mice, n = 16 cells) are shown. F, Coomassie stain and ProQ phosphostain analysis demonstrating increased phosphorylation of proteins from RADΔ/Δ hearts after treatment with ISO of cMyBPC and TnI, quantification to the right. Saline, n = 6 mice; and ISO, n = 6 mice were used. For all panels: *, p = 0.01; **, p = 0.0002; ****, p < 10−4.
Discussion
There is need for inotropic agents; however, targeting excitation-contraction coupling has been met with failure (34). Consistent with this literature, early findings with constitutive global RAD−/− (gRAD−/−) suggested that elevated trigger Ca2+ led to increased cardiac hypertrophy (19). The importance of the present study is the finding that cardiomyocyte-restricted RAD deletion results in a salutary cardiac phenotype, devoid of the wall remodeling observed in gRAD−/− mouse studies. Here, we successfully achieved conditional cardiomyocyte selective RAD deletion. Through functional measurements and biochemical analysis, we demonstrate that: 1) RAD is expressed in cardiomyocytes of the heart and myocardial RAD deficiency promotes chronic enhanced heart function without structural remodeling or fibrosis. 2) Cardiomyocyte-specific RAD deletion leads to enhanced basal ICa,L resembling β-AR modulated ICa,L. 3) Myocardial physiological function (Ca2+ handling and sarcomere dynamics) remain β-AR responsive. Moreover, RAD protein is reduced in nonischemic heart failure patients (17) suggesting that regulation of RAD contributes to a compensatory response.
There are two dogmatic paradigms that our present results challenge: first, that increases of calcium dynamics necessarily promote pathogenic cardiac hypertrophy; and second, that absence of RAD exacerbates pathological cardiac remodeling (15, 22). Each of these are discussed below.
Increased calcium does not necessarily promote cardiac hypertrophy
A major finding of this study is that myocardial RAD deletion, with increased ICa,L, did not demonstrate structural remodeling (Fig. 3). Previous studies have reported a connection between calcium influx through the LTCC and adverse growth of the heart wall (24, 36, 37). Increasing ICa,L through overexpression of the pore subunit α1C or the β2 subunit of the LTCC led to cardiac hypertrophy, and eventual heart failure growth (38). An ICa,L phenotypic difference with these over-expression models and RADΔ/Δ is in the kinetics of ICa,L. RADΔ/Δ demonstrates a biphasic decay of current with fast and slow components (Fig. 4, Fig. S1). The time course of ICa,L is reminiscent of calcium-dependent fast inactivation contributed by the calcium load of the sarcoplasmic reticulum (39). Fast decay in RADΔ/Δ ICa,L attributed to a large calcium release from the SR might serve as a regulatory negative feedback mechanism to limit total LTCC Ca2+ flux, and in turn might not necessarily activate pathways to initiate pathological hypertrophy. Thus, even if dynamical total Ca2+ influx was increased in the intact myocardium, there may not be obligatory hypertrophic signaling by LTCC (40).
Over-expression of SERCA2a preserves myocardial function under conditions that otherwise would be expected to produce failure (41–44). RADΔ/Δ hearts display increased SERCA2a expression without a detectable change in PLN phosphorylation (Figs. 5 and 9), resulting in a change in the ratio of SERCA2a to PLN that is consistent with overall faster Ca2+ transient decay dynamics. This favorable alteration toward less regulated SERCA2a complexes in our model has also demonstrated cardiac protection in high-intensity exercise models (41) and PLN-null mice (47), as well as in phase 1 and phase 2 trials of patients suffering from advanced heart failure (43, 44). To summarize, we observe contemporaneous mechanistic changes in RADΔ/Δ myocardium that plausibly sum to a stable gain of heart function including: increased, faster-decaying trigger Ca2+ and increased SERCA2a.
Davis et al. (31) proposed a tension-integral index model that distinguishes between hypertrophic and dilated cardiac remodeling. A key insight of the tension-integral index study was that the tension-integral predicted myocardial structural changes, but there were poor correlations to changes in calcium handling indices (31). The RADΔ/Δ approximates a WT tension-integral as a consequence of larger amplitude contractions offset by faster kinetics. Regardless, the RADΔ/Δ heart showed elevated EF. Thus, how RADΔ/Δ accommodates the increased demands of chronic increased cardiac function will be an important area of follow-up studies.
In a similar vein to sarcomere dynamics, the faster kinetics of ICa,L in RADΔ/Δ could also potentially serve to attenuate arrhythmogenic potential. Mahajan et al. (46) proposed modifying ICa,L kinetics as a superior way to prevent arrhythmias, rather than through inhibiting ICa,L activation. This would allow early peak ICa,L to initiate normal SR calcium release, and diminish the effects of late ICa,L on repolarization. As encouraging as the present results are for suggesting myocardial RAD deletion as a therapeutic direction for cardioprotection, thorough analysis of arrhythmogenic potential will require further studies, including, but not limited to alternative model systems to the mouse.
