Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2020 Jun 8;117(25):14522–14531. doi: 10.1073/pnas.2001270117

GPCR-dependent biasing of GIRK channel signaling dynamics by RGS6 in mouse sinoatrial nodal cells

Allison Anderson a, Ikuo Masuho b, Ezequiel Marron Fernandez de Velasco a, Atsushi Nakano c, Lutz Birnbaumer d,e, Kirill A Martemyanov b, Kevin Wickman a,1
PMCID: PMC7322085  PMID: 32513692

Significance

Mechanisms promoting the compartmentalization of G-protein signaling are poorly understood in native cells. This study examined muscarinic (M2R) and adenosine (A1R) receptor signaling to a common effector—the G protein-gated inwardly rectifying K+ (GIRK) channel—in mouse sino-atrial nodal (SAN) cells. Regulator of G protein Signaling 6 (RGS6) exerts a GPCR-dependent influence on the efficacy, kinetics, and sensitivity of GIRK-dependent signaling in SAN cells, suppressing M2R-GIRK signaling kinetics and sensitivity and A1R-GIRK signaling amplitude. The distinctions can be explained by the differential utilization of G protein isoforms by M2R and A1R and a Gαo substrate preference of RGS6. Thus, GPCR-G protein signaling and RGS-G protein substrate biases shape GIRK-dependent signaling dynamics in the mouse heart.

Keywords: muscarinic, adenosine, Kir3, G protein, heart rate

Abstract

How G protein-coupled receptors (GPCRs) evoke specific biological outcomes while utilizing a limited array of G proteins and effectors is poorly understood, particularly in native cell systems. Here, we examined signaling evoked by muscarinic (M2R) and adenosine (A1R) receptor activation in the mouse sinoatrial node (SAN), the cardiac pacemaker. M2R and A1R activate a shared pool of cardiac G protein-gated inwardly rectifying K+ (GIRK) channels in SAN cells from adult mice, but A1R-GIRK responses are smaller and slower than M2R-GIRK responses. Recordings from mice lacking Regulator of G protein Signaling 6 (RGS6) revealed that RGS6 exerts a GPCR-dependent influence on GIRK-dependent signaling in SAN cells, suppressing M2R-GIRK coupling efficiency and kinetics and A1R-GIRK signaling amplitude. Fast kinetic bioluminescence resonance energy transfer assays in transfected HEK cells showed that RGS6 prefers Gαo over Gαi as a substrate for its catalytic activity and that M2R signals preferentially via Gαo, while A1R does not discriminate between inhibitory G protein isoforms. The impact of atrial/SAN-selective ablation of Gαo or Gαi2 was consistent with these findings. Gαi2 ablation had minimal impact on M2R-GIRK and A1R-GIRK signaling in SAN cells. In contrast, Gαo ablation decreased the amplitude and slowed the kinetics of M2R-GIRK responses, while enhancing the sensitivity and prolonging the deactivation rate of A1R-GIRK signaling. Collectively, our data show that differences in GPCR-G protein coupling preferences, and the Gαo substrate preference of RGS6, shape A1R- and M2R-GIRK signaling dynamics in mouse SAN cells.


G protein-coupled receptors (GPCRs) constitute the largest family of cell-surface receptors in the mammalian genome, and they mediate the influence of a diverse array of ligands on cell signaling, organ physiology, and behavior (1). GPCRs evoke biological responses by coupling to a limited array of heterotrimeric G proteins, which then regulate the activity of a relatively small pool of effectors. Mechanisms underlying the compartmentalization of G protein-dependent signaling pathways in native cell types are not well understood.

Regulation of heart rate (HR) offers an opportunity to investigate how different GPCRs engage intracellular signaling networks to shape a critical physiological response. Activation of the parasympathetic branch of the autonomic nervous system elicits bradycardia (2), an effect mediated by the acetylcholine (ACh)-induced activation of M2 muscarinic receptors (M2R) on sinoatrial nodal (SAN) cells (3). M2R activation stimulates inhibitory G proteins, which then modulate the activity of downstream effectors, including the G protein-gated inwardly rectifying K+ (GIRK) channel (46). The cardiac GIRK channel is a heterotetramer consisting of GIRK1 and GIRK4 subunits (79). Studies involving Girk1−/− and Girk4−/− mice have shown that activation of cardiac GIRK channels mediates most of the parasympathetic impact on HR, as well as atrial and atrio-ventricular (AV) impulse conduction (1013). Cardiac GIRK channels are also activated by adenosine (Ado), an autacoid generated under ischemic conditions that activates the A1 adenosine receptor (A1R), resulting in GIRK-dependent bradycardia (10, 1416). The ability of Ado to suppress HR and AV impulse conduction is exploited clinically to treat and diagnose supraventricular tachycardias (14).

GPCR-dependent signaling dynamics are influenced by Regulator of G protein Signaling (RGS) proteins (17). RGS proteins possess GTPase-activating protein (GAP) catalytic activity that accelerates the intrinsic GTP hydrolysis rate of G protein alpha (Gα) subunits. Previous work from our group and others has identified RGS6 as a critical negative regulator of M2R-GIRK signaling in mouse SAN cells and atrial myocytes (1820). RGS6 is a member of the R7 RGS subfamily of RGS proteins, members of which form obligate dimers with the atypical Gβ subunit Gβ5 (21). Genetic ablation of Rgs6 in mice increased GIRK channel sensitivity to muscarinic receptor activation, prolonged the deactivation rate of currents elicited by the nonselective cholinergic agonist carbachol (CCh), and enhanced CCh-induced bradycardia (1820). The extent to which RGS6 regulates A1R-GIRK signaling in the heart is currently unknown.

Here, we probed the functional compartmentalization of GPCR-dependent signaling by investigating signaling engaged by M2R and A1R activation in SAN cells. Despite converging on a common effector, M2R-GIRK and A1R-GIRK signaling pathways display notable differences in amplitude and kinetics. Using an array of knockout mouse lines, as well as electrophysiological, molecular, and optical imaging approaches, we found that the differences in M2R-GIRK and A1R-GIRK signaling dynamics in mouse SAN cells can be explained by GPCR-specific coupling biases for inhibitory G protein isoforms and the Gαo substrate specificity of RGS6.

Results

A1R-GIRK and M2R-GIRK Signaling Distinctions in Mouse SAN Cells.

We began by measuring whole-cell currents evoked by a saturating concentration of either Ado (10 μM) or CCh (10 μM) in isolated SAN cells from C57BL/6J (wild-type) mice (Fig. 1). Both agonists reliably evoked currents in SAN cells from wild-type mice, but not in SAN cells from congenic Girk4−/− mice (Fig. 1 A and B), indicating that the evoked responses were mediated by GIRK channel activation. Currents elicited by Ado in wild-type SAN cells were smaller than CCh-induced currents (Fig. 1C) and did not exhibit acute desensitization (Fig. 1D). In addition, activation and deactivation rates of Ado-induced GIRK currents were slower than those of CCh-induced responses (Fig. 1 E and F).

Fig. 1.

Fig. 1.

