Abstract

Objective We sought to determine if different β-adrenergic receptor (βAR) subtypes, and their associated signalling machinery, are functionally localized to nuclear membranes.

Methods Employing enriched nuclear preparations, we assayed the specific presence of βAR by measuring 125I-cyanopindolol (CYP) binding, Western blotting, confocal microscopy and functional assays.

Results Western blots of rat heart nuclear fractions and confocal immunofluorescent analysis of adult rat and mouse ventricular cardiomyocytes displayed the presence of β1AR and β3AR but, surprisingly, not the β2AR on nuclear membranes. Nuclear localization of downstream signalling partners Gs, Gi and adenylyl cyclases II and V/VI was also demonstrated. The functional relevance of nuclear βAR was shown by receptor-mediated stimulation of adenylyl cyclase activity by isoproterenol but not the β3AR-selective agonist CL 316243 in enriched nuclear preparations. We also examined the effect of subtype-selective ligands on the initiation of RNA synthesis in isolated nuclei. Both isoproterenol and another β3AR-selective agonist, BRL 37344, increased RNA synthesis which was inhibited by pertussis toxin (PTX). Neither a β1AR-selective agonist, xamoterol, nor a β2AR-selective agonist, procaterol, was able to stimulate transcription. However, both CGP 20712A and ICI 118,551 blocked isoproterenol-mediated effects to varying extents. PTX treatment also revealed that nuclear βAR may be coupled to other signalling pathways in addition to Gi, as stimulation under these conditions reduced initiation of transcription below basal levels.

Conclusion These results highlight differential subcellular localization for βAR subtypes and indicate that βAR may have specific roles in regulating nuclear function in cardiomyocytes.

This article is referred to in the Editorial by J. Neumann (pages 6–7) in this issue.

1 Introduction

An increasing number of GPCRs are targetted to the nuclear membrane, including lysophosphatidic acid receptors [1], metabotropic glutamate receptors (MGluR5, [2]), apelin receptors [3], platelet activating factor (PAF) receptors [4], bradykinin B2 receptors [3], angiotensin 2 type I receptors [3,5–7], prostaglandin receptors [8] and endothelin receptors [9]. In addition, a large number of proteins, classically associated with receptor-mediated events at the cell surface, including heterotrimeric G proteins ([8,10,11]; reviewed in [12]), adenylyl cyclase isoforms [13,14], phospholipase A2[15], phospholipase Cβ [16], phospholipase D [17], RGS proteins (reviewed in [18]), β-arrestin1 [19,20], G protein-coupled receptor kinases [21–23], A kinase anchoring proteins (AKAPs) and PKA [24], among others, have been demonstrated to be trafficked to the nucleus and/or nuclear membrane.

In mammalian cardiomyocytes, three βAR subtypes are found which differ in their signalling partners, subcellular localization and desensitization profiles. β1ARs, localized to the sarcolemma and t-tubular network, plays a predominant role in regulating myocyte contractility. β2ARs, similarly distributed, play a more modest role in regulating inotropic and lusitropic responses. Both receptors signal through Gs and adenylyl cyclase with similar efficacy although the signals are compartmentalized differently possibly due to localization in distinct membrane microdomains and/or dual coupling of the β2AR to Gs and Gi (reviewed in [25–27]). β3ARs are also found in cardiomyocytes although their function remains ill-defined. Hence, βAR subtypes may serve unique functions and play non-redundant roles within the cardiomyocyte.

In the process of studying receptor trafficking and heterodimerization in mouse ventricular cardiomyocytes using recombinant adenoviruses, we noted a perinuclear distribution of overexpressed β1AR and β2AR [28]. We therefore wished to study receptor distribution in native myocytes and assess what function putative nuclear βAR might have.

2 Experimental procedures

2.1 Materials

Polyclonal rabbit anti-β1AR, β2AR, Gαs, Gαi1-3, adenylyl cyclases II, V/VI and polyclonal goat anti-β3AR were from Santa Cruz Biotechnology (Santa Cruz, CA). Mouse anti-nucleoporin p62 (Nup-62) monoclonal antibodies (1:200) were from BD Transduction Laboratories. Appropriate TRITC-, CY5-, and HRP-conjugated secondary antibodies were from Jackson ImmunoResearch Laboratories (West Grove, PA). Chemiluminescence reagent Renaissance Plus was from Perkin-Elmer Life Sciences (Woodbridge, Ontario). Triton X-100, leupeptin, Protector RNAse inhibitor and PMSF were from Roche (Laval, Quebec). SDS-polyacrylamide gel electrophoresis reagents, nitrocellulose (0.22 μm) were from Bio-Rad (Mississauga, Ontario). Essential fatty acid-free BSA, CL 316243, PTX, isoproterenol, CGP 20712A and ICI 118,551 were from Sigma (Mississauga, Ontario). Xamoterol, procaterol and BRL 37344 were from Tocris (Ellisville, MO). Unless otherwise stated, all reagents were of analytical grade and were purchased from VWR Canlab (Ville Mont-Royal, Quebec) or Fisher Scientific (Mississauga, Ontario). [α32P]-UTP (specific activity 3000 Ci/mmol), [125I]-cyanopindolol (CYP) and [α32P]-ATP were from Perkin-Elmer.

2.2 Isolation of ventricular myocytes

Calcium-tolerant cardiomyocytes were isolated from adult rats [29] or mice [30] as described. These preparations provided 8–10 million cells/heart with 70–85% viability, as assessed by the presence of quiescent cells with rod-shaped morphology. The experimental protocol conformed with the Guide for the Care and Use of Laboratory Animals published by the U.S. National Institutes of Health (NIH Publication No. 85-23, revised 1996).