The RADΔ/Δ model also differs from other models of increased ICa,L because responsiveness to β-adrenergic receptor (β-AR) stimulation is retained. A hallmark feature of HFrEF is insensitivity of β-AR to chronic stimulation (47) that ultimately limits the contractile reserve of the heart. RADΔ/Δ hearts retain β-AR responsiveness at the level of the cell (Figs. 9 and 10) and whole heart (Fig. 8), and did not develop heart failure. ICa,L did not change after treatment with isoproterenol in our RADΔ/Δ model (Fig. 7), similar to overexpression of the α1C model (12). ICa,L was larger in RADΔ/Δ than in RADfl/fl stimulated with ISO. This suggests that RAD may play a critical role in the ability for the LTCC to be modulated by β-AR stimulation; when RAD is deleted, basal ICa,L is larger than currently seen following typical modulation. Further studies are warranted to define the molecular role for RAD in β-adrenergic Ca2+ current modulation. Finally, enhancing Ca2+ dynamics via targeting of β-AR signaling by G-protein coupling receptor kinase (48) or Raf kinase inhibition (49) also provides protection against pressure overload induced heart failure. In summary, increasing Ca2+ dynamics in a manner that bypasses chronic β-AR activity is a potentially attractive target of therapeutic strategies for heart failure therapy.
Myocardial-restricted RAD deletion differs from whole body RAD deletion
Key differences exist between RADΔ/Δ and the whole body deletion of RAD leading to the existing erroneous conclusion that RAD-reduction is pro-hypertrophic (19). In the global RAD knockout model (gRAD−/−), channel function and cellular calcium dynamics were similar to RADΔ/Δ, and in both models the myocardium retained responsiveness to β-AR stimulation (18). However, myocardial RAD deficiency promotes enhanced heart function without structural remodeling or fibrosis. A major consideration is that gRAD−/− mice experience hypertension (50), whereas RADΔ/Δ do not. This may result from loss of RAD in vascular smooth muscle (25, 45); increased ICa,L in vascular smooth muscle could lead to increased vascular resistance to progress toward hypertension. Regardless, gRAD−/− hearts have likely undergone adaptive signaling and therefore interpretation of studies of Ca2+ dynamics in gRAD−/− must consider the myriad of myocardial alterations secondary to pressure overload.
In summary, our findings suggest that increasing myocardial calcium handling does not necessarily promote pathological remodeling, in fact, by imposing a chronic modulated ICa,L phenotype in the basal state we show that heart function increases stably into senescence. These findings are consistent with the notion that decreased RAD in heart failure patients (17, 19) contributes to a compensatory response. There is a major unmet need for treatments targeting inotropy (1). We conclude that myocardial RAD deletion might provide a potential new therapeutic target for positive inotropy without introducing damaging effects to the heart.
Experimental procedures
All experimental procedures and protocols were approved by the Animal Care and Use Committee of the University of Kentucky and conformed to the National Institute of Health “Guide for the Care and Use of Laboratory Animals.”
Animal model
Transgenic animals were generated on a C57BL/6J background in the Transgenic Animal and Genome Editing Core at Cincinnati Children's Hospital Medical Center. The conditional RRAD allele was made via a sequential insertion strategy using CRISPR-Cas9 technology, in which exons 2 and 3 were flanked by Cre recombinase-dependent loxP (flox: fl) recognition sequences (see Fig. 1A). The sgRNA's were engineered to contain either HindIII (5′) or EcoRI (3′) restriction sites, with offspring screened by genomic PCR analysis, and loxP insertion confirmed by Sanger sequencing. These mice (RADfl/fl) were then crossed (or not, as controls) with mice expressing a tamoxifen-inducible Cre recombinase (MerCreMer) under α-myosin heavy chain promoter (28) to produce RADfl/fl-MHC animals. RAD deficiency was induced by a single intraperitoneal injection in control (RADfl/fl) and experimental mice with tamoxifen dissolved in sunflower seed oil (40 mg/kg body weight). All mice received tamoxifen and were used ≥2 weeks after tamoxifen treatment unless otherwise stated. This single tamoxifen injection protocol minimizes cardiomyopathological effects observed with multiday administration of tamoxifen (35). The age of the oldest mice used for study were limited to 18 months, but the cellular studies were from mice <12 months of age.