Ado- and CCh-induced GIRK currents in SAN cells. (A and B) Whole-cell currents (Vhold = −70 mV) evoked by Ado (10 μM; A) or CCh (10 μM; B) in SAN cells from wild-type (Left, black) and Rgs6−/− (Right, red) mice; these currents were not observed in SAN cells from Girk4−/− mice (Bottom, blue). (Scale bars: 5 s/500 pA.) (C) Peak current density of responses elicited by Ado (Left) and CCh (Right) in SAN cells from wild-type (black) and Rgs6−/− (red) mice. There was a significant interaction between genotype and agonist (F1,49 = 14.9, P < 0.001; two-way ANOVA); group sizes ranged from 10 to 20 cells (5–7 mice). **P < 0.01 wild-type vs. Rgs6−/− (within agonist); ####P < 0.0001 Ado vs. CCh (wild-type). (D) Acute desensitization of responses elicited by Ado (Left) and CCh (Right) in SAN cells from wild-type (black) and Rgs6−/− (red) mice. Statistical analysis revealed a main effect of agonist (F1,49 = 30.4, P < 0.0001; two-way ANOVA), but no main effect of genotype (F1,49 = 0.13, P = 0.72; two-way ANOVA) or genotype x agonist interaction (F1,49 = 1.6, P = 0.21; two-way ANOVA); group sizes ranged from 9 to 19 cells (5–7 mice). (E) Activation rates of responses elicited by Ado (Left) and CCh (Right) in SAN cells from wild-type (black) and Rgs6−/− (red) mice. Statistical analysis revealed a genotype x agonist interaction (F1,45 = 6.8, P < 0.05; two-way ANOVA); group sizes ranged from 9 to 17 cells (5–7 mice). **P < 0.01 wild-type vs. Rgs6−/− (within agonist); ####P < 0.0001 Ado vs. CCh (wild type). (F) Deactivation rates of responses elicited by Ado (Left) and CCh (Right) in SAN cells from wild-type (black) and Rgs6−/− (red) mice. Statistical analysis revealed a genotype x agonist interaction (F1,48 = 8.0, P < 0.01; two-way ANOVA); group sizes ranged from 12 to 19 cells (5–7 mice). ***P < 0.001 wild-type vs. Rgs6−/− (within agonist); ###P < 0.001 Ado vs. CCh (wild type).

RGS6 Exerts a GPCR-Dependent Influence on GIRK-Dependent Signaling.

In parallel, we explored the impact of Rgs6 ablation on Ado- and CCh-induced currents in SAN cells. While Ado-induced currents were significantly larger in SAN cells from Rgs6−/− mice relative to wild-type controls (Fig. 1 A and C), there was no impact of Rgs6 ablation on CCh-induced current amplitudes (Fig. 1 B and C). Rgs6 ablation also exerted a GPCR-dependent impact on response kinetics. Loss of RGS6 correlated with prolonged activation rate of Ado-induced currents (Fig. 1E), but there was no impact on deactivation rate (Fig. 1F). In contrast, and consistent with published reports (18, 19), there was no impact of Rgs6 ablation on the activation rate of CCh-induced currents (Fig. 1F), but deactivation rate was prolonged (Fig. 1F). Acute desensitization of CCh- and Ado-induced currents was not impacted by Rgs6 ablation (Fig. 1D). Thus, RGS6 normally suppresses the amplitude and accelerates the activation rate of A1R-GIRK signaling in mouse SAN cells, while accelerating the deactivation rate of M2R-GIRK signaling.

Ado-induced currents in SAN cells from Rgs6−/− mice were completely reversed by the GIRK channel inhibitor rTertiapin-Q (TTQ, 300 nM; SI Appendix, Fig. S1) (22, 23), indicating that the enhanced Ado-induced responses in these cells involves an increase in GIRK-dependent signaling. There was no difference in GIRK1, GIRK4, or A1R mRNA levels in atrial tissue samples containing the SAN region from wild-type and Rgs6−/− mice (SI Appendix, Fig. S2). Additionally, GIRK channel currents evoked by ML297, a direct and selective activator of GIRK1-containing GIRK channels (24, 25), were modestly but significantly smaller in SAN cells from Rgs6−/− mice (SI Appendix, Fig. S3). Thus, the increase in Ado-induced GIRK currents in SAN cells from Rgs6−/− mice does not appear to be linked to an increase in the expression of A1R or GIRK channels.

To address the impact of Rgs6 ablation on the sensitivity of M2R-GIRK and A1R-GIRK signaling pathways, we measured GIRK currents evoked by increasing concentrations of Ado and CCh in SAN cells from wild-type and Rgs6−/− mice. While there was no impact of Rgs6 ablation on the sensitivity of GIRK channels to Ado (Fig. 2 AC), GIRK channels in SAN cells from Rgs6−/− mice were more sensitive to CCh than GIRK channels in SAN cells from wild-type controls (Fig. 2 DF) (19). Thus, RGS6 decreases GIRK channel sensitivity to CCh, but not Ado, in SAN cells.

Fig. 2.

Fig. 2.

Impact of Rgs6 ablation on GIRK channel sensitivity to CCh and Ado. (A) Concentration-response experiments for Ado in SAN cells from wild-type (Scale bars: Top, 10 s/200 pA.) and Rgs6−/− (Scale bars: Bottom, 10 s/500 pA.) mice. (B and C) Summary of concentration-response experiments for Ado-induced currents in SAN cells from wild-type and Rgs6−/− mice. There was no difference in EC50 values for Ado-induced currents in SAN cells from wild-type (n = 24 cells/7 mice) and Rgs6−/− (n = 13 cells/5 mice) mice (t35 = 1.8, P = 0.08; unpaired t test). (D) Concentration-response experiments for CCh-induced currents in SAN cells from wild-type (Scale bars: Top, 10 s/200 pA.) and Rgs6−/− (Scale bars: Bottom, 10 s/500 pA.) mice. (E and F) Summary of concentration-response experiments of CCh-induced currents in SAN cells from wild-type and Rgs6−/− mice. The EC50 values for CCh-induced currents in SAN cells from Rgs6−/− (n = 12 cells/4 mice) mice were lower than the EC50 values measured in wild-type counterparts (n = 10 cells/4 mice) (t20 = 4.8, ***P < 0.001; unpaired t test).

RGS6 Impacts GIRK-Dependent Bradycardia in a GPCR-Dependent Manner.

We also evaluated the impact of Rgs6 ablation on the bradycardic effect of A1R and M2R activation using an isolated heart model (Fig. 3). Both the stable A1R agonist N6-cyclopentyl-adenosine (CPA) (Fig. 3 A and B) and CCh (Fig. 3 C and D) evoked dose-dependent decreases in HR in wild-type hearts. The bradycardic effect of CPA and CCh was strongly diminished in hearts from Girk4−/− mice (Fig. 3 B and D), suggesting that M2R- and A1R-induced bradycardia is largely dependent on GIRK channel activation. The magnitude of CPA-induced bradycardia was larger in Rgs6−/− as compared to wild-type hearts (Fig. 3B), but there was no detectable shift in CPA potency. While CCh-induced bradycardia was also more pronounced in Rgs6−/− hearts (18, 19, 26), there was a clear leftward shift in the dose–response curve (Fig. 3D). Thus, the results from the isolated heart model mirror the results obtained from isolated SAN cells, indicating that RGS6 exerts a prominent negative influence on A1R-GIRK signaling amplitude and M2R-GIRK signaling sensitivity.

Fig. 3.

Fig. 3.