2.3 Isolation of nuclei

Rat cardiac nuclei were isolated as described with modifications [31]. Briefly, rat hearts were pulverized under liquid nitrogen, resuspended in cold PBS and homogenized (Polytron, 8000 rpm; 2 × 10 s, Fraction 1). All subsequent steps were carried out on ice or at 5 °C. Homogenates were centrifuged for 15 min at 500 × g and supernatants, referred to as Fraction 2, diluted 1:1 with buffer A (10 mM K-HEPES (pH 7.9), 1.5 mM MgCl2, 10 mM KCl, 1 mM DTT, 25 μg/ml leupeptin, 0.2 mM Na3VO4), incubated 10 min on ice, and centrifuged for 15 min at 2000 × g. The resulting supernatant was discarded. The pellet, referred to as crude nuclei (Fraction 3) was resuspended in buffer B (0.3 M K-HEPES pH 7.9, 1.5 M KCl, 0.03 M MgCl2, 25 μg/ml leupeptin, 0.2 mM Na3VO4), incubated on ice for 10 min, and centrifuged for 15 min at 2000 × g. The pellet, an enriched nuclear fraction (Fraction 4), was resuspended in buffer C (20 mM Na-HEPES (pH 7.9), 25% (v/v) glycerol, 0.42 M NaCl, 1.5 mM MgCl2, 0.2 mM EDTA, 0.2 mM EGTA, 0.5 mM PMSF, 0.5 mM DTT, 25 μg/ml leupeptin, 0.2 mM Na3VO4) or 1X transcription buffer (see below) and either used fresh or aliquoted, snap-frozen using liquid nitrogen, and stored at − 80 °C.

2.4 Receptor quantification

Nuclei were prepared and washed as described above. Total βAR number was calculated using a saturating concentration (300 pM) of [125I]-cyanopindolol (CYP; Perkin Elmer) as the radioligand as described [32].

2.5 Measurement of adenylyl cyclase activity

Adenylyl cyclase activity was assayed in nuclear preparations as described [33] using 100 μg protein in a total volume of 50 μl. Enzyme activities were determined following a 15-min incubation in the presence of various receptor ligands, 100 μM forskolin, 10 mM NaF or vehicle at 37 °C. Data were calculated as pmol cAMP/min/mg protein, and analyzed by least squares regression using Prism 4.0cx (GraphPad Software).

2.6 Transcription initiation

Measurements of transcription initiation were as described [34]. Briefly, 10 μl of isolated nuclei were incubated at 30 °C for 30 min in 20 μl of 1 × transcription buffer (50 mM Tris pH 7.9, 0.15 M KCl, 1 mM MnCl2, 6 mM MgCl2, 1 mM ATP, 2 mM DTT, 1 U/μl RNAse inhibitor) and 10 μCi [α32P]-UTP (3000 Ci/mmol) in the absence of CTP and GTP to prevent chain elongation. After termination of reactions by digestion with DNase I, nuclei were lysed with 10 mM Tris–HCl pH 8.0, 10 mM EDTA and 1% SDS. Duplicate of 5 μl aliquots were dried onto Whatman GF/C glass fibre filters which were batch washed with 5% TCA, containing 20 mM sodium pyrophosphate and dried. 32P incorporation was determined by scintillation counting. DNA concentration was determined by spectrophotometry and incorporation expressed as dpm/μg DNA. Ligands were used as for activation of adenylyl cyclase. Where indicated, nuclei were pretreated with PTX (5 μg/ml) for 2 h.

2.7 Western blotting

Immunoblots were performed on the subcellular fractions described above. Protein concentrations were measured as described [35] using bovine γ-globulin as a standard [9]. We first demonstrated using HEK 293 cells, transfected or not with the three βAR subtypes that individual antibodies recognized cognate receptors specifically in both Western and immunocytochemical applications (data not shown). These antibodies have been used in other studies on rat ventricular cardiomyocytes under similar conditions [36,37]. Similarly, we performed control experiments for our other antibodies.

2.8 Confocal microscopy

The intracellular localization of βAR subtypes and downstream signalling partners was studied using scanning confocal microscopy (LSM 510 Carl Zeiss). Freshly isolated adult ventricular myocytes were plated on laminin-coated coverslips for 1 h (37 °C, 95% O2/5% CO2), fixed for 20 min in PBS (pH 7.4) containing 2% (w/v) paraformaldehyde, then washed thrice in PBS and blocked 1 h at room temperature in PBS containing 5% normal donkey serum (NDS) plus 0.2% (v/v) TX-100. Coverslips were rinsed thrice in PBS and incubated 1 h at 4 °C in PBS containing 10% NDS. Excess serum was removed and cells were incubated overnight at 4 °C with primary antibody diluted 1:100 in PBS containing 2% NDS. Coverslips were rinsed gently three times with PBS, drained, and incubated for 1 h at room temperature with a 1:500 (v/v) dilution of appropriate secondary antibodies (TRITC-conjugated anti-rabbit antibody, or ALEXA 488-conjugated anti-sheep antibody) in PBS containing 2% NDS. Coverslips were washed three times with PBS, drained, and mounted onto glass slides. For co-localization experiments, protocols were modified slightly. Freshly isolated mouse cardiomyocytes were plated on laminin-coated coverslips for 1 h (37 °C, 95% O2/5% CO2) and fixed for 20 min in 2% paraformaldehyde. Coverslips were rinsed three times in PBS and incubated 1 h at 4 °C in PBS containing 2% normal goat serum (NGS) and Triton 0.2% (v/v) when anti-β1AR (1:100) and β2AR (1:100) were used and 2% normal donkey serum (NDS) and Triton 0.2% (v/v) when β3AR (1:100) was used. ALEXA 555-conjugated anti-mouse antibody (1:500 in NGS) was used when murine cardiomyocytes were stained for Nup62. Fluorescence was visualized as serial 0.5-μm-thick optical sections in the z-axis plane of each cell. For each secondary antibody, control experiments were performed in the absence of primary antibody.