Immunoblotting
Hearts freshly excised from RADfl/fl and RADΔ/Δ were flash frozen, and tissue was pulverized using Freezer Mill (6775, SPEX SamplePrep) before sonication in cell lysis buffer containing (in mmol/liter), 50 Hepes, pH 7.5, 150 NaCl, 0.5% Triton X-100, 1 EDTA, 1 EGTA, 10 NaF, 2.5 NaVO4, complete Protease Inhibitor Mixture (Roche 11697498001-1 tablet per 50 ml), phosphatase inhibitor mixture 2 (Sigma, P5726, ×100 solution), and phosphatase inhibitor mixture 3 (Sigma, P0044, ×100 solution) (500 μl of each inhibitor mixture). Lysates were centrifuged at 14,000 rpm and protein concentrations of supernatants were measured. Lysates were prepared in 4× SDS-PAGE loading buffer with 40.0 μg of total protein per lane resolved on 8, 15, or 4–20% SDS-PAGE gels and transferred to nitrocellulose/PVDF membranes. Immunoblotting was performed with: calsequestrin (1:3000, Abcam); RAD (1:2000, Everest Biotech); Cav1.2 (1:1000, Alomone); CaMKIId (1:1000, Santa Cruz Biotechnology); pCaMKII (1:1000, Santa Cruz Biotechnology); SERCA2a (1:1000, Badrilla); phospholamban phosphoserine 16 (1:1000, Badrilla); phosphor-threonine 17 (1:1000, Badrilla), and total PLN (1:1000, Invitrogen) antibodies, CTGF (1:1000, Santa Cruz Biotech) and GAPDH (1:1000, Cell Signaling Technology). Proteins were detected using SuperSignal enhanced chemiluminescence (Pierce), and immunoblots were developed and quantified using the Bio-Rad ChemiDoc MP Imaging System. The total protein loaded was quantified by staining the membranes with Coomassie staining after the detection of the desired proteins or using stain-free gels (Bio-Rad).
Quantification of myofibril protein phosphorylation
Myofibrils were prepared from frozen mouse ventricular tissue homogenized in relaxing solution containing protease and phosphatase inhibitors (PhosSTOP and cOmplete ULTRA Tablets, Roche Applied Science). Samples were skinned for 15 min using 1% Triton X-100, centrifuged at 10,000 × g for 5 min, and resuspended in fresh relax. Samples were reduced using 2-mercaptoethanol in Laemmli buffer and boiled for 5 min. 5 μg of the solubilized myofibrils were loaded and electrophoretically separated using 4–20% Tris glycine minigels at 160 V for 70 min. Gels were fixed and stained with Pro-Q diamond phosphoprotein stain (Invitrogen) per the manufacturer's protocol. The same gels were then stained with Coomassie Blue for total protein expression. Densitometry of bands in Pro-Q and Coomassie-stained gels was performed using ImageJ software (National Institutes of Health). The degree of phosphorylation of a given protein was determined by the ratio of the Pro-Q signal to the protein contented loaded.
Ventricular myocyte isolation
Isolated ventricular cardiomyocytes were prepared as previously described (15). Prior to heart excision mice were anesthetized with ketamine + xylazine (90 + 10 mg/kg intraperitoneal). Hearts were excised from adult RADfl/fl and RADΔ/Δ and immediately perfused on a Langendorff apparatus with a high-potassium Tyrode buffer and then digested with 5 to 7 mg of liberase (Roche Applied Science). After digestion, atria were removed and ventricular myocytes were mechanically dispersed. Calcium concentrations were gradually restored to physiological levels in a stepwise fashion, and only healthy quiescent ventricular myocytes were used for electrophysiological analysis or calcium imaging within 12 h.
Single-cell assays
Electrophysiological recordings and calcium transients
ICa,L was recorded in the whole-cell configuration of the patch clamp technique as previously described (15). All recordings were performed at room temperature (20 to 22 °C). The pipette solution consisted of (in mmol/liter) 125 Cs-methanosulfonate, 15 TEA-Cl, 1 MgCl2, 10 EGTA, and 5 Hepes, 5 MgATP, 5 phosphocreatine, pH 7.2. Bath solution contained (in mmol/liter) 140 NaCl, 5.4 KCl, 1.2 KH2PO4, 5 Hepes, 5.55 glucose, 1 MgCl2, 1.8 CaCl2, pH 7.4. Once a cell was successfully patched, zero sodium bath solution was introduced into the chamber (mmol/liter) 150 N-methyl-d-glucamine, 2.5 CaCl2, 1 MgCl2, 10 glucose, 10 Hepes, 4-amino-pyridine, pH 7.2. Recordings of isoproterenol response were recorded in zero sodium bath solution containing 300 nm isoproterenol. Activation voltage dependence parameters were obtained by first transforming the peak current/voltage relationship to a conductance transform by fitting the ascending phase (typically Vtest +15 to +40 mV) to a linear regression to obtain a reversal potential. Using G = I/V the conductance as a function of voltage transform was then fitted to a Boltzmann distribution of the form G(V) = Gmax/(1 + exp(V½/k)), where Gmax is the maximal conductance and V½ is the activation midpoint. For steady-state availability curve, we pre-pulsed cells to Vpre ranging from −40 to +10 mV in 5-mV increments and recorded a Vtest to 0 mV. Peak currents were plotted as a function of Vpre, and a Boltzmann distribution was fitted to the resulting curve.