CPA- and CCh-induced bradycardia in isolated hearts from wild-type and Rgs6−/− mice. (A) Segments of ECG traces from isolated wild-type (Top) and Rgs6−/− (Bottom) hearts perfused with the A1R-selective agonist CPA (30 nM). (Scale bar: 5 s.) (B) Percentage decrease in HR (relative to baseline) following perfusion of increasing concentrations of CPA in hearts from wild-type (n = 6), Rgs6−/− (n = 7), and Girk4−/− (n = 4) mice; there was a genotype x CPA concentration interaction for wild-type and Rgs6−/− hearts (F4,44 = 2.81; P < 0.05; two-way ANOVA with repeated measures). **P < 0.01 wild type vs. Rgs6−/−. (C) Segments of ECG traces of isolated wild-type (Top) and Rgs6−/− (Bottom) hearts perfused with CCh (1 μM). (Scale bar: 5 s.) (D) Percentage decrease in HR (relative to baseline) following perfusion of increasing concentrations of CCh in hearts from wild-type (n = 3), Rgs6−/− (n = 5), and Girk4−/− (n = 6) mice; there was a genotype x CCh concentration interaction for wild-type and Rgs6−/− hearts (F3,18 = 5.1, P < 0.05; two-way ANOVA with repeated measures). *P < 0.05 and ***0.001, respectively, wild type vs. Rgs6−/−.

RGS6 Limits the Access of A1R to GIRK Channels.

The differential impact of Rgs6 ablation on M2R-GIRK and A1R-GIRK signaling in mouse SAN cells could reflect the coupling of the GPCRs to separate pools of GIRK channels. To test this possibility, we examined whether Ado-induced currents were occluded by a maximal CCh-induced GIRK channel response. In both wild-type and Rgs6−/− SAN cells, Ado failed to evoke an additive current during application of a saturating concentration of CCh (Fig. 4 A and B). When the order of agonist application was reversed, CCh evoked an additive current during the Ado-induced response (Fig. 4C). The size of the CCh-induced additive response was substantially smaller, however, in SAN cells from Rgs6−/− mice (Fig. 4D). Indeed, the Ado-induced GIRK current occluded nearly all of the CCh-induced response. Collectively, these findings show that M2R and A1R couple to a shared pool of GIRK channels in SAN cells and that RGS6 effectively limits the access of A1R to GIRK channels in these cells.

Fig. 4.

Fig. 4.

Occlusion of CCh- and Ado-induced GIRK currents. (A) Occlusion experiments showing whole-cell currents elicited by a maximal concentration of CCh (10 μM), followed by Ado application (10 μM) in SAN cells from wild-type (Left) and Rgs6−/− (Right) mice. (Scale bars: 10 s/500 pA.) (B) There was no difference in the Ado-induced additive response in SAN cells isolated from wild-type (n = 9 cells/4 mice) and Rgs6−/− (n = 6 cells/5 mice) mice (t13 = 0.11, P = 0.91; unpaired t test). (C) Occlusion experiments showing whole-cell currents elicited by a maximal concentration of Ado (10 μM), followed by CCh application (10 μM) in SAN cells from wild-type (Left) and Rgs6−/− (Right) mice. (Scale bars: 10 s/500 pA.) (D) There was a significant difference in the CCh-induced additive response in SAN cells from wild-type (n = 7 cells/4 mice) and Rgs6−/− (n = 6 cells/4 mice) mice (t11 = 6.0; ****P < 0.0001; unpaired t test).

M2R and A1R Display Overlapping but Distinct G Protein Coupling Preferences.

We next investigated whether the utilization of different Gα isoforms by M2R and A1R could explain the differences in GIRK channel-dependent signaling and modulation by RGS6. Treatment with pertussis toxin abolished CCh- and Ado-induced currents in wild-type SAN cells (SI Appendix, Fig. S4), indicating that M2R– and A1R-GIRK signaling pathways are mediated by inhibitory (Gi/o) G proteins. To probe the inhibitory G protein coupling preferences for M2R and A1R, we employed a Bioluminescence Resonance Energy Transfer (BRET) assay to measure G protein activation in response to GPCR stimulation in living cells (Fig. 5A) (27).

Fig. 5.

Fig. 5.

G protein coupling preferences of M2R and A1R. (A) Schematic representation of the BRET assay for real-time optical imaging of G protein activity. Agonist-induced activation of a GPCR leads to the dissociation of Gα-GTP and Venus-Gβγ subunits. The released Venus-Gβγ then interacts with the Gβγ effector mimetic masGRK3ct-Nluc-HA to produce the BRET signal. (B) Representative real-time monitoring of G protein activation by M2R (Top) or A1R (Bottom). HEK293T/17 cells were transfected with the BRET sensor pair (A) and GPCR, together with either GαoA (black) or Gαi1 (green). Acetylcholine (100 μM) or Ado (100 μM) was applied at the 5-s time point, and the BRET signal was followed across time. (C and D) G protein coupling summary diagrams for M2R and A1R. Maximum amplitudes (red) and activation rate constants (blue) for 15 different G proteins were normalized to the largest value and plotted in the wheel diagrams. Line thickness represents the SEM of three technical replicates performed independently.

We quantified maximal BRET response amplitudes and G protein activation rates for M2R and A1R in the presence of specific G protein isoforms (Fig. 5B). Analysis of BRET amplitudes showed that both M2R and A1R can activate all members of the Gi/o subfamily, as well as G15 (Fig. 5C). M2R displayed faster activation rates with G⍺o relative to G⍺i isoforms, however, while A1R exhibited similar activation rates for G⍺o and G⍺i isoforms (Fig. 5D). Since G protein activation rates closely reflect the catalytic activity of GPCRs (28), whereas amplitudes in the BRET assay could be influenced by intrinsic differences in the extent of heterotrimer dissociation (29), we relied on activation rate as a proxy for G⍺ coupling preferences for M2R and A1R. Accordingly, M2R exhibits a coupling preference for G⍺o over G⍺i, whereas A1R does not discriminate between the inhibitory G protein isoforms.

o Is the Preferred Substrate for RGS6 GAP Activity.

Given the distinct G protein isoform coupling preferences displayed by M2R and A1R, and the differential impact of RGS6 on G protein-mediated regulation of GIRK channels, we next tested whether the RGS6 exhibits a G protein isoform preference for its catalytic activity. The G protein deactivation rates of all Gαi/o members were measured in the presence of RGS6/Gβ5 using a modified version of the BRET assay (Fig. 6A). The D2 dopamine receptor (D2R), dopamine, and haloperidol were chosen for this study due to previous work showing that D2R can efficiently activate all Gαi/o G proteins and that GTPase activity is the rate-limiting step in response deactivation for this combination of receptor and ligands (28). Consistent with results from a study involving purified RGS6/Gβ5 and G proteins (30), RGS6/Gβ5 strongly accelerated the deactivation of GαoA/B, while showing only weak activity toward Gαi1-3 isoforms and no activity toward Gαz (Fig. 6 B and C). Thus, Gαo is the preferred substrate for RGS6 and its GAP activity.

Fig. 6.

Fig. 6.

Gα substrate specificity of RGS6/Gβ5. (A) Schematic representation of the BRET assay for measuring RGS GAP activity. Application of antagonist after G protein activation by a GPCR agonist initiates the deactivation of G proteins, decreasing the BRET signal. (B) Real-time monitoring of G protein deactivation. HEK293T/17 cells were transfected with D2R, Gα, Venus-Gβγ, and masGRK3ct-Nluc-HA with (red) or without (black) RGS6/Gβ5. Dopamine (100 μM) and haloperidol (100 μM) were applied to activate D2R and initiate G protein deactivation, respectively. Representative data from three independent experiments are shown. (C) Comparison of kGAP activity of RGS6/Gβ5 on the Gi/o isoforms (F5,12 = 372.6, P < 0.0001; one-way ANOVA). ****P < 0.0001 vs. Gαo isoforms.

o and Gαi2 Ablation Impact GIRK-Dependent Signaling in a GPCR-Dependent Manner.