2.9 Statistical analysis

Data are presented as the mean±the standard error of the mean (S.E.). The significance of differences between groups was estimated by one-way ANOVA followed by Tukey's multiple comparison test (Prism 4.0cx, GraphPad Software). Differences were considered significant when p<0.05.

3 Results

3.1 Localization of β-adrenergic signalling machinery on nuclear membranes

To determine whether βAR subtypes were trafficked to the nuclear membrane, we used a standard protocol for enrichment of nuclei. As shown in Fig. 1, binding of the βAR-specific ligand [125I]-CYP was detected in both crude (Fraction 3) and enriched (Fraction 4) nuclear preparations. The efficiency of fractionation is well characterized [9]. Fraction 3 and Fraction 4 are significantly enriched for the nuclear membrane marker, Nup62 [38] and have clearly lost both annexin II (a cell surface marker [39]), BiP (an ER chaperone [40]) and GM 130 (a Golgi resident protein [41]). The nuclear fraction is never totally pure but in going from Fraction 3 to Fraction 4, there is a significant enrichment for Nup62 (which was undetectable in Fractions 1 and 2) with concomitant depletion of markers for the cell surface (annexin II is reduced significantly in Fraction 4), ER (BiP is almost undetectable in Fraction 4), and Golgi (GM 130 is reduced significantly in Fraction 4). Initial experiments to determine the exact subtypes present in enriched nuclear fractions were performed using competition experiments with various subtype-selective ligands. These experiments could not clearly distinguish between the different subtypes in isolated nuclei. However, as described below, we used subtype-selective agonists to elicit specific responses in isolated nuclei.

Fig. 1

125I-CYP binding in enriched nuclear fractions. Specific βAR binding (total minus non-specific as measured using 10 μM alprenolol) in various fractions associated with the nuclear enrichment procedure. Fractions are as described in Materials and methods. Data represent mean±S.E. of at least three separate experiments performed in triplicate.

Similar results to those obtained in ligand binding studies were obtained using Western blots on the different crude and enriched nuclear fractions (Fig. 2). However, whereas all three subtypes could be detected in crude lysates (Fraction 1), only the β1AR and β3AR were detected in the enriched nuclear fraction (Fraction 4). A lower molecular weight protein (around 45 kDa) was revealed as a faint band by β2AR antiserum in Fractions 1–3; however this band was unrelated to the β2AR.

Fig. 2

Detection of βAR isoforms in crude and enriched nuclei using Western blotting. Cell fractions were prepared as in Fig. 1. Lanes contained 100 μg protein from Fractions 1–4. Blots were probed using anti-β1AR (A), anti-β2AR (B) or anti-β3AR (C). Molecular weight markers are indicated on the left of the figure. Arrows indicate specific antibody signals previously described in the literature (see text for details). Data are representative of three independent experiments.

Next, confocal microscopy was used in order to further investigate the subcellular localization of βAR subtypes. Confocal image stacks were deconvolved to reduce the amount of out-of-focus haze from secondary antibodies [11]. Immunofluorescence for all three receptors was detected as punctate signals localized on the cell surface and in a striated pattern within myocytes (Fig. 3A, C, E). Although the t-tubular system is formed of continuous invaginations of the plasma membrane, β1AR and β2AR immunofluorescence was more intense on t-tubular membranes than surface membranes. Most interestingly, cell images of β1AR- or β3AR-decorated myocytes revealed an intense intracellular signal on the periphery of the nucleus (Fig. 3A) and in structures that appeared to extend outwards from its apex (Fig. 3E), indicating that they were localized in nuclear or perinuclear membranes. In contrast, no β2AR staining was detected on the nuclear membranes (arrows, Fig. 3A, C, E). No staining was observed in the absence of the primary antibody (Fig. 3B, D, F). Co-localization studies were also performed using mouse ventricular cardiomyocytes where relevant secondary antibodies were available and these confirmed our results using isolated rat cardiomyocytes. Nup-62 colocalized with β1AR and β3AR but not with β2AR (Fig. 4). Colocalization of all three subtypes was detected with caveolin-3 but not with the Golgi marker GM 130 (data not shown) confirming our results using subcellular fractionation.

Fig. 4

Colocalization of βAR isoforms in adult mouse ventricular cardiomyocytes with a marker of the nuclear membrane. Panels A, B and C are representative confocal images showing the colocalization of β1AR, β2AR or β3AR, respectively with Nup-62, a component of the nuclear pore complex. Controls were performed in the absence of primary antibody (D). Data are representative of at least three independent experiments.

Fig. 3

Detection of βAR isoforms in isolated adult rat ventricular cardiomyocytes using immunocytochemistry. Panels A, C and E are representative confocal images showing the distribution of β1AR (A), β2AR (C) or β3AR (E), respectively. Controls were performed in the absence of primary antibody (B, D and F). Scale bar is 10 μm. Arrows indicate nuclei. Data are representative of three independent experiments.