Calcium transients were recorded from ventricular cardiomyocytes loaded with cell permeable fura2-AM (Invitrogen) at 1.0 Hz to determine transient amplitude, upstroke velocity, and rate of decay. Sarcomere dynamics were simultaneously recorded by acquiring an optical signal of visible striations of the cardiomyocyte. All measurements were made following >2 min of conditioning of 1-Hz field stimuli to induce steady-state. Transients were recorded at 1 Hz. All Ca2+ transient/sarcomere dynamic data were analyzed using IonOptix IonWizard 6.3. Background fluorescence (Fbackground) for F380 and F340 were determined from cell-free regions. Data are expressed as F340/380 and were corrected for Fbackground.
Calcium transients were also recorded from ventricular cardiomyocytes loaded with cell permeable fluo4-AM (Invitrogen) at 1.0 Hz to determine the transient rate of decay. All measurements were made following >5 min of conditioning of 1-Hz field stimuli to induce steady-state. Using confocal microscopy, a line scan was taken of an individual cardiomyocyte. Transient decays from line scans were then fitted with a single exponential to determine the rate of decay.
Quantitative RT-PCR
Mice were anesthetized with ketamine and xylazine and hearts were quickly excised, after which the apex of the left ventricle was removed and snap frozen in liquid nitrogen. Frozen tissue was then homogenized, and RNA was isolated using the RNAqueous kit (Life Technologies) and quantified using a Nanodrop (ThermoScientific). cDNA was generated from 500 ng of RNA, which was then amplified via RT-PCR using TaqMan probes from Life Technologies: gapdh (Mm99999915_g1), nppa (Mm01255747_g1), rcan1 (Mm01213406_m1), cacna1c (Mm01188822_m1), cacna1d (Mm01209927_g1), and ctgf (Mm01192933_g1). Threshold values (CT) for nppa, rrad, and rcan1 were normalized by subtraction from gapdh, and WT was then subtracted from RAD−/− (ΔΔCT) and fold-changes were calculated as 2−ΔΔCT.
Histology
Mice were anesthetized with ketamine and xylazine and hearts were perfused with PBS followed by 10% formalin in PBS. Fixed hearts were halved along the short axis with papillary muscle visible, and both basal and apical regions were sectioned at 5-μm sections. Picosirius red staining (Direct red 80, Sirius red, Sigma 365548; icric acid, Sigma P6744; in acetic acid) for 1 h at room temperature was used to assess fibrosis (collagen). Sections were 100% ethanol treated and cleared with xylene. Bright field images were captured with polarized light and analyzed with FIJI (ImageJ2).
Echocardiography
Transthoracic echocardiography was performed using the Visual Sonics 770 imaging system equipped with a 30-MHz probe. Mice underwent transthoracic echocardiography, under light anesthesia (inhaled Isoflurane, 1–2%), with heart rate (350–500 beats per minute) and core temperature (37 °C) continuously monitored. The heart was visualized in 2-dimensional form from modified parasternal long axis, short axis, and apical views. The left ventricular dimensions and calculated left ventricular EF were measured from the short axis M-mode display. All measurements were obtained in triplicate and averaged. The sonographer was blinded to animal genotype, and data analysis was performed with animal genotype blinded.
For the pharmacological stress echocardiogram, a single intraperitoneal injection of isoproterenol (ISO, 30 mg/kg, USP) was given immediately after baseline echocardiography measurements were recorded. Heart rate was monitored, within 5 min of increased heart rate the drug effect was confirmed, and echocardiography measures were repeated.
Radiotelemetry
RADfl/fl-αMHC-CreERT2 mice were chronically instrumented in the left common carotid artery with a radiotelemetry probe (HD-X11, Data Sciences International; St. Paul, MN). Recordings commenced 2 weeks after surgical implantation of probes. Blood pressure was recorded continuously for 1 week prior to tamoxifen administration and then continuously for 2 weeks after tamoxifen.
Statistical analysis
For statistical analyses performed on all cellular observations the mouse is the primary unit of analysis. Cellular mean ± S.E. values are represented in the figures. The within-mouse averages of the cellular observations were used for analysis to perform the analysis on the level of the experimental units in the mice. The factors of mouse type and isoproterenol treatment (2 × 2 factorial design) were analyzed using 2-way ANOVA. In the studies of responses across multiple time points, a repeated-measures ANOVA was performed. Post hoc t tests were performed to compare particular groups of interest. In the analyses involving the difference of means between WT and RADΔ/Δ mice under the assumption of normality of response variable, 2-sample t tests were used, and paired t tests were used before and after isoproterenol treatment comparisons. p < 0.05 was considered statistically significant. All statistical analyses were performed using GraphPad Prism7 (San Diego, CA).
Author contributions
B. M. A., D. A. A., and J. S. conceptualization; B. M. A. and B. M. L. data curation; B. M. A., B. M. L., S. V., M. S., M. C. G., J. L., D. A. A., and J. S. formal analysis; B. M. A., D. A. A., and J. S. supervision; B. M. A., D. A. A., and J. S. funding acquisition; B. M. A., B. M. L., S. V., M. S., N. A., A. S., W. S., M. C. G., J. L., J. E. S., D. A. A., and J. S. investigation; B. M. A., B. M. L., D. A. A., and J. S. writing-original draft; B. M. A., D. A. A., and J. S. project administration; J. S. resources.
