Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 336(2): 524-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21071556

ABSTRACT

Astrocytomas and glioblastomas have been particularly difficult to treat and refractory to chemotherapy. However, significant evidence has been presented that demonstrates a decrease in astrocytoma cell proliferation subsequent to an increase in cAMP levels. The 1321N1 astrocytoma cell line, as well as other astrocytomas and glioblastomas, expresses ß(2)-adrenergic receptors (ß(2)-ARs) that are coupled to G(s) activation and consequent cAMP production. Experiments were conducted to determine whether the ß(2)-AR agonist (R,R')-fenoterol and other ß(2)-AR agonists could attenuate mitogenesis and, if so, by what mechanism. Receptor binding studies were conducted to characterize ß(2)-AR found in 1321N1 and U118 cell membranes. In addition, cells were incubated with (R,R')-fenoterol and analogs to determine their ability to stimulate intracellular cAMP accumulation and inhibit [(3)H]thymidine incorporation into the cells. 1321N1 cells contain significant levels of ß(2)-AR as determined by receptor binding. (R,R')-fenoterol and other ß(2)-AR agonists, as well as forskolin, stimulated cAMP accumulation in a dose-dependent manner. Accumulation of cAMP induced a decrease in [(3)H]thymidine incorporation. There was a correlation between concentration required to stimulate cAMP accumulation and inhibit [(3)H]thymidine incorporation. U118 cells have a reduced number of ß(2)-ARs and a concomitant reduction in the ability of ß(2)-AR agonists to inhibit cell proliferation. These studies demonstrate the efficacy of ß(2)-AR agonists for inhibition of growth of the astrocytoma cell lines. Because a significant portion of brain tumors contain ß(2)-ARs to a greater extent than whole brain, (R,R')-fenoterol, or some analog, may be useful in the treatment of brain tumors after biopsy to determine ß(2)-AR expression.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Astrocytoma/drug therapy , Astrocytoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Fenoterol/pharmacology , G1 Phase/drug effects , Humans , Propanolamines/metabolism , Thymidine/metabolism
3.
Sci STKE ; 2001(104): re15, 2001 Oct 16.
Article in English | MEDLINE | ID: mdl-11604549

ABSTRACT

Beta-adrenergic receptor (AR) subtypes are archetypical members of the G protein-coupled receptor (GPCR) superfamily. Whereas both beta1AR and beta2AR stimulate the classic G(s)-adenylyl cyclase-3',5'-adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling cascade, beta2AR couples to both G(s) and G(i) proteins, activating bifurcated signaling pathways. In the heart, dual coupling of the beta2AR to G(s) and G(i) results in compartmentalization of the G(s)-stimulated cAMP signal, thus selectively affecting plasma membrane effectors (such as L-type Ca(2+) channels) and bypassing cytoplasmic target proteins (such as phospholamban and myofilament contractile proteins). More important, the beta2AR-to-G(i) branch delivers a powerful cell survival signal that counters apoptosis induced by the concurrent G(s)-mediated signal or by a wide range of assaulting factors. This survival pathway sequentially involves G(i), G(beta)(gamma), phosphoinositide 3-kinase, and Akt. Furthermore, cardiac-specific transgenic overexpression of betaAR subtypes in mice results in distinctly different phenotypes in terms of the likelihood of cardiac hypertrophy and heart failure. These findings indicate that stimulation of the two betaAR subtypes activates overlapping, but different, sets of signal transduction mechanisms, and fulfills distinct or even opposing physiological and pathophysiological roles. Because of these differences, selective activation of cardiac beta2AR may provide catecholamine-dependent inotropic support without cardiotoxic consequences, which might have beneficial effects in the failing heart.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Myocardium/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Cardiomegaly/etiology , Cell Survival , Cyclic AMP/physiology , Heart/embryology , Heart Failure/drug therapy , Heart Failure/etiology , Humans , Mice , Models, Biological , Rats , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 98(21): 12283-8, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11593045