3.2 Functional effects of nuclear β-adrenergic receptors

To assess potential function of the nuclear βAR receptors, immunofluorescence was employed to localize downstream effector molecules. Known downstream effectors, Gαi- and Gαs-subunits as well as adenylyl cyclases (AC) II and V/VI, were examined. Gαi immunofluorescence was apparent on t-tubules and surface membranes, including the intercalated disc region (Fig. 5A). In addition, punctate Gαi immunofluorescence was observed on the nuclear periphery and in intranuclear structures (arrow, Fig. 5A). Images from anti-Gαs-labeled cardiomyocytes revealed punctate immunofluorescence on plasma membranes, t-tubules and nuclear membranes, where staining was most intense (arrow, Fig. 5C). Thus, both G proteins known to couple to βAR subtypes in adult ventricular myocytes [25] were present on nuclear membranes and intranuclear structures. AC II antibodies decorated the cell surface, showed a striated intracellular pattern and labeled nuclei (Fig. 5E). AC II localization to t-tubules was mainly peripheral. AC V/VI immunofluorescence was observed at the surface membranes, t-tubules and was most intense in the intercalated disc regions and nuclei (Fig. 5G). Hence, the relevant βAR signalling machinery is present on nuclear membranes in cardiomyocytes.

Fig. 5

Detection of βAR signalling partners in isolated adult rat left ventricular cardiomyocytes using immunocytochemistry. Panels A, C, E and G are representative confocal images showing the distribution of Giα (A), Gsα (C) ACII (E) or ACV/VI (G), respectively. Controls were performed in the absence of primary antibody (B, D, F and H). Scale bar is 10 μm. Arrows indicate nuclei. Data are representative of three independent experiments.

To more directly explore the functional consequences of nuclear localization of βAR, we first measured the ability of a non-selective agonist, isoproterenol, to stimulate adenylyl cyclase activity in isolated nuclei from rat hearts. We recognize that only 30–40% of the nuclei in the heart come from cardiomyocytes [42]. However, the mechanical procedure we used for nuclear isolation is much more efficient on whole tissues than isolated cells. As can be seen in Fig. 6A, there was a dose-dependent stimulation of adenylyl cyclase activity in enriched nuclear preparations demonstrating that the entire signalling system from receptor to Gs to effector is indeed functional. In the absence of ligand, basal enzyme activity was detected suggesting that the system may also be functional constitutively, i.e. even in the absence of the endogenous ligand (Fig. 6B). Direct activation of G protein with NaF or adenylyl cyclase with forskolin also led to an increase in the production of cAMP in isolated nuclei, confirming the functional presence of the signalling system downstream of the receptor (Fig. 6B). Interestingly, a β3AR-selective agonist CL 316243 had no effect on adenylyl cyclase activity, suggesting that the β1AR is primarily responsible for activation of this effector in nuclei.

Fig. 6

Functional coupling of β1AR but not β3AR to adenylyl cyclase stimulation in enriched nuclear preparations. (A) Membranes isolated from enriched nuclear fractions were stimulated with increasing concentrations of isoproterenol (1 nM to 100 μM) and the production of [32P]-cAMP from [α32P]-ATP was assessed. (B) Stimulation of adenylyl cyclase activity in response to activation by 1 μM isoproterenol, the β3AR-selective agonist CL 316243 (1 μM), direct activation of G protein with 10 mM NaF or adenylyl cyclase with 100 μM forskolin. Data represent mean±S.E. of at least three separate experiments performed in triplicate.

To determine if there were consequences downstream of βAR activation particular to nuclear function, purified nuclei were incubated with [α32P]-UTP to measure initiation of RNA transcription in response to both non-selective and subtype-selective ligands. In addition to isoproterenol, subtype-selective ligands, xamoterol (β1AR), procaterol (β2AR) and BRL 37344 (β3AR) were used to stimulate transcription. Isoproterenol and BRL 37344, but not xamoterol (Fig. 7a) or procaterol (data not shown), could stimulate initiation of transcription. Since both Gs and Gi were detected on cardiomyocyte nuclear membrane, we next sought to determine the G protein coupling of each receptor. Nuclei were pretreated with PTX for 2 h prior to agonist stimulation in the presence of [α32P]-UTP. For both isoproterenol- and BRL 37344-stimulated nuclei, treatment with PTX abolished initiation of transcription demonstrating that the nuclear β3AR coupling to Gi is responsible for this activity. Further, basal activity was not effected by PTX treatment although the responses to both ligands became inhibitory. It is noteworthy to mention that stimulation of nuclear endothelin receptors induces a similar decrease in basal transcriptional activity (data not shown). This indicates that, under basal conditions, a balance exists between Gi-mediated stimulatory signals and inhibitory signals mediated by other G proteins. This notion is also supported by the observation that forskolin, a direct activator of adenylyl cyclase, tended toward an inhibitory effect (7525±2394 pmol cAMP/mg/min, basal versus 5637.2±1600 pmol cAMP/mg/min, 100 μM forskolin, n=6).