Supplementary Material
Acknowledgments
We thank Wendy Katz for histology support provided by an Institutional Development Award (IDeA) from NIGMS National Institutes of Health Grant P20 GM103527. We thank Tanya Seward for surgical support for radiotelemetry in the Physiology Department core facility. The Vevo3100 was supported by The Saha Cardiovascular Research Center.
This work was supported by National Institutes of Health Grants HL131782 (to D. A. A. and J. S.), HL11470 and HL146676 (to J. E. S.), and GM007250 and T32-HL007567 (to J. L.), American Heart Association Grant 17SDG33670578 (to S. V.), and American Heart Association Pre-doctoral Fellowship 19PRE34380909 and NIGMS T32GM118292 (to B. M. A.). The authors declare that they have no conflicts of interest with the contents of this article. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
This article contains Figs. S1–S4.
- LTCC
- L-type Ca2+ channel complex
- ANF
- atrial natriuretic factor
- β-AR
- β-adrenergic receptor
- CTGF
- connective tissue growth factor
- EF
- ejection fraction
- gRAD−/−
- whole body RAD gene deletion
- HFrEF
- heart failure with reduced ejection fraction
- ICa,L
- L-type calcium channel current
- ISO
- isoproterenol
- PLN
- phospholamban
- RAD
- Rad associated with diabetes
- RADfl/fl
- RAD with loxP sites flanking exons 3–4
- RADΔ/Δ
- inducible, myocardial-restricted RAD knockout
- RCAN1
- regulator of calcineurin 1
- SERCA2a
- sarco/endoplasmic reticulum calcium ATPase
- SR
- sarcoplasmic reticulum
- qRT
- quantitative RT
- MHC
- α-myosin heavy chain
- HR
- heart rate
- ANOVA
- analysis of variance
- EF
- ejection fraction
- GAPDH
- glyceraldehyde-3-phosphate dehydrogenase.
References
- 1. Maack C., Eschenhagen T., Hamdani N., Heinzel F. R., Lyon A. R., Manstein D. J., Metzger J., Papp Z., Tocchetti C. G., Yilmaz M. B., Anker S. D., Balligand J. L., Bauersachs J., Brutsaert D., Carrier L., et al. (2018) Treatments targeting inotropy. Eur. Heart J. 10.1093/eurheartj/ehy600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Mebazaa A., Parissis J., Porcher R., Gayat E., Nikolaou M., Boas F. V., Delgado J. F., and Follath F. (2011) Short-term survival by treatment among patients hospitalized with acute heart failure: the global ALARM-HF registry using propensity scoring methods. Intensive Care Med. 37, 290–301 10.1007/s00134-010-2073-4 [DOI] [PubMed] [Google Scholar]
- 3. Tacon C. L., McCaffrey J., and Delaney A. (2012) Dobutamine for patients with severe heart failure: a systematic review and meta-analysis of randomised controlled trials. Intensive Care Med. 38, 359–367 10.1007/s00134-011-2435-6 [DOI] [PubMed] [Google Scholar]
- 4. Bristow M. R. (2011) Treatment of chronic heart failure with beta-adrenergic receptor antagonists: a convergence of receptor pharmacology and clinical cardiology. Circ. Res. 109, 1176–1194 10.1161/CIRCRESAHA.111.245092 [DOI] [PubMed] [Google Scholar]
- 5. Lohse M. J., Engelhardt S., and Eschenhagen T. (2003) What is the role of β-adrenergic signaling in heart failure? Circ. Res. 93, 896–906 10.1161/01.RES.0000102042.83024.CA [DOI] [PubMed] [Google Scholar]
- 6. Satin J. (2017) Regulation of Cardiac Calcium Channels. in Cardiac Electrophysiology: From Cell to Bedside (Zipes D. P., Jalife J., and Stevenson W., eds) 7th Ed., pp. 1424, Elsevier, New York [Google Scholar]
- 7. Béguin P., Nagashima K., Gonoi T., Shibasaki T., Takahashi K., Kashima Y., Ozaki N., Geering K., Iwanaga T., and Seino S. (2001) Regulation of Ca2+ channel expression at the cell surface by the small G-protein kir/Gem. Nature 411, 701–706 10.1038/35079621 [DOI] [PubMed] [Google Scholar]
- 8. Finlin B. S., Crump S. M., Satin J., and Andres D. A. (2003) Regulation of voltage-gated calcium channel activity by the Rem and Rad GTPases. Proc. Natl. Acad. Sci. U.S.A. 100, 14469–14474 10.1073/pnas.2437756100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Finlin B. S., Correll R. N., Pang C., Crump S. M., Satin J., and Andres D. A. (2006) Analysis of the complex between Ca2+ channel β-subunit and the Rem GTPase. J. Biol. Chem. 281, 23557–23566 10.1074/jbc.M604867200 [DOI] [PubMed] [Google Scholar]