ABSTRACT

Stress-induced mitogen-activated protein kinase (MAP) p38 is activated in various forms of heart failure, yet its effects on the intact heart remain to be established. Targeted activation of p38 MAP kinase in ventricular myocytes was achieved in vivo by using a gene-switch transgenic strategy with activated mutants of upstream kinases MKK3bE and MKK6bE. Transgene expression resulted in significant induction of p38 kinase activity and premature death at 7-9 weeks. Both groups of transgenic hearts exhibited marked interstitial fibrosis and expression of fetal marker genes characteristic of cardiac failure, but no significant hypertrophy at the organ level. Echocardiographic and pressure-volume analyses revealed a similar extent of systolic contractile depression and restrictive diastolic abnormalities related to markedly increased passive chamber stiffness. However, MKK3bE-expressing hearts had increased end-systolic chamber volumes and a thinned ventricular wall, associated with heterogeneous myocyte atrophy, whereas MKK6bE hearts had reduced end-diastolic ventricular cavity size, a modest increase in myocyte size, and no significant myocyte atrophy. These data provide in vivo evidence for a negative inotropic and restrictive diastolic effect from p38 MAP kinase activation in ventricular myocytes and reveal specific roles of p38 pathway in the development of ventricular end-systolic remodeling.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomyopathy, Restrictive/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Ventricular Remodeling , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cardiomyopathy, Restrictive/metabolism , Cardiotonic Agents , Cells, Cultured , Gene Expression , Gene Targeting , Heart Ventricles/cytology , Hemodynamics , Humans , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Rats , p38 Mitogen-Activated Protein Kinases
5.
Circ Res ; 88(8): 794-801, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11325871

ABSTRACT

To elucidate microscopic mechanisms underlying the modulation of cardiac excitation-contraction (EC) coupling by beta-adrenergic receptor (beta-AR) stimulation, we examined local Ca(2+) release function, ie, Ca(2+) spikes at individual transverse tubule-sarcoplasmic reticulum (T-SR) junctions, using confocal microscopy and our recently developed technique for release flux measurement. beta-AR stimulation by norepinephrine plus an alpha(1)-adrenergic blocker, prazosin, increased the amplitude of SR Ca(2+) release flux (J(SR)), its running integral (integralJ(SR)), and L-type Ca(2+) channel current (I(Ca)), and it shifted their bell-shaped voltage dependence leftward by approximately 10 mV, with the relative effects ranking I(Ca)> J(SR)>integralJ(SR). Confocal imaging revealed that the bell-shaped voltage dependence of SR Ca(2+) release is attributable to a graded recruitment of T-SR junctions as well as to changes in Ca(2+) spike amplitudes. beta-AR stimulation increased the fractional T-SR junctions that fired Ca(2+) spikes and augmented Ca(2+) spike amplitudes, without altering the SR Ca(2+) load, suggesting that more release units were activated synchronously among and within T-SR junctions. Moreover, beta-AR stimulation decreased the latency and temporal dispersion of Ca(2+) spike occurrence at a given voltage, delivering most of the Ca(2+) at the onset of depolarization rather than spreading it out throughout depolarization. Because the synchrony of Ca(2+) spikes affects Ca(2+) delivery per unit of time to contractile myofilaments, and because the myofilaments display a steep Ca(2+) dependence, our data suggest that synchronization of SR Ca(2+) release represents a heretofore unappreciated mechanism of beta-AR modulation of cardiac inotropy.