Fig. 7

Functional coupling of β3AR but not β1AR to initiation of de novo transcription in enriched nuclear preparations. (A) Enriched nuclear fractions were stimulated with 1 μM isoproterenol, the β1AR-selective agonist xamoterol (1 μM) or the β3AR-selective agonist BRL 37344. [α32P]-UTP incorporation was measured in membranes either untreated or pretreated with 5 μg/ml PTX for 2 h to inhibit Gi-specific signalling. Significant differences (* for p<0.05 between control and ligand-stimulated) were determined by a paired t test for three or more experiments. (B) Enriched nuclear fractions were stimulated with 1 μM isoproterenol, with and without the β1AR-selective antagonist CGP 20712A (1 nM) or the β2AR-selective antagonist ICI 118,551 (0.5 nM). [α32P]-UTP incorporation was also measured in response to CGP 20712A or ICI 118,551 alone. Data represent mean±S.E. of at least three separate experiments performed in triplicate and are normalized to control. Significant differences (** and * for p<0.05 between vehicle and PTX-treated and control and ligand-stimulated, respectively) were determined by a paired t test for three or more experiments.

To further define the subtype selectivity for the transcriptional response and to overcome difficulties with the use of weak partial agonists such as xamoterol, we use either CGP 20712A or ICI 118,551 to block β1AR and β2AR, respectively. Curiously, we noted that isoproterenol-stimulated transcription initiation was inhibited in both cases (Fig. 7b). There was residual stimulation in the case of CGP 20712A. However, when we used these compounds, in the absence of agonist, CGP 20712A had no effect, consistent with its role as an antagonist. Surprisingly, ICI 118,551 had a strong inhibitory effect on the basal level of transcription initiation by itself. Further, we have shown that ICI 118,551 can compete for CYP binding (data not shown). These data suggest the possibility that levels of β2AR undetectable by immunological methods may be found in the nuclei as well.

Taken together, our data suggest that two effector systems are stimulated by βAR in isolated rat heart nuclei, one coupled to Gs which leads to an activation of adenylyl cyclase and another acting through Gi which leads to an increase in RNA transcription initiation.

4 Discussion

Our results demonstrate functional βAR receptors on the nuclear membrane. By immunological, ligand-binding and functional criteria, these seem to be predominantly β1AR and β3AR. However, it is also possible that the β2AR may be there but undetectable using available antibodies. These receptors were inhibited by ICI 118,551 even in the absence of agonist, i.e. ICI 118,551 is acting as an inverse agonist reducing constitutive receptor activity. CGP 20712 blocked the effects of isoproterenol as well. It has been demonstrated that CGP 20712 can antagonize β3AR [43] and this may explain the effects we have seen here. We have so far considered the effects of stimulation on nuclear βAR as if the receptors were monomeric or homodimeric entities. Alternatively, we [44] and others [45,46] have demonstrated that receptor heterodimers may comprise unique receptors with altered pharmacological properties, i.e. subtype-selective ligands may not be informative. Our results may thus be complicated by the possibility that the β1AR and β3AR may form heterodimers. Both β1AR and β2AR [47,48] and β2AR and β3AR [49] form heterodimeric receptors and it remains to be seen (although likely) if β1AR and β3AR do as well. Other potentially confounding and cell- or even organelle-specific factors might affect the pharmacology, signalling and/or trafficking of homodimeric or heterodimeric GPCRs to different subcellular destinations. As βAR signalling changes during development [50,51], and during the progression to heart failure [52], it is possible that trafficking itineraries on an absolute or relative basis may be altered as well.

β1AR and β3AR also seem to serve different functions in the nuclear membrane, as they are coupled to distinct G protein partners. β3AR stimulation, coupled to a PTX-sensitive G protein, likely Gi, which colocalizes with the receptor to the nuclear membrane, can initiate de novo transcription in isolated nuclei. The β3AR-mediated transcriptional activity is independent of adenylyl cyclase as stimulation with forskolin does not mimic these results. Our studies, being performed in native tissues, confirm that these results are not simply artifacts due to overexpression in heterologous expression systems and suggest a novel role for the β3AR in cardiac function. Also, we verified the existence of these receptors on the nucleus using multiple techniques, not only relying on antibodies but also demonstrating the presence of receptors and functional signalling pathways using ligand binding and by measuring two distinct agonist-mediated events. Further, as has been demonstrated for a number of other GPCRs, not only are the receptors themselves trafficked to the nuclear membrane but in many cases entire signalling pathways are also present.

A recent study that deorphanized GPR30 has demonstrated that this receptor is retained in the ER where it serves as a receptor for estrogen [53]. For other GPCRs such as the PAF or prostaglandin receptors, which are also activated by hydrophobic ligands which can cross the plasma membrane, it is quite possible to envision particular roles for these receptors in intracellular compartments. For receptors with peptidic ligands such as those for angiotensin, bradykinin or endothelin, it is intrinsically easier to imagine how these intracrine signalling pathways might be activated as well. This is because the ligands themselves may be released into intracellular compartments as part of their own independent trafficking itineraries following synthesis. In some cases, as for EGF receptors, a fragment of a cell surface receptor is trafficked to the nucleus after a proteolytic event [54,55]. This is unlikely to be the case here as Western blots of enriched nuclei indicate molecular weights for the β1AR and β3AR consistent with published values for full-length receptors [36,37]. Furthermore, no reduction in molecular weight was observed for the β1AR and β3AR in the nuclear fraction (Fraction 4) compared to total cell extract (Fraction 1). For adrenergic and other small ligand-activated receptors to have intracellular effects, one could consider an uptake mechanism. The classic reuptake system seen in neurons is unlikely to operate in cardiomyocytes. However, there are other non-selective uptake systems such as EMT (extraneuronal monoamine transporter, or OCT3) found in several tissues including heart ([56,57], reviewed in [58]) which could transport catecholamines to intracellular targets. One study using neonatal rat cardiomyocytes determined that extracellular [3H]-norepinephrine is taken up into these cells and >60% is distributed to nuclear fractions [59]. Further, these authors also demonstrate that nuclei isolated from these cells (purity confirmed by electron microscopy) possess approximately 60 fmol/mg βAR (roughly comparable to our values of 15–20 fmol/mg) as determined by ligand binding. It has also become clear in recent years that many GPCRs including βAR have constitutive, agonist-independent activity [60,61]. Depending on local expression levels in a given subcellular fraction, this constitutive activity could be quite important.