- 10. Puckerin A. A., Chang D. D., Shuja Z., Choudhury P., Scholz J., and Colecraft H. M. (2018) Engineering selectivity into RGK GTPase inhibition of voltage-dependent calcium channels. Proc. Natl. Acad. Sci. U.S.A. 115, 12051–12056 10.1073/pnas.1811024115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gunton J. E., Sisavanh M., Stokes R. A., Satin J., Satin L. S., Zhang M., Liu S. M., Cai W., Cheng K., Cooney G. J., Laybutt D. R., So T., Molero J. C., Grey S. T., Andres D. A., Rolph M. S., and Mackay C. R. (2012) Mice deficient in GEM GTPase show abnormal glucose homeostasis due to defects in beta-cell calcium handling. PLoS ONE 7, e39462 10.1371/journal.pone.0039462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Puhl H. L. 3rd, Lu V. B., Won Y.-J., Sasson Y., Hirsch J. A., Ono F., and Ikeda S. R. (2014) Ancient origins of RGK protein function: modulation of voltage-gated calcium channels preceded the protostome and deuterostome split. PloS ONE 9, e100694–e100694 10.1371/journal.pone.0100694 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Reynet C., and Kahn C. R. (1993) Rad: a member of the Ras family overexpressed in muscle of type II diabetic humans. Science 262, 1441–1444 10.1126/science.8248782 [DOI] [PubMed] [Google Scholar]
- 14. Correll R. N., Pang C., Niedowicz D. M., Finlin B. S., and Andres D. A. (2008) The RGK family of GTP-binding proteins: regulators of voltage-dependent calcium channels and cytoskeleton remodeling. Cell Signal. 20, 292–300 10.1016/j.cellsig.2007.10.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Magyar J., Kiper C. E., Sievert G., Cai W., Shi G. X., Crump S. M., Li L., Niederer S. A., Smith N. P., Andres D. A., and Satin J. (2012) Rem-GTPase regulates cardiac myocyte L-type calcium current. Channels (Austin) 6, 166–173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Yang T., and Colecraft H. M. (2013) Regulation of voltage-dependent calcium channels by RGK proteins. Biochim. Biophys. Acta 1828, 1644–1654 10.1016/j.bbamem.2012.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Levitan B. M., Manning J. R., Withers C. N., Smith J. D., Shaw R. M., Andres D. A., Sorrell V. L., and Satin J. (2016) Rad-deletion phenocopies tonic sympathetic stimulation of the heart. J. Cardiovasc. Transl. Res. 9, 432–444 10.1007/s12265-016-9716-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Manning J. R., Yin G., Kaminski C. N., Magyar J., Feng H. Z., Penn J., Sievert G., Thompson K., Jin J. P., Andres D. A., and Satin J. (2013) Rad GTPase deletion increases L-type calcium channel current leading to increased cardiac contraction. J. Am. Heart Assoc. 2, e000459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Chang L., Zhang J., Tseng Y. H., Xie C. Q., Ilany J., Brüning J. C., Sun Z., Zhu X., Cui T., Youker K. A., Yang Q., Day S. M., Kahn C. R., and Chen Y. E. (2007) Rad GTPase deficiency leads to cardiac hypertrophy. Circulation 116, 2976–2983 10.1161/CIRCULATIONAHA.107.707257 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wang G., Zhu X., Xie W., Han P., Li K., Sun Z., Wang Y., Chen C., Song R., Cao C., Zhang J., Wu C., Liu J., and Cheng H. (2010) Rad as a novel regulator of excitation-contraction coupling and β-adrenergic signaling in heart. Circ. Res. 106, 317–327 10.1161/CIRCRESAHA.109.208272 [DOI] [PubMed] [Google Scholar]
- 21. Manning J. R., Withers C. N., Levitan B., Smith J. D., Andres D. A., and Satin J. (2015) Loss of Rad-GTPase produces a novel adaptive cardiac phenotype resistant to systolic decline with aging. Am. J. Physiol. Heart Circ. Physiol. 309, H1336–H1345 10.1152/ajpheart.00389.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Zhang J., Chang L., Chen C., Zhang M., Luo Y., Hamblin M., Villacorta L., Xiong J.-W., Chen Y. E., Zhang J., and Zhu X. (2011) Rad GTPase inhibits cardiac fibrosis through connective tissue growth factor. Cardiovasc. Res. 91, 90–98 10.1093/cvr/cvr068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Muth J. N., Bodi I., Lewis W., Varadi G., and Schwartz A. (2001) A Ca2+-dependent transgenic model of cardiac hypertrophy: a role for protein kinase Cα. Circulation 103, 140–147 10.1161/01.CIR.103.1.140 [DOI] [PubMed] [Google Scholar]
- 24. Chen X., Nakayama H., Zhang X., Ai X., Harris D. M., Tang M., Zhang H., Szeto C., Stockbower K., Berretta R. M., Eckhart A. D., Koch W. J., Molkentin J. D., and Houser S. R. (2011) Calcium influx through Cav1.2 is a proximal signal for pathological cardiomyocyte hypertrophy. J. Mol. Cell Cardiol. 50, 460–470 10.1016/j.yjmcc.2010.11.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Fu M., Zhang J., Tseng Y.-H., Cui T., Zhu X., Xiao Y., Mou Y., De Leon H., Chang M. M., Hamamori Y., Kahn C. R., and Chen Y. E. (2005) Rad GTPase attenuates vascular lesion formation by inhibition of vascular smooth muscle cell migration. Circulation 111, 1071–1077 10.1161/01.CIR.0000156439.55349.AD [DOI] [PubMed] [Google Scholar]