Subject(s)
Calcium/metabolism , Intracellular Fluid/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Receptors, Adrenergic, beta/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Adrenergic alpha-1 Receptor Antagonists , Animals , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cell Separation , Fluorescent Dyes , Myocardial Contraction/drug effects , Myocardium/cytology , Norepinephrine/pharmacology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Vasoconstrictor Agents/pharmacology
6.
Proc Natl Acad Sci U S A ; 98(4): 1607-12, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11171998

ABSTRACT

The goal of this study was to determine whether beta(1)-adrenergic receptor (AR) and beta(2)-AR differ in regulating cardiomyocyte survival and apoptosis and, if so, to explore underlying mechanisms. One potential mechanism is that cardiac beta(2)-AR can activate both G(s) and G(i) proteins, whereas cardiac beta(1)-AR couples only to G(s). To avoid complicated crosstalk between beta-AR subtypes, we expressed beta(1)-AR or beta(2)-AR individually in adult beta(1)/beta(2)-AR double knockout mouse cardiac myocytes by using adenoviral gene transfer. Stimulation of beta(1)-AR, but not beta(2)-AR, markedly induced myocyte apoptosis, as indicated by increased terminal deoxynucleotidyltransferase-mediated UTP end labeling or Hoechst staining positive cells and DNA fragmentation. In contrast, beta(2)-AR (but not beta(1)-AR) stimulation elevated the activity of Akt, a powerful survival signal; this effect was fully abolished by inhibiting G(i), G(beta gamma), or phosphoinositide 3 kinase (PI3K) with pertussis toxin, beta ARK-ct (a peptide inhibitor of G(beta gamma)), or LY294002, respectively. This indicates that beta(2)-AR activates Akt via a G(i)-G(beta gamma)-PI3K pathway. More importantly, inhibition of the G(i)-G(beta gamma)-PI3K-Akt pathway converts beta(2)-AR signaling from survival to apoptotic. Thus, stimulation of a single class of receptors, beta(2)-ARs, elicits concurrent apoptotic and survival signals in cardiac myocytes. The survival effect appears to predominate and is mediated by the G(i)-G(beta gamma)-PI3K-Akt signaling pathway.


Subject(s)
Apoptosis , Myocardium/cytology , Protein Serine-Threonine Kinases , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Animals , Cell Survival , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heterotrimeric GTP-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , p38 Mitogen-Activated Protein Kinases
7.
J Biol Chem ; 275(51): 40635-40, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11018034

ABSTRACT

Increasing evidence shows that stimulation of beta-adrenergic receptor (AR) activates mitogen-activated protein kinases (MAPKs), in addition to the classical G(s)-adenylyl cyclase-cAMP-dependent protein kinase (PKA) signaling cascade. In the present study, we demonstrate a novel beta(2)-AR-mediated cross-talk between PKA and p38 MAPK in adult mouse cardiac myocytes expressing beta(2)-AR, with a null background of beta(1)beta(2)-AR double knockout. beta(2)-AR stimulation by isoproterenol increased p38 MAPK activity in a time- and dose-dependent manner. Inhibiting G(i) with pertussis toxin or scavenging Gbetagamma with betaARK-ct overexpression could not prevent beta(2)-AR-induced p38 MAPK activation. In contrast, a specific peptide inhibitor of PKA, PKI (5 microm), completely abolished the stimulatory effect of beta(2)-AR, suggesting that beta(2)-AR-induced p38 MAPK activation is mediated via a PKA-dependent mechanism, rather than by G(i) or Gbetagamma. This conclusion was further supported by the ability of forskolin (10 microm), an adenylyl cyclase activator, to elevate p38 MAPK activity in a PKI-sensitive manner. Furthermore, inhibition of p38 MAPK with SB203580 (10 microm) markedly enhanced the beta(2)-AR-mediated contractile response, without altering base-line contractility. These results provide the first evidence that cardiac beta(2)-AR activates p38 MAPK via a PKA-dependent signaling pathway, rather than by G(i) or Gbetagamma, and reveal a novel role of p38 MAPK in regulating cardiac contractility.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myocardium/enzymology , Receptors, Adrenergic, beta-2/physiology , Animals , Cells, Cultured , Enzyme Activation , Mice , Myocardium/cytology , Myocardium/metabolism , p38 Mitogen-Activated Protein Kinases
8.
Biophys J ; 79(5): 2547-56, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053129