Previous work utilizing constitutive or conditional knockout mice has implicated G⍺o and Gαi2 isoforms in HR regulation (3133). Accordingly, we crossed an atrial-specific Cre driver line (SLNCre) (34) with conditional G⍺o (G⍺ofl/fl) (35) or Gαi2 (G⍺i2fl/fl) (32) knockout mice to generate mice lacking G⍺o (SLNCre:G⍺ofl/fl) or Gαi2 (SLNCre:Gαi2fl/fl) in atrial (including SAN) tissue. We then recorded currents evoked by CCh or Ado in SAN cells from Cre(+) and Cre(−) littermates (Fig. 7). While maximal CCh-induced currents and desensitization were smaller in SLNCre(+):G⍺ofl/fl SAN cells compared to Cre(−) littermates (Fig. 7 AC), Ado-induced currents and desensitization were not impacted by G⍺o ablation (Fig. 7 FH). Gαi2 ablation had no impact on CCh- or Ado-induced response amplitudes or desensitization (Fig. 7 B, C, G, and H). Loss of G⍺o prolonged activation and deactivation rates of CCh-induced GIRK currents, whereas Gαi2 ablation was without effect (Fig. 7 D and E). The deactivation rate of Ado-induced GIRK currents was also prolonged by G⍺o ablation (Fig. 7J), but interestingly, the activation rate of Ado-induced GIRK currents was accelerated by Gαi2 ablation (Fig. 7I). Collectively, these results align with observations that M2R signals preferentially through G⍺o, whereas A1R does not discriminate between G⍺o and G⍺i. Moreover, the lack of impact of Gαi2 ablation on the CCh- and Ado-induced GIRK current deactivation rates is consistent with the Gαo substrate preference of RGS6.

Fig. 7.

Fig. 7.

Impact of Gαo and Gαi2 ablation on CCh- and Ado-induced GIRK currents. (A) Whole-cell currents evoked by CCh (10 μM) in SAN cells from SLNCre(−):Gαofl/fl (Left, gray) and SLNCre(+):Gαofl/fl (Right, green) mice. (Scale bars: 5 s/500 pA.) (B and C) Peak CCh-induced current density (t20 = 4.3, ***P < 0.001; unpaired t test) and current desensitization (t19 = 2.6, *P < 0.05; unpaired t test) were smaller in SAN cells from SLNCre(+):Gαofl/fl mice as compared to their SLNCre(−):Gαofl/fl counterparts; group sizes ranged from 9 to 12 cells (5 mice) per genotype. There was no difference in CCh-induced current density (t13 = 0.4, P = 0.69; unpaired t test) or desensitization (t13 = 0.5, P = 0.62; unpaired t test) in SAN cells from SLNCre(−):Gαi2fl/fl mice and SLNCre(+):Gαi2fl/fl mice; group sizes ranged from 7 to 8 cells (3 mice) per genotype. (D and E) Activation (t19 = 3.0, **P < 0.01; unpaired t test) and deactivation (t18 = 2.5, *P < 0.05; unpaired t test) rates of CCh-induced currents were prolonged in SAN cells from SLNCre(+):Gαofl/fl mice, relative to SLNCre(−):Gαofl/fl counterparts; group sizes ranged from 8 to 12 cells (5 mice) per genotype. There was no difference in activation (t11 = 0.9, P = 0.38; unpaired t test) or deactivation (t13 = 1.0, P = 0.36; unpaired t test) rates of CCh-induced currents in SAN cells from SLNCre(−):Gαi2fl/fl and SLNCre(+):Gαi2fl/fl mice; group sizes ranged from 6 to 8 cells (3 mice) per genotype. (F) Whole-cell currents evoked by Ado (10 μM) in SAN cells from SLNCre(−):Gαofl/fl (Left, gray) and SLNCre(+):Gαofl/fl (Right, green) mice. (Scale bars: 5 s/500 pA.) (G and H) There was no difference in peak current density (t20 = 1.8, P = 0.08; unpaired t test) or desensitization (t21 = 0.9, P = 0.38; unpaired t test) of Ado-induced currents in SAN cells from SLNCre(−):Gαofl/fl and SLNCre(+):Gαofl/fl mice; group sizes ranged from 11 to 12 cells (4 mice) per genotype. There were no differences in Ado-induced current density (t13 = 0.1, P = 0.91; unpaired t test) or desensitization (t13 = 0.7, P = 0.51; unpaired t test) in SAN cells from SLNCre(−):Gαi2fl/fl and SLNCre(+):Gαi2fl/fl mice; group sizes ranged from 7 to 8 cells (3 mice) per genotype. (I and J) There was no difference in peak current density (t20 = 1.8, P = 0.08; unpaired t test) or desensitization (t21 = 0.9, P = 0.38; unpaired t test) of Ado-induced currents in SAN cells from SLNCre(−):Gαofl/fl and SLNCre(+):Gαofl/fl mice; group sizes ranged from 11 to 12 cells (4 mice) per genotype. There was an increase in activation rate (t13 = 2.2, *P < 0.05; unpaired t test) of Ado-induced currents in SAN cells from SLNCre(+):Gαi2fl/fl mice as compared to SLNCre(−):Gαi2fl/fl littermates. There was no difference in deactivation rate (t12 = 0.08, P = 0.94; unpaired t test) of Ado-induced currents in SAN cells from SLNCre(−):Gαi2fl/fl and SLNCre(+):Gαi2fl/fl mice; group sizes ranged from 7 to 8 cells (3 mice) per genotype.

Finally, we evaluated the impact of G⍺o or Gαi2 ablation on GIRK channel sensitivity to CCh and Ado. While the loss of Gαo did not impact GIRK channel sensitivity to CCh (Fig. 8 AC), Gαi2 ablation increased the sensitivity of GIRK channels to CCh. Conversely, Gαo ablation increased GIRK channel sensitivity to Ado, whereas Gαi2 ablation was without effect (Fig. 8 DF). Thus, loss of Gαo increases the sensitivity of A1R-GIRK channel signaling, whereas loss of Gαi2 increases the sensitivity of M2R-GIRK channel signaling.

Fig. 8.

Fig. 8.

Impact of Gαo or Gαi2 ablation on GIRK channel sensitivity to CCh and Ado. (A) Concentration-response experiments of CCh-induced currents in SAN cells from SLNCre(−):Gαofl/fl (Top) and SLNCre(+):Gαofl/fl (Bottom) mice. (B) Summary of CCh sensitivity experiments in SAN cells from SLNCre(−):Gαofl/fl and SLNCre(+):Gαofl/fl mice. (C) There was no difference in EC50 values of CCh-induced GIRK currents in SAN cells from SLNCre(−):Gαofl/fl (n = 12 cells/3 mice) and SLNCre(+):Gαofl/fl (n = 8 cells/3 mice) mice (t18 = 0.8, P = 0.44; unpaired t test), but there was a decrease in the EC50 value in SAN cells from SLNCre(+):Gαi2fl/fl (n = 5 cells/3 mice) compared to SLNCre(−):Gαi2fl/fl (n = 8 cells/3 mice) mice (t11 = 2.5, *P < 0.05; unpaired t test). (D) Concentration-response experiments of Ado-induced GIRK currents in SAN cells from SLNCre(−):Gαofl/fl (Top) and SLNCre(+):Gαofl/fl (Bottom) mice. (E) Summary of concentration-response experiments of Ado-induced GIRK currents in SAN cells from SLNCre(−):Gαofl/fl and SLNCre(+):Gαofl/fl mice. (F) The EC50 value for Ado-induced signaling in SAN cells from SLNCre(+):Gαofl/fl (n = 15 cells/3 mice) mice was lower than that measured in SAN cells from SLNCre(−):Gαofl/fl (n = 12 cells/3 mice) mice (t25 = 3.3, **P < 0.01; unpaired t test), but there was no difference in EC50 value in SAN cells from SLNCre(−):Gαi2fl/fl (n = 7 cells/3 mice) and SLNCre(+):Gαi2fl/fl (n = 7 cells/3 mice) mice (t12 = 1.0, P = 0.35; unpaired t test).