- 26. Withers C. N., Brown D. M., Byiringiro I., Allen M. R., Condon K. W., Satin J., and Andres D. A. (2017) Rad GTPase is essential for the regulation of bone density and bone marrow adipose tissue in mice. Bone 103, 270–280 10.1016/j.bone.2017.07.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Correll R. N., Pang C., Finlin B. S., Dailey A. M., Satin J., and Andres D. A. (2007) Plasma membrane targeting is essential for Rem-mediated Ca2+ channel inhibition. J. Biol. Chem. 282, 28431–28440 10.1074/jbc.M706176200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Sohal D. S., Nghiem M., Crackower M. A., Witt S. A., Kimball T. R., Tymitz K. M., Penninger J. M., and Molkentin J. D. (2001) Temporally regulated and tissue-specific gene manipulations in the adult and embryonic heart using a tamoxifen-inducible Cre protein. Circ. Res. 89, 20–25 10.1161/hh1301.092687 [DOI] [PubMed] [Google Scholar]
- 29. Wang Y., Li G., Mao F., Li X., Liu Q., Chen L., Lv L., Wang X., Wu J., Dai W., Wang G., Zhao E., Tang K. F., and Sun Z. S. (2014) Ras-induced epigenetic inactivation of the RRAD (Ras-related associated with diabetes) gene promotes glucose uptake in a human ovarian cancer model. J. Biol. Chem. 289, 14225–14238 10.1074/jbc.M113.527671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kobirumaki-Shimozawa F., Oyama K., Shimozawa T., Mizuno A., Ohki T., Terui T., Minamisawa S., Ishiwata S., and Fukuda N. (2016) Nano-imaging of the beating mouse heart in vivo: importance of sarcomere dynamics, as opposed to sarcomere length per se, in the regulation of cardiac function. J. Gen. Physiol. 147, 53–62 10.1085/jgp.201511484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Davis J., Davis L. C., Correll R. N., Makarewich C. A., Schwanekamp J. A., Moussavi-Harami F., Wang D., York A. J., Wu H., Houser S. R., Seidman C. E., Seidman J. G., Regnier M., Metzger J. M., Wu J. C., and Molkentin J. D. (2016) A tension-based model distinguishes hypertrophic versus dilated cardiomyopathy. Cell 165, 1147–1159 10.1016/j.cell.2016.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Gresham K. S., Mamidi R., Li J., Kwak H., and Stelzer J. E. (2017) Sarcomeric protein modification during adrenergic stress enhances cross-bridge kinetics and cardiac output. J. Appl. Physiol. 122, 520–530 10.1152/japplphysiol.00306.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Gresham K. S., and Stelzer J. E. (2016) The contributions of cardiac myosin binding protein C and troponin I phosphorylation to β-adrenergic enhancement of in vivo cardiac function. J. Physiol. 594, 669–686 10.1113/JP270959 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Bertero E., and Maack C. (2018) Metabolic remodelling in heart failure. Nat. Rev. Cardiol. 15, 780–781 10.1038/s41569-018-0116-7 [DOI] [PubMed] [Google Scholar]
- 35. Lexow J., Poggioli T., Sarathchandra P., Santini M. P., and Rosenthal N. (2013) Cardiac fibrosis in mice expressing an inducible myocardial-specific Cre driver. Dis. Model Mech. 6, 1470–1476 10.1242/dmm.010470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Makarewich C. A., Correll R. N., Gao H., Zhang H., Yang B., Berretta R. M., Rizzo V., Molkentin J. D., and Houser S. R. (2012) A caveolae-targeted L-type Ca2+ channel antagonist inhibits hypertrophic signaling without reducing cardiac contractility. Circ. Res. 110, 669–674 10.1161/CIRCRESAHA.111.264028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Gao H., Wang F., Wang W., Makarewich C. A., Zhang H., Kubo H., Berretta R. M., Barr L. A., Molkentin J. D., and Houser S. R. (2012) Ca2+ influx through L-type Ca2+ channels and transient receptor potential channels activates pathological hypertrophy signaling. J. Mol. Cell Cardiol. 53, 657–667 10.1016/j.yjmcc.2012.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Muth J. N., Yamaguchi H., Mikala G., Grupp I. L., Lewis W., Cheng H., Song L. S., Lakatta E. G., Varadi G., and Schwartz A. (1999) Cardiac-specific overexpression of the α1 subunit of the L-type voltage-dependent Ca2+ channel in transgenic mice. Loss of isoproterenol-induced contraction. J. Biol. Chem. 274, 21503–21506 10.1074/jbc.274.31.21503 [DOI] [PubMed] [Google Scholar]