A growing number of GPCRs are trafficked to the nuclear or other endomembranes where they can activate signalling pathways that may be intrinsically different than those activated at the plasma membrane. The actual subcellular location of a given receptor may be critical for determining which signalling pathways get activated. Thus decisions about trafficking and formation of signalling complexes with distinct compositions may represent key events in determining the specificity of cellular communication.

Acknowledgements

TEH and BGA currently hold senior scholarships from the Fonds de la Recherche en Santé du Québec (FRSQ). BB was the recipient a studentship from the FRSQ. This work was supported by grants from NSERC, the Heart and Stroke Foundation of Québec and the Canadian Institutes of Health Research (CIHR) to TEH (MOP-36379) and BGA (MOP-64183) and from a CIHR group grant (FRN-12814). We thank Angelo Calderone for the discussion and critical advice. We also thank Céline Fiset and Marie-Andrée Lupien for assistance with isolation of mouse cardiomyocytes.

References

[1]

Gobeil
F.
Jr.
Bernier
S.G.
Vazquez-Tello
A.
Brault
S.
Beauchamp
M.H.
Quiniou
C.
et al. 
Modulation of pro-inflammatory gene expression by nuclear lysophosphatidic acid receptor type-1
J Biol Chem
2003
278
38875
38883

[2]

O'Malley
K.L.
Jong
Y.J.
Gonchar
Y.
Burkhalter
A.
Romano
C.
Activation of metabotropic glutamate receptor mGlu5 on nuclear membranes mediates intranuclear Ca2+ changes in heterologous cell types and neurons
J Biol Chem
2003
278
28210
28219

[3]

Lee
D.K.
Lanca
A.J.
Cheng
R.
Nguyen
T.
Ji
X.D.
Gobeil
F.
Jr.
et al. 
Agonist-independent nuclear localization of the Apelin, angiotensin AT1, and bradykinin B2 receptors
J Biol Chem
2004
279
7901
7908

[4]

Marrache
A.M.
Gobeil
F.
Jr.
Bernier
S.G.
Stankova
J.
Rola-Pleszczynski
M.
Choufani
S.
et al. 
Proinflammatory gene induction by platelet-activating factor mediated via its cognate nuclear receptor
J Immunol
2002
169
6474
6481

[5]

Lu
D.
Yang
H.
Shaw
G.
Raizada
M.K.
Angiotensin II-induced nuclear targeting of the angiotensin type 1 (AT1) receptor in brain neurons
Endocrinology
1998
139
365
375

[6]

Zhuo
J.L.
Imig
J.D.
Hammond
T.G.
Orengo
S.
Benes
E.
Navar
L.G.
Ang II accumulation in rat renal endosomes during Ang II-induced hypertension: role of AT(1) receptor
Hypertension
2002
39
116
121

[7]

Chen
R.
Mukhin
Y.V.
Garnovskaya
M.N.
Thielen
T.E.
Iijima
Y.
Huang
C.
et al. 
A functional angiotensin II receptor-GFP fusion protein: evidence for agonist-dependent nuclear translocation
Am J Physiol Renal Physiol
2000
279
F440
F448

[8]

Gobeil
F.
Jr.
Dumont
I.
Marrache
A.M.
Vazquez-Tello
A.
Bernier
S.G.
Abran
D.
et al. 
Regulation of eNOS expression in brain endothelial cells by perinuclear EP(3) receptors
Circ Res
2002
90
682
689

[9]

Boivin
B.
Chevalier
D.
Villeneuve
L.R.
Rousseau
E.
Allen
B.G.
Functional endothelin receptors are present on nuclei in cardiac ventricular myocytes
J Biol Chem
2003
278
29153
29163

[10]

Zhang
J.H.
Barr
V.A.
Mo
Y.
Rojkova
A.M.
Liu
S.
Simonds
W.F.
Nuclear localization of G protein β5 and regulator of G protein signaling 7 in neurons and brain
J Biol Chem
2001
276
10284
10289

[11]

Boivin
B.
Villeneuve
L.R.
Farhat
N.
Chevalier
D.
Allen
B.G.
Sub-cellular distribution of endothelin signaling pathway components in ventricular myocytes and heart: lack of preformed caveolar signalosomes
J Mol Cell Cardiol
2005
38
665
676

[12]

Willard
F.S.
Crouch
M.F.
Nuclear and cytoskeletal translocation and localization of heterotrimeric G-proteins
Immunol Cell Biol
2000
78
387
394

[13]

Yamamoto
S.
Kawamura
K.
James
T.N.
Intracellular distribution of adenylate cyclase in human cardiocytes determined by electron microscopic cytochemistry
Microsc Res Tech
1998
40
479
487

[14]

Schulze
W.
Buchwalow
I.B.
Adenylyl cyclase in the heart: an enzymocytochemical and immunocytochemical approach
Microsc Res Tech
1998
40
473
478

[15]

Schievella
A.R.
Regier
M.K.
Smith
W.L.
Lin
L.L.
Calcium-mediated translocation of cytosolic phospholipase A2 to the nuclear envelope and endoplasmic reticulum
J Biol Chem
1995
270
30749
30754