Discussion

While M2R and A1R signal to a shared pool of GIRK channels in mouse SAN cells, the amplitude and kinetics of these signaling pathways are distinct. RGS6 exerts a GPCR-dependent influence on GIRK-dependent signaling in mouse SAN cells, modulating the sensitivity and kinetics of M2R-GIRK signaling and the amplitude of A1R-GIRK signaling. Innate GPCR-G protein coupling preferences appear to contribute to the distinctions in M2R- and A1R-GIRK signaling pathways in SAN cells. Our data suggest that Gαo plays a more important role in mediating M2R-GIRK than A1R-GIRK signaling in mouse SAN cells. This, together with the strong Gαo substrate preference of RGS6, provides a plausible explanation for the GPCR-dependent influence of RGS6 on GIRK-dependent signaling in mouse SAN cells.

A1R-GIRK responses are smaller than M2R-GIRK responses in mouse SAN cells; parallel observations have been noted in mouse, rat, and guinea pig atrial myocytes (3638). A maximal M2R-induced GIRK response completely occludes the A1R-GIRK response in SAN cells, suggesting that M2R activation can elicit a maximal GIRK-dependent response, while A1R activation can engage only a fraction of available GIRK channels. Different levels of M2R and A1R expression likely contribute to the differences in response amplitudes and activation kinetics. Indeed, overexpression of A1R in rat atrial myocytes increased Ado-induced GIRK currents and accelerated the current activation rate (39). Thus, the level of A1R and/or G protein intermediate appears to limit A1R-GIRK signaling amplitude in mouse SAN cells.

Considering the GAP activity of RGS6, Rgs6 ablation should result in increased steady-state levels of activated Gαo (and Gβγ) following M2R and A1R activation. Indeed, A1R-GIRK response amplitudes were enhanced by Rgs6 ablation without up-regulation of A1R or GIRK channels. Thus, the amplitude of A1R-GIRK signaling in mouse SAN cells is likely limited by the amount of activated G protein released by A1R activation. Interestingly, loss of RGS6 had no impact on CCh-induced GIRK current amplitude, suggesting that the GIRK channel limits the M2R-GIRK responses in these cells, a contention supported by our occlusion data. The enhanced sensitivity of GIRK channels to CCh in SAN cells from Rgs6−/− mice further suggests that the loss of RGS6 effectively increases the fraction of spare M2R receptors in these cells. Interestingly, we recently reported an increase in M2R-GIRK current amplitude in ventricular myocytes from Rgs6−/− mice, where expression M2R is lower than in the atria (40). Thus, RGS6 may impact GPCR-effector response amplitudes when the level of G protein, rather than effector, is limiting.

RGS proteins may also facilitate GIRK-dependent signaling in a GAP-independent manner. Mice heterozygous for an RGS-insensitive Gαo, for example, exhibit blunted GIRK channel responses to mu opioid receptor (MOR) activation in some neurons (41), suggesting that RGS proteins promote MOR-GIRK coupling. Notably, we observed a modest decrease in GIRK currents elicited by the GIRK channel activator ML297 in SAN cells from Rgs6−/− mice. Thus, RGS6 may facilitate efficient GIRK channel signaling in SAN cells, perhaps via physical interaction (42). The increase in A1R-GIRK channel signaling seen with Rgs6 ablation, however, suggests that this is not the main influence of RGS6 in SAN cells. Other work has called attention to the role of adaptor proteins, including R7BP and GPR158, in fine-tuning the influence of RGS proteins (21, 4345). While R7BP or GPR158 expression has not been observed in the heart (46, 47), an unknown adaptor protein(s) may influence the impact of RGS6/Gβ5 on GIRK-dependent signaling in cardiomyocytes.

Previous work has implicated Gαo in the bradycardic effects of A1R and M2R activation. For example, isolated hearts from mice with a global knockout of Gαo, but not of Gαi2 or Gαi3, exhibited diminished CCh-induced bradycardia (33). Additionally, work with embryonic stem cell-derived cardiomyocytes (ESDCs) showed that the reduction in isoproterenol-stimulated beating rate elicited by M2R and A1R activation was exaggerated in cells containing an RGS-insensitive version of Gαo (48). Our findings that Gαo is a critical mediator of M2R-GIRK signaling in mouse SAN cells, and that Rgs6 ablation enhances both M2R- and A1R-mediated bradycardia, are in line with these reports. Interestingly, the enhanced A1R-induced suppression of isoproterenol-stimulated beating rate seen in ESDCs harboring a RGS-insensitive Gαo mutant was not blunted by TTQ, suggesting that this effect of A1R activation may not be GIRK-dependent (48). This contrasts with our finding that A1R-mediated bradycardia is absent in Girk4−/− mouse hearts. These discrepancies likely arise from the different cell types and model systems employed, where the relative complements of GPCRs, G proteins, effectors, and RGS proteins may differ.

There is little consensus on relevance of Gαi2 to HR regulation. Mice with a global deletion of Gαi2 exhibited diminished CCh-induced bradycardia and resting tachycardia (31). Mice harboring an RGS-insensitive Gαi2 displayed enhanced M2R-induced bradycardia, and the enhanced effect of CCh on beating rate of ESDCs harboring this mutation was reversed by TTQ (48). These findings are consistent with a prominent role for Gαi2 in mediating M2R-GIRK signaling in SAN cells. However, atrial myocytes from mice lacking Gαi2 exhibited increased CCh-induced GIRK responses, which was attributed to increased GIRK subunit expression (49). Moreover, mice lacking Gαi2 selectively in the cardiac conduction system exhibited resting tachycardia but normal CCh-induced bradycardia (32). Thus, the impact of Gαi2 ablation and RGS-insensitive Gαi2 on CCh-induced bradycardia is model-dependent and may reflect an indirect (noncardiac) influence of Gαi2 on HR regulation. Additionally, the impact of Gαi2 on resting HR may be exerted through GIRK-independent mechanisms (50). For example, M2R activation inhibits the hyperpolarization-activated channel (If) by suppressing adenylyl cyclase activity and cAMP production (3). Interestingly, M2R-If coupling is more sensitive than M2R-GIRK signaling in SAN cells (51). Our data show that Gαo plays a critical role in M2R-GIRK signaling. This pathway is subject to negative regulation by RGS6 and therefore may be engaged only by strong parasympathetic activation. Gαi2, which is less susceptible to the GAP activity of RGS6, may mediate M2R-If coupling during periods of weaker parasympathetic activity.

It is important to recognize that genetic ablation of an individual Gα isoform redirects signaling to residual G protein isoforms. Ablation of Gαo, for example, routes M2R-GIRK signaling through Gαi. In our hands, this yielded small CCh-induced GIRK currents with slow activation rates, reminiscent of Ado-induced GIRK currents in wild-type SAN cells. In contrast, routing A1R-GIRK signaling through Gαi had no impact on Ado-induced current amplitude or activation kinetics. Gαo ablation did increase the sensitivity of GIRK channels to Ado, suggesting that (residual) Gαi is more efficient than Gαo at mediating A1R-GIRK signaling in mouse SAN cells. Similarly, Gαi2 ablation increased the sensitivity of GIRK channels to CCh, suggesting that (residual) Gαo is a more efficient mediator than Gαi2 of M2R-GIRK signaling in mouse SAN cells. The modest overall impact of Gαi2 ablation, particularly on A1R-GIRK signaling, is likely attributable to the presence of residual Gαi1 and Gαi3 isoforms (49). Indeed, this likely explains why there was no impact of global ablation of any single G-protein isoform (Gαo, Gαi2, Gαi1/3) on GIRK-dependent bradycardia elicited by the A1R agonist 2-chloro-N6-cyclopentyladenosine (CCPA) (32).