- 39. Sipido K. R., Callewaert G., and Carmeliet E. (1995) Inhibition and rapid recovery of Ca2+ current during Ca2+ release from sarcoplasmic reticulum in guinea pig ventricular myocytes. Circ. Res. 76, 102–109 10.1161/01.RES.76.1.102 [DOI] [PubMed] [Google Scholar]
- 40. Correll R. N., Makarewich C. A., Zhang H., Zhang C., Sargent M. A., York A. J., Berretta R. M., Chen X., Houser S. R., and Molkentin J. D. (2017) Caveolae-localized L-type Ca2+ channels do not contribute to function or hypertrophic signalling in the mouse heart. Cardiovasc. Res. 113, 749–759 10.1093/cvr/cvx046 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Bupha-Intr T., Laosiripisan J., and Wattanapermpool J. (2009) Moderate intensity of regular exercise improves cardiac SR Ca2+ uptake activity in ovariectomized rats. J. Appl. Physiol. 107, 1105–1112 10.1152/japplphysiol.00407.2009 [DOI] [PubMed] [Google Scholar]
- 42. Freeman K., Lerman I., Kranias E. G., Bohlmeyer T., Bristow M. R., Lefkowitz R. J., Iaccarino G., Koch W. J., and Leinwand L. A. (2001) Alterations in cardiac adrenergic signaling and calcium cycling differentially affect the progression of cardiomyopathy. J. Clin. Invest. 107, 967–974 10.1172/JCI12083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Jaski B. E., Jessup M. L., Mancini D. M., Cappola T. P., Pauly D. F., Greenberg B., Borow K., Dittrich H., Zsebo K. M., Hajjar R. J., and Calcium Up-Regulation by Percutaneous Administration of Gene Therapy In Cardiac Disease Trial Investigators (2009) Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID Trial), a first-in-human phase 1/2 clinical trial. J. Card. Fail. 15, 171–181 10.1016/j.cardfail.2009.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Jessup M., Greenberg B., Mancini D., Cappola T., Pauly D. F., Jaski B., Yaroshinsky A., Zsebo K. M., Dittrich H., Hajjar R. J., and Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease Investigators (2011) Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID): a phase 2 trial of intracoronary gene therapy of sarcoplasmic reticulum Ca2+-ATPase in patients with advanced heart failure. Circulation 124, 304–313 10.1161/CIRCULATIONAHA.111.022889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Wang J.-N., Shi N., Xie W.-B., Guo X., and Chen S.-Y. (2011) Response gene to complement 32 promotes vascular lesion formation through stimulation of smooth muscle cell proliferation and migration. Arterioscler. Thromb. Vasc. Biol. 31, e19–e26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Mahajan A., Sato D., Shiferaw Y., Baher A., Xie L. H., Peralta R., Olcese R., Garfinkel A., Qu Z., and Weiss J. N. (2008) Modifying L-type calcium current kinetics: consequences for cardiac excitation and arrhythmia dynamics. Biophys. J. 94, 411–423 10.1529/biophysj.106.98590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Goldhaber J. I., and Hamilton M. A. (2010) Role of inotropic agents in the treatment of heart failure. Circulation 121, 1655–1660 10.1161/CIRCULATIONAHA.109.899294 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Raake P. W., Zhang X., Vinge L. E., Brinks H., Gao E., Jaleel N., Li Y., Tang M., Most P., Dorn G. W. 2nd, Houser S. R., Katus H. A., Chen X., and Koch W. J. (2012) Cardiac G-protein–coupled receptor kinase 2 ablation induces a novel Ca2+ handling phenotype resistant to adverse alterations and remodeling after myocardial infarction. Circulation 125, 2108–2118 10.1161/CIRCULATIONAHA.111.044255 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Schmid E., Neef S., Berlin C., Tomasovic A., Kahlert K., Nordbeck P., Deiss K., Denzinger S., Herrmann S., Wettwer E., Weidendorfer M., Becker D., Schäfer F., Wagner N., Ergün S., et al. (2015) Cardiac RKIP induces a beneficial [beta]-adrenoceptor-dependent positive inotropy. Nat. Med. 21, 1298–1306 10.1038/nm.3972 [DOI] [PubMed] [Google Scholar]
- 50. Manning J. R., Chelvarajan L., Levitan B. M., Withers C. N., Nagareddy P. R., Haggerty C. M., Fornwalt B. K., Gao E., Tripathi H., Abdel-Latif A., Andres D. A., and Satin J. (2018) Rad GTPase deletion attenuates post-ischemic cardiac dysfunction and remodeling. JACC Basic Transl. Sci. 3, 83–96 10.1016/j.jacbts.2017.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.