ABSTRACT

A plausible determinant of the specificity of receptor signaling is the cellular compartment over which the signal is broadcast. In rat heart, stimulation of beta(1)-adrenergic receptor (beta(1)-AR), coupled to G(s)-protein, or beta(2)-AR, coupled to G(s)- and G(i)-proteins, both increase L-type Ca(2+) current, causing enhanced contractile strength. But only beta(1)-AR stimulation increases the phosphorylation of phospholamban, troponin-I, and C-protein, causing accelerated muscle relaxation and reduced myofilament sensitivity to Ca(2+). beta(2)-AR stimulation does not affect any of these intracellular proteins. We hypothesized that beta(2)-AR signaling might be localized to the cell membrane. Thus we examined the spatial range and characteristics of beta(1)-AR and beta(2)-AR signaling on their common effector, L-type Ca(2+) channels. Using the cell-attached patch-clamp technique, we show that stimulation of beta(1)-AR or beta(2)-AR in the patch membrane, by adding agonist into patch pipette, both activated the channels in the patch. But when the agonist was applied to the membrane outside the patch pipette, only beta(1)-AR stimulation activated the channels. Thus, beta(1)-AR signaling to the channels is diffusive through cytosol, whereas beta(2)-AR signaling is localized to the cell membrane. Furthermore, activation of G(i) is essential to the localization of beta(2)-AR signaling because in pertussis toxin-treated cells, beta(2)-AR signaling becomes diffusive. Our results suggest that the dual coupling of beta(2)-AR to both G(s)- and G(i)-proteins leads to a highly localized beta(2)-AR signaling pathway to modulate sarcolemmal L-type Ca(2+) channels in rat ventricular myocytes.


Subject(s)
Calcium Channels, L-Type/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Biophysical Phenomena , Biophysics , In Vitro Techniques , Myocardium/metabolism , Patch-Clamp Techniques , Phosphorylation , Rats , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction
9.
Circ Res ; 87(9): 760-7, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11055979

ABSTRACT

Cardiac beating arises from the spontaneous rhythmic excitation of sinoatrial (SA) node cells. Here we report that SA node pacemaker activity is critically dependent on Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). In freshly dissociated rabbit single SA node cells, inhibition of CaMKII by a specific peptide inhibitor, autocamtide-2 inhibitory peptide (AIP, 10 micromol/L), or by KN-93 (0.1 to 3.0 micromol/L), but not its inactive analog, KN-92, depressed the rate and amplitude of spontaneous action potentials (APs) in a dose-dependent manner. Strikingly, 10 micromol/L AIP and 3 micromol/L KN-93 completely arrested SA node cells, which indicates that basal CaMKII activation is obligatory to the genesis of pacemaker AP. To understand the ionic mechanisms of the CaMKII effects, we measured L-type Ca(2+) current (I(Ca, L)), which contributes both to AP upstroke and to pacemaker depolarization. KN-93 (1 micromol/L), but not its inactive analog, KN-92, decreased I:(Ca, L) amplitude from 12+/-2 to 6+/-1 pA/pF without altering the shape of the current-voltage relationship. Both AIP and KN-93 shifted the midpoint of the steady-state inactivation curve leftward and markedly slowed the recovery of I(Ca, L) from inactivation. Similar results were observed using the fast Ca(2+) chelator BAPTA, whereas the slow Ca(2+) chelator EGTA had no significant effect, which suggests that CaMKII activity is preferentially regulated by local Ca(2+) transients. Indeed, confocal immunocytochemical imaging showed that active CaMKII is highly localized beneath the surface membrane in the vicinity of L-type channels and that AIP and KN-93 significantly reduced CaMKII activity. Thus, we conclude that CaMKII plays a vital role in regulating cardiac pacemaker activity mainly via modulating I(Ca, L) inactivation and reactivation, and local Ca(2+) is critically involved in these processes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Egtazic Acid/analogs & derivatives , Heart Rate/physiology , Sinoatrial Node/physiology , Action Potentials/drug effects , Animals , Benzylamines/pharmacology , Calcium/chemistry , Calcium/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Egtazic Acid/chemistry , Enzyme Activation , Enzyme Inhibitors/pharmacology , Immunohistochemistry , In Vitro Techniques , Lysine/pharmacology , Microscopy, Confocal , Nifedipine/pharmacology , Nitrobenzenes/pharmacology , Patch-Clamp Techniques , Rabbits , Sinoatrial Node/cytology , Sulfonamides/pharmacology
11.
Mol Pharmacol ; 58(5): 887-94, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11040034