[16]

Kim
C.G.
Park
D.
Rhee
S.G.
The role of carboxyl-terminal basic amino acids in Gqa-dependent activation, particulate association, and nuclear localization of phospholipase C-β1
J Biol Chem
1996
271
21187
21192

[17]

Freyberg
Z.
Sweeney
D.
Siddhanta
A.
Bourgoin
S.
Frohman
M.
Shields
D.
Intracellular localization of phospholipase D1 in mammalian cells
Mol Biol Cell
2001
12
943
955

[18]

Burchett
S.A.
In through the out door: nuclear localization of the regulators of G protein signaling
J Neurochem
2003
87
551
559

[19]

Scott
M.G.
Le Rouzic
E.
Perianin
A.
Pierotti
V.
Enslen
H.
Benichou
S.
et al. 
Differential nucleocytoplasmic shuttling of β-arrestins. Characterization of a leucine-rich nuclear export signal in β-arrestin2
J Biol Chem
2002
277
37693
37701

[20]

Wang
P.
Wu
Y.
Ge
X.
Ma
L.
Pei
G.
Subcellular localization of β-arrestins is determined by their intact N domain and the nuclear export signal at the C terminus
J Biol Chem
2003
278
11648
11653

[21]

Yi
X.P.
Gerdes
A.M.
Li
F.
Myocyte redistribution of GRK2 and GRK5 in hypertensive, heart-failure-prone rats
Hypertension
2002
39
1058
1063

[22]

Yi
X.P.
Zhou
J.
Baker
J.
Wang
X.
Gerdes
A.M.
Li
F.
Myocardial expression and redistribution of GRKs in hypertensive hypertrophy and failure
Anat Rec A Discov Mol Cell Evol Biol
2005
282
13
23

[23]

Johnson
L.R.
Scott
M.G.
Pitcher
J.A.
G protein-coupled receptor kinase 5 contains a DNA-binding nuclear localization sequence
Mol Cell Biol
2004
24
10169
10179

[24]

Sastri
M.
Barraclough
D.M.
Carmichael
P.T.
Taylor
S.S.
A-kinase-interacting protein localizes protein kinase A in the nucleus
Proc Natl Acad Sci U S A
2005
102
349
354

[25]

Xiao
R.P.
Zhu
W.
Zheng
M.
Chakir
K.
Bond
R.
Lakatta
E.G.
et al. 
Subtype-specific β-adrenoceptor signaling pathways in the heart and their potential clinical implications
Trends Pharmacol Sci
2004
25
358
365

[26]

Steinberg
S.F.
Brunton
L.L.
Compartmentation of G protein-coupled signaling pathways in cardiac myocytes
Annu Rev Pharmacol Toxicol
2001
41
751
773

[27]

Steinberg
S.F.
β(2)-Adrenergic receptor signaling complexes in cardiomyocyte caveolae/lipid rafts
J Mol Cell Cardiol
2004
37
407
415

[28]

Zhu
W.Z.
Chakir
K.
Zhang
S.
Yang
D.
Lavoie
C.
Bouvier
M.
et al. 
Heterodimerization of β1- and β2-adrenergic receptor subtypes optimizes β-adrenergic modulation of cardiac contractility
Circ Res
2005
9
244
251

[29]

Boivin
B.
Allen
B.G.
Regulation of membrane-bound PKC in adult cardiac ventricular myocytes
Cell Signal
2003
15
217
224

[30]

Fiset
C.
Clark
R.B.
Larsen
T.S.
Giles
W.R.
A rapidly activating sustained K+ current modulates repolarization and excitation–contraction coupling in adult mouse ventricle
J Physiol
1997
504
Pt 3
557
563

[31]

Dawn
B.
Xuan
Y.T.
Marian
M.
Flaherty
M.P.
Murphree
S.S.
Smith
T.L.
et al. 
Cardiac-specific abrogation of NF-κ B activation in mice by transdominant expression of a mutant IκBα
J Mol Cell Cardiol
2001
33
161
173

[32]

Hebert
T.E.
Moffett
S.
Morello
J.P.
Loisel
T.P.
Bichet
D.G.
Barret
C.
et al. 
A peptide derived from a β2-adrenergic receptor transmembrane domain inhibits both receptor dimerization and activation
J Biol Chem
1996
271
16384
16392

[33]

Salomon
Y.
Londos
C.
Rodbell
M.
A highly sensitive adenylate cyclase assay
Anal Biochem
1974
58
541
548

[34]

Xu
J.
Manning
F.C.
O'Brien
T.J.
Ceryak
S.
Patierno
S.R.
Mechanisms of chromium-induced suppression of RNA synthesis in cellular and cell-free systems: relationship to RNA polymerase arrest
Mol Cell Biochem
2004
255
151
160

[35]

Bradford
M.M.
A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding
Anal Biochem
1976
72
248
254

[36]

Rybin
V.O.
Xu
X.
Lisanti
M.P.
Steinberg
S.F.
Differential targeting of β-adrenergic receptor subtypes and adenylyl cyclase to cardiomyocyte caveolae. A mechanism to functionally regulate the cAMP signaling pathway
J Biol Chem
2000
275
41447
41457

[37]

Steinle
J.J.
Booz
G.W.
Meininger
C.J.
Day
J.N.
Granger
H.J.
β3-adrenergic receptors regulate retinal endothelial cell migration and proliferation
J Biol Chem
2003
278
20681
20686

[38]