Ado can provoke atrial fibrillation through shortening of the effective refractory period, an effect thought to be mediated by GIRK channel activation in atrial myocytes (14). Enhanced A1R and GIRK channel expression are associated with increased bradycardia in a tachypacing-induced chronic heart failure model in dogs (52). Moreover, atrial fibrillation induced by A1R activation or M2R activation (via vagal nerve stimulation) is thought to be mediated in part by GIRK channel activity (53, 54). While loss of GIRK4 in mice conferred resistance to pacing-induced atrial fibrillation (55), Rgs6 ablation resulted in a higher incidence of atrial fibrillation induction (26). Similar to the impact of Girk4 ablation, peptides targeting specific Gαi isoforms reduced atrial fibrillation susceptibility during vagal nerve stimulation in dogs (56). These and related observations, together with our results, show that interventions that suppress the influence of inhibitory G-protein signaling in atria by targeting signaling mediators and regulators or effectors could prove beneficial for treatment of certain arrhythmias and heart failure.

Insights gained in this study are likely to be relevant to the compartmentalization of GPCR-dependent signaling in other cell types, including neurons. GIRK channel regulation by inhibitory GPCRs has been implicated in neurological disorders including epilepsy, pain and analgesia, anxiety and depression, and addiction (57, 58). Neuronal GIRK-dependent signaling pathways regulated by GABABR, 5-HT1AR, and A1R exhibit distinct amplitude and kinetic profiles and are differentially susceptible to plasticity (5962). Our previous work has highlighted the integral role of RGS7, another member of the R7 RGS family, in modulating the kinetics and sensitivity of GABABR-GIRK signaling in hippocampal neurons (42, 43, 63, 64). Furthermore, RGS7 prefers Gαo over Gαi as a substrate for its GAP activity (28). Thus, GPCR-G protein and RGS-G protein substrate preferences likely shape neuronal GPCR-GIRK signaling dynamics in neurons and may help the functional compartmentalization of these signaling pathways.

Materials and Methods

Animals.

All procedures involving mice were approved by the Institutional Animal Care and Use Committee of the University of Minnesota, and experiments were conducted in accordance with guidelines set by the NIH. The generation of Girk4−/− and Rgs6−/− mice was described previously (10, 19). C57BL/6J mice, bred on site or purchased from The Jackson Laboratory, were used as wild-type controls for these studies. Mice lacking either Gαo or Gαi2 selectively in atrial/SAN tissue were generated by crossing the SLNCre driver mouse line (34) with floxed versions of either Gαo (35) or Gαi2 (32), resulting in SLNCre:Gαofl/fl and SLNCre:Gαi2fl/fl mouse lines, respectively. Male and female mice were group-housed on a 12-h light/dark cycle, given free access to food and water, and used for experiments at ages 8 to 12 wk.

SAN Cell Culture and Recordings.

SAN cells were prepared for electrophysiological analysis as described (40). See SI Appendix for further details.

Isolated Heart Recordings.

Hearts were excised and placed into ice-cold, oxygenated Tyrode’s solution, and the aorta was quickly cannulated. Cannulated hearts were then placed into a warm (37 ± 1 °C) Tyrode’s bath, and iWorx platinum recording electrodes were placed near/on the surface of the heart. Oxygenated Tyrode’s solution was then perfused at 2 to 3 mL/min, and a baseline HR was recorded. Increasing concentrations of CCh or CPA (Tocris Bioscience) were then perfused via peristaltic pump for at least 15 min per dose. The electrocardiogram (ECG) signal was acquired with LabScribe v.3 software (iWorx) and filtered as appropriate. The derivative of that channel was computed to account for movements in baseline and to amplify the signal for subsequent analysis. A 30-s segment from the last minute of exposure to each agonist dose was then exported to Kubios HRV v.2 (65) for HR analysis, utilizing artifact correction as appropriate.

Quantitative RT-PCR.

Total RNA was isolated from freshly isolated atrial tissue samples from adult wild-type mice and Rgs6−/− mice using the RNeasy fibrous tissue kit (Qiagen), according to manufacturer recommendations. Reverse transcription of 1.2 μg of total RNA per sample was performed using Maxima H Minus First Strand cDNA Synthesis Kit (ThermoFisher Scientific). Quantitative PCR was performed in a QuantStudio3 Real Time PCR System (Applied Biosystems) with the Fast SYBR Green Master Mix (ThermoFisher Scientific). See SI Appendix for further details, including target-specific primer sequences, reaction conditions, and analysis information.

Fast Kinetic BRET Assay.

HEK293T/17 cells were grown in Dulbecco’s Modified Eagle Media (Thermo Fisher Scientific) supplemented with 10% fetal bovine serum (Millipore Sigma), minimum Eagle’s medium nonessential amino acids, 1 mM sodium pyruvate, and antibiotics (100 units/mL penicillin and 100 mg/mL streptomycin) at 37 °C in a humidified incubator containing 5% CO2. Culture dishes (3.5 cm) were incubated for 10 min at 37 °C with 1 mL of 10 mg/mL growth-factor–reduced Matrigel (BD Biosciences) in culture medium. For transfection, cells were seeded into 3.5-cm dishes at a density of 2 × 106 cells/dish. After 2 h, expression constructs (total 5 μg/dish) were added to the cells using PLUS (5 μg/dish) and Lipofectamine LTX (6 μL/dish) reagents (Thermo Fisher Scientific). BRET measurements of Venus-Gβ1γ2 and masGRK3ct-Nluc-HA were performed to measure G protein activation by M2R and A1R, or the GAP activity of RGS6/Gβ5 in living cells, as described (27, 66). See SI Appendix for further details, including information about DNA constructs and transfection as well as BRET assay procedures and analysis.

Statistical Analysis.

All data were analyzed using Prism v.8.2.1 software (GraphPad Software) and are presented as mean ± SEM. Statistical outliers were identified and excluded with Grubb’s outlier test. The level of statistical significance was set at P < 0.05. Specific statistical analyses are denoted within the figure legends.

Data Availability.

All experimental procedures and data supporting the findings of this paper are included in the article and SI Appendix. Requests for reagents used in this study should be directed to K.W.

Supplementary Material

Supplementary File
pnas.2001270117.sapp.pdf (422.4KB, pdf)

Acknowledgments

We thank Dr. C. David Weaver and Dr. Corey Hopkins for providing ML297, Nickolas Skamangas for technical support, and Hannah Oberle and Mehrsa Zahiremami for exceptional care of the mouse colony. This work was supported by NIH Grants HL105550 (to K.W. and K.A.M.), R21 HL124503 (to A.N.), and F31 HL139090 (A.A.) and by NIH Intramural Research Program Project Grant Z01-ES-101643 (to L.B.).

Footnotes

The authors declare no competing interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at https://www.pnas.org/lookup/suppl/doi:10.1073/pnas.2001270117/-/DCSupplemental.