ABSTRACT

Although ligand-free, constitutive beta(2)-adrenergic receptor (AR) signaling has been demonstrated in naive cell lines and in transgenic mice overexpressing cardiac beta(2)-AR, it is unclear whether the dominant cardiac beta-AR subtype, beta(1)-AR, shares the ability of spontaneous activation. In the present study, we expressed human beta(1)- or beta(2)-AR via recombinant adenoviral infection in ventricular myocytes isolated from beta(1)beta(2)-AR double knockout mice, creating pure beta(1)-AR and beta(2)-AR systems with variable receptor densities. A contractile response to a nonselective beta-AR agonist, isoproterenol, was absent in double knockout mouse myocytes but was fully restored after adenoviral beta(1)-AR or adenoviral beta(2)-AR infection. Increasing the titer of adenoviral vectors (multiplicity of infection 10-1000) led to a dose-dependent expression of beta(1)- or beta(2)-AR with a maximal density of 1207 +/- 173 (36-fold over the wild-type control value) and 821+/-38 fmol/mg protein (69-fold), respectively. Using confocal immunohistochemistry, we directly visualized the cellular distribution of beta(1)-AR and beta(2)-AR and found that both subtypes were distributed on the cell surface membrane and transverse tubules, resulting in a striated pattern. In the absence of ligand, beta(2)-AR expression resulted in graded increases in baseline cAMP and contractility up to 428% and 233% of control, respectively, at the maximal beta(2)-AR density. These effects were specifically reversed by a beta(2)-AR inverse agonist, ICI 118,551 (10(-7) M). In contrast, overexpression of beta(1)-AR, even at a greater density, failed to enhance either basal cAMP or contractility; the alleged beta(1)-AR inverse agonist, CGP 20712A (10(-6) M), had no significant effect on basal contraction in these cells. Thus, we conclude that acute beta(2)-AR overexpression in cardiac myocytes elicits significant physiological responses due to spontaneous receptor activation; however, this property is beta-AR subtype specific because beta(1)-AR does not exhibit agonist-independent spontaneous activation.


Subject(s)
Myocardium/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Adrenergic Agonists/pharmacology , Animals , Heart Ventricles/cytology , Heart Ventricles/metabolism , Mice , Mice, Knockout , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics
12.
Am J Physiol Heart Circ Physiol ; 279(1): H429-36, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10899083

ABSTRACT

Rapid development of transgenic and gene-targeted mice and acute genetic manipulation via gene transfer vector systems have provided powerful tools for cardiovascular research. To facilitate the phenotyping of genetically engineered murine models at the cellular and subcellular levels and to implement acute gene transfer techniques in single mouse cardiomyocytes, we have modified and improved current enzymatic methods to isolate a high yield of high-quality adult mouse myocytes (5.3 +/- 0.5 x 10(5) cells/left ventricle, 83.8 +/- 2.5% rod shaped). We have also developed a technique to culture these isolated myocytes while maintaining their morphological integrity for 2-3 days. The high percentage of viable myocytes after 1 day in culture (72.5 +/- 2.3%) permitted both physiological and biochemical