Ben-Efraim
I.
Gerace
L.
Gradient of increasing affinity of importin b for nucleoporins along the pathway of nuclear import
J Cell Biol
2001
152
411
417

[39]

Chung
C.Y.
Erickson
H.P.
Cell surface annexin II is a high affinity receptor for the alternatively spliced segment of tenascin-C
J Cell Biol
1994
126
539
548

[40]

Haas
I.G.
BiP (GRP78), an essential hsp70 resident protein in the endoplasmic reticulum
Experientia
1994
50
1012
1020

[41]

Stroud
W.J.
Jiang
S.
Jack
G.
Storrie
B.
Persistence of Golgi matrix distribution exhibits the same dependence on Sar1p activity as a Golgi glycosyltransferase
Traffic
2003
4
631
641

[42]

Herget
G.W.
Neuburger
M.
Plagwitz
R.
Adler
C.P.
DNA content, ploidy level and number of nuclei in the human heart after myocardial infarction
Cardiovasc Res
1997
36
45
51

[43]

Kaumann
A.J.
Molenaar
P.
Differences between the third cardiac beta-adrenoceptor and the colonic β3-adrenoceptor in the rat
Br J Pharmacol
1996
118
2085
2098

[44]

Lavoie
C.
Hebert
T.E.
Pharmacological characterization of putative β1-β2-adrenergic receptor heterodimers
Can J Physiol Pharmacol
2003
81
186
195

[45]

Jordan
B.A.
Devi
L.A.
G-protein-coupled receptor heterodimerization modulates receptor function
Nature
1999
399
697
700

[46]

Pfeiffer
M.
Koch
T.
Schroder
H.
Klutzny
M.
Kirscht
S.
Kreienkamp
H.J.
et al. 
Homo- and heterodimerization of somatostatin receptor subtypes. Inactivation of sst(3) receptor function by heterodimerization with sst(2A)
J Biol Chem
2001
276
14027
14036

[47]

Lavoie
C.
Mercier
J.F.
Salahpour
A.
Umapathy
D.
Breit
A.
Villeneuve
L.R.
et al. 
β1/β2-adrenergic receptor heterodimerization regulates β2-adrenergic receptor internalization and ERK signaling efficacy
J Biol Chem
2002
277
35402
35410

[48]

Mercier
J.-F.
Salahpour
A.
Angers
S.
Breit
A.
Bouvier
M.
Quantitative Assessment of β1- and β2-Adrenergic Receptor Homo- and Heterodimerization by Bioluminescence Resonance Energy Transfer
J Biol Chem
2002
277
44925
44931

[49]

Breit
A.
Lagace
M.
Bouvier
M.
Hetero-oligomerization between β2- and β3-adrenergic receptors generates a beta-adrenergic signaling unit with distinct functional properties
J Biol Chem
2004
279
28756
28765

[50]

Rybin
V.O.
Pak
E.
Alcott
S.
Steinberg
S.F.
Developmental changes in β2-adrenergic receptor signaling in ventricular myocytes: the role of Gi proteins and caveolae microdomains
Mol Pharmacol
2003
63
1338
1348

[51]

Steinberg
S.F.
The molecular basis for distinct b-adrenergic receptor subtype actions in cardiomyocytes
Circ Res
1999
85
1101
1111

[52]

Lohse
M.J.
Engelhardt
S.
Eschenhagen
T.
What is the role of b-adrenergic signaling in heart failure?
Circ Res
2003
93
896
906

[53]

Revankar
C.M.
Cimino
D.F.
Sklar
L.A.
Arterburn
J.B.
Prossnitz
E.R.
A transmembrane intracellular estrogen receptor mediates rapid cell signaling
Science
2005
307
1625
1630

[54]

Lin
S.Y.
Makino
K.
Xia
W.
Matin
A.
Wen
Y.
Kwong
K.Y.
et al. 
Nuclear localization of EGF receptor and its potential new role as a transcription factor
Nat Cell Biol
2001
3
802
808

[55]

Wells
A.
Marti
U.
Signalling shortcuts: cell-surface receptors in the nucleus?
Nat Rev Mol Cell Biol
2002
3
697
702

[56]

Eisenhofer
G.
The role of neuronal and extraneuronal plasma membrane transporters in the inactivation of peripheral catecholamines
Pharmacol Ther
2001
91
35
62

[57]

Grundemann
D.
Schechinger
B.
Rappold
G.A.
Schomig
E.
Molecular identification of the corticosterone-sensitive extraneuronal catecholamine transporter
Nat Neurosci
1998
1
349
351

[58]

Jonker
J.W.
Schinkel
A.H.
Pharmacological and physiological functions of the polyspecific organic cation transporters: OCT1, 2, and 3 (SLC22A1-3)
J Pharmacol Exp Ther
2004
308
2
9

[59]

Buu
N.T.
Hui
R.
Falardeau
P.
Norepinephrine in neonatal rat ventricular myocytes: association with the cell nucleus and binding to nuclear α1- and β-adrenergic receptors
J Mol Cell Cardiol
1993
25
1037
1046

[60]

Chidiac
P.
Hebert
T.E.
Valiquette
M.
Dennis
M.
Bouvier
M.
Inverse agonist activity of b-adrenergic antagonists
Mol Pharmacol
1994
45
490
499

[61]

Samama
P.
Pei
G.
Costa
T.
Cotecchia
S.
Lefkowitz
R.J.
Negative antagonists promote an inactive conformation of the β2-adrenergic receptor
Mol Pharmacol
1994
45
390
394

Author notes

Time for primary review 24 days