References

  • 1.Hauser A. S., Attwood M. M., Rask-Andersen M., Schiöth H. B., Gloriam D. E., Trends in GPCR drug discovery: New agents, targets and indications. Nat. Rev. Drug Discov. 16, 829–842 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Dyavanapalli J., Dergacheva O., Wang X., Mendelowitz D., Parasympathetic vagal control of cardiac function. Curr. Hypertens. Rep. 18, 22 (2016). [DOI] [PubMed] [Google Scholar]
  • 3.Dhein S., van Koppen C. J., Brodde O. E., Muscarinic receptors in the mammalian heart. Pharmacol. Res. 44, 161–182 (2001). [DOI] [PubMed] [Google Scholar]
  • 4.Sakmann B., Noma A., Trautwein W., Acetylcholine activation of single muscarinic K+ channels in isolated pacemaker cells of the mammalian heart. Nature 303, 250–253 (1983). [DOI] [PubMed] [Google Scholar]
  • 5.Logothetis D. E., Kurachi Y., Galper J., Neer E. J., Clapham D. E., The β γ subunits of GTP-binding proteins activate the muscarinic K+ channel in heart. Nature 325, 321–326 (1987). [DOI] [PubMed] [Google Scholar]
  • 6.Wickman K. D. et al., Recombinant G-protein β γ-subunits activate the muscarinic-gated atrial potassium channel. Nature 368, 255–257 (1994). [DOI] [PubMed] [Google Scholar]
  • 7.Corey S., Clapham D. E., Identification of native atrial G-protein-regulated inwardly rectifying K+ (GIRK4) channel homomultimers. J. Biol. Chem. 273, 27499–27504 (1998). [DOI] [PubMed] [Google Scholar]
  • 8.Corey S., Krapivinsky G., Krapivinsky L., Clapham D. E., Number and stoichiometry of subunits in the native atrial G-protein-gated K+ channel, IKACh. J. Biol. Chem. 273, 5271–5278 (1998). [DOI] [PubMed] [Google Scholar]
  • 9.Krapivinsky G. et al., The G-protein-gated atrial K+ channel IKACh is a heteromultimer of two inwardly rectifying K(+)-channel proteins. Nature 374, 135–141 (1995). [DOI] [PubMed] [Google Scholar]
  • 10.Wickman K., Nemec J., Gendler S. J., Clapham D. E., Abnormal heart rate regulation in GIRK4 knockout mice. Neuron 20, 103–114 (1998). [DOI] [PubMed] [Google Scholar]
  • 11.Bettahi I., Marker C. L., Roman M. I., Wickman K., Contribution of the Kir3.1 subunit to the muscarinic-gated atrial potassium channel IKACh. J. Biol. Chem. 277, 48282–48288 (2002). [DOI] [PubMed] [Google Scholar]
  • 12.Mesirca P. et al., The G-protein-gated K+ channel, IKACh, is required for regulation of pacemaker activity and recovery of resting heart rate after sympathetic stimulation. J. Gen. Physiol. 142, 113–126 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lee S. W. et al., Atrial GIRK channels mediate the effects of vagus nerve stimulation on heart rate dynamics and arrhythmogenesis. Front. Physiol. 9, 943 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Layland J., Carrick D., Lee M., Oldroyd K., Berry C., Adenosine: Physiology, pharmacology, and clinical applications. JACC Cardiovasc. Interv. 7, 581–591 (2014). [DOI] [PubMed] [Google Scholar]
  • 15.Mustafa S. J., Morrison R. R., Teng B., Pelleg A., Adenosine receptors and the heart: Role in regulation of coronary blood flow and cardiac electrophysiology. Handb. Exp. Pharmacol. 193, 161–188 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Belardinelli L., Giles W. R., West A., Ionic mechanisms of adenosine actions in pacemaker cells from rabbit heart. J. Physiol. 405, 615–633 (1988). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Woodard G. E., Jardín I., Berna-Erro A., Salido G. M., Rosado J. A., Regulators of G-protein-signaling proteins: Negative modulators of G-protein-coupled receptor signaling. Int. Rev. Cell Mol. Biol. 317, 97–183 (2015). [DOI] [PubMed] [Google Scholar]
  • 18.Posokhova E., Wydeven N., Allen K. L., Wickman K., Martemyanov K. A., RGS6/Gβ5 complex accelerates IKACh gating kinetics in atrial myocytes and modulates parasympathetic regulation of heart rate. Circ. Res. 107, 1350–1354 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wydeven N., Posokhova E., Xia Z., Martemyanov K. A., Wickman K., RGS6, but not RGS4, is the dominant regulator of G protein signaling (RGS) modulator of the parasympathetic regulation of mouse heart rate. J. Biol. Chem. 289, 2440–2449 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang J. et al., RGS6, a modulator of parasympathetic activation in heart. Circ. Res. 107, 1345–1349 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Anderson G. R., Posokhova E., Martemyanov K. A., The R7 RGS protein family: Multi-subunit regulators of neuronal G protein signaling. Cell Biochem. Biophys. 54, 33–46 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jin W., Klem A. M., Lewis J. H., Lu Z., Mechanisms of inward-rectifier K+ channel inhibition by tertiapin-Q. Biochemistry 38, 14294–14301 (1999). [DOI] [PubMed] [Google Scholar]
  • 23.Jin W., Lu Z., Synthesis of a stable form of tertiapin: A high-affinity inhibitor for inward-rectifier K+ channels. Biochemistry 38, 14286–14293 (1999). [DOI] [PubMed] [Google Scholar]
  • 24.Kaufmann K. et al., ML297 (VU0456810), the first potent and selective activator of the GIRK potassium channel, displays antiepileptic properties in mice. ACS Chem. Neurosci. 4, 1278–1286 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wydeven N. et al., Mechanisms underlying the activation of G-protein-gated inwardly rectifying K+ (GIRK) channels by the novel anxiolytic drug, ML297. Proc. Natl. Acad. Sci. U.S.A. 111, 10755–10760 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Posokhova E. et al., Essential role of the m2R-RGS6-IKACh pathway in controlling intrinsic heart rate variability. PLoS One 8, e76973 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Masuho I. et al., Distinct profiles of functional discrimination among G proteins determine the actions of G protein-coupled receptors. Sci. Signal. 8, ra123 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Masuho I., Xie K., Martemyanov K. A., Macromolecular composition dictates receptor and G protein selectivity of regulator of G protein signaling (RGS) 7 and 9-2 protein complexes in living cells. J. Biol. Chem. 288, 25129–25142 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Digby G. J., Sethi P. R., Lambert N. A., Differential dissociation of G protein heterotrimers. J. Physiol. 586, 3325–3335 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hooks S. B. et al., RGS6, RGS7, RGS9, and RGS11 stimulate GTPase activity of Gi family G-proteins with differential selectivity and maximal activity. J. Biol. Chem. 278, 10087–10093 (2003). [DOI] [PubMed] [Google Scholar]
  • 31.Zuberi Z., Birnbaumer L., Tinker A., The role of inhibitory heterotrimeric G proteins in the control of in vivo heart rate dynamics. Am. J. Physiol. Regul. Integr. Comp. Physiol. 295, R1822–R1830 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sebastian S. et al., The in vivo regulation of heart rate in the murine sinoatrial node by stimulatory and inhibitory heterotrimeric G proteins. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305, R435–R442 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Duan S. Z., Christe M., Milstone D. S., Mortensen R. M., Go but not Gi2 or Gi3 is required for muscarinic regulation of heart rate and heart rate variability in mice. Biochem. Biophys. Res. Commun. 357, 139–143 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nakano H. et al., Cardiac origin of smooth muscle cells in the inflow tract. J. Mol. Cell. Cardiol. 50, 337–345 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ang R., Abramowitz J., Birnbaumer L., Gourine A. V., Tinker A., The role of GαO-mediated signaling in the rostral ventrolateral medulla oblongata in cardiovascular reflexes and control of cardiac ventricular excitability. Physiol. Rep. 4, e12860 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cho H., Regulation of adenosine-activated GIRK channels by Gq-coupled receptors in mouse atrial myocytes. Korean J. Physiol. Pharmacol. 14, 145–150 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bünemann M., Pott L., Down-regulation of A1 adenosine receptors coupled to muscarinic K+ current in cultured Guinea-pig atrial myocytes. J. Physiol. 482, 81–92 (1995). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wellner-Kienitz M. C., Bender K., Meyer T., Bünemann M., Pott L., Overexpressed A(1) adenosine receptors reduce activation of acetylcholine-sensitive K(+) current by native muscarinic M(2) receptors in rat atrial myocytes. Circ. Res. 86, 643–648 (2000). [DOI] [PubMed] [Google Scholar]
  • 39.Kienitz M.-C., Littwitz C., Bender K., Pott L., Remodeling of inward rectifying K(+) currents in rat atrial myocytes by overexpression of A(1)-adenosine receptors. Basic Res. Cardiol. 106, 953–966 (2011). [DOI] [PubMed] [Google Scholar]
  • 40.Anderson A. et al., Expression and relevance of the G protein-gated K+ channel in the mouse ventricle. Sci. Rep. 8, 1192 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.McPherson K. B. et al., Regulators of G-protein signaling (RGS) proteins promote receptor coupling to G-protein-coupled inwardly rectifying potassium (GIRK) channels. J. Neurosci. 38, 8737–8744 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Xie K. et al., Gbeta5 recruits R7 RGS proteins to GIRK channels to regulate the timing of neuronal inhibitory signaling. Nat. Neurosci. 13, 661–663 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ostrovskaya O. I. et al., Inhibitory signaling to ion channels in hippocampal neurons is differentially regulated by alternative macromolecular complexes of RGS7. J. Neurosci. 38, 10002–10015 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jayaraman M., Zhou H., Jia L., Cain M. D., Blumer K. J., R9AP and R7BP: Traffic cops for the RGS7 family in phototransduction and neuronal GPCR signaling. Trends Pharmacol. Sci. 30, 17–24 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Orlandi C. et al., Orphan receptor GPR158 is an allosteric modulator of RGS7 catalytic activity with an essential role in dictating its expression and localization in the brain. J. Biol. Chem. 290, 13622–13639 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Grabowska D. et al., Postnatal induction and localization of R7BP, a membrane-anchoring protein for regulator of G protein signaling 7 family-Gbeta5 complexes in brain. Neuroscience 151, 969–982 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Orlandi C. et al., GPR158/179 regulate G protein signaling by controlling localization and activity of the RGS7 complexes. J. Cell Biol. 197, 711–719 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fu Y. et al., Endogenous RGS proteins and Galpha subtypes differentially control muscarinic and adenosine-mediated chronotropic effects. Circ. Res. 98, 659–666 (2006). [DOI] [PubMed] [Google Scholar]
  • 49.Nobles M., Montaigne D., Sebastian S., Birnbaumer L., Tinker A., Differential effects of inhibitory G protein isoforms on G protein-gated inwardly rectifying K+ currents in adult murine atria. Am. J. Physiol. Cell Physiol. 314, C616–C626 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ye C., Sowell M. O., Vassilev P. M., Milstone D. S., Mortensen R. M., Galpha(i2), Galpha(i3)and Galpha(o) are all required for normal muscarinic inhibition of the cardiac calcium channels in nodal/atrial-like cultured cardiocytes. J. Mol. Cell. Cardiol. 31, 1771–1781 (1999). [DOI] [PubMed] [Google Scholar]
  • 51.DiFrancesco D., Ducouret P., Robinson R. B., Muscarinic modulation of cardiac rate at low acetylcholine concentrations. Science 243, 669–671 (1989). [DOI] [PubMed] [Google Scholar]
  • 52.Long V. P. III et al., Chronic heart failure increases negative chronotropic effects of adenosine in canine sinoatrial cells via A1R stimulation and GIRK-mediated IKado. Life Sci. 240, 117068 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hashimoto N., Yamashita T., Tsuruzoe N., Tertiapin, a selective IKACh blocker, terminates atrial fibrillation with selective atrial effective refractory period prolongation. Pharmacol. Res. 54, 136–141 (2006). [DOI] [PubMed] [Google Scholar]
  • 54.Li N. et al., Adenosine-induced atrial fibrillation: Localized reentrant drivers in lateral right atria due to heterogeneous expression of adenosine A1 receptors and GIRK4 subunits in the human heart. Circulation 134, 486–498 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kovoor P. et al., Evaluation of the role of I(KACh) in atrial fibrillation using a mouse knockout model. J. Am. Coll. Cardiol. 37, 2136–2143 (2001). [DOI] [PubMed] [Google Scholar]
  • 56.Aistrup G. L. et al., Targeted G-protein inhibition as a novel approach to decrease vagal atrial fibrillation by selective parasympathetic attenuation. Cardiovasc. Res. 83, 481–492 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lüscher C., Slesinger P. A., Emerging roles for G protein-gated inwardly rectifying potassium (GIRK) channels in health and disease. Nat. Rev. Neurosci. 11, 301–315 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Luján R., Marron Fernandez de Velasco E., Aguado C., Wickman K., New insights into the therapeutic potential of Girk channels. Trends Neurosci. 37, 20–29 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Leaney J. L., Contribution of Kir3.1, Kir3.2A and Kir3.2C subunits to native G protein-gated inwardly rectifying potassium currents in cultured hippocampal neurons. Eur. J. Neurosci. 18, 2110–2118 (2003). [DOI] [PubMed] [Google Scholar]
  • 60.Chung H. J. et al., G protein-activated inwardly rectifying potassium channels mediate depotentiation of long-term potentiation. Proc. Natl. Acad. Sci. U.S.A. 106, 635–640 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nassirpour R. et al., Morphine- and CaMKII-dependent enhancement of GIRK channel signaling in hippocampal neurons. J. Neurosci. 30, 13419–13430 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Marron Fernandez de Velasco E. et al., GIRK2 splice variants and neuronal G protein-gated K+ channels: Implications for channel function and behavior. Sci. Rep. 7, 1639 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ostrovskaya O. et al., RGS7/Gβ5/R7BP complex regulates synaptic plasticity and memory by modulating hippocampal GABABR-GIRK signaling. eLife 3, e02053 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fajardo-Serrano A. et al., Association of Rgs7/Gβ5 complexes with Girk channels and GABAB receptors in hippocampal CA1 pyramidal neurons. Hippocampus 23, 1231–1245 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Tarvainen M. P., Niskanen J.-P., Lipponen J. A., Ranta-Aho P. O., Karjalainen P. A., Kubios HRV: Heart rate variability analysis software. Comput. Methods Programs Biomed. 113, 210–220 (2014). [DOI] [PubMed] [Google Scholar]
  • 66.Masuho I., Martemyanov K. A., Lambert N. A., Monitoring G protein activation in cells with BRET. Methods Mol. Biol. 1335, 107–113 (2015). [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.

Supplementary Materials

Supplementary File
pnas.2001270117.sapp.pdf (422.4KB, pdf)

Data Availability Statement

All experimental procedures and data supporting the findings of this paper are included in the article and SI Appendix. Requests for reagents used in this study should be directed to K.W.


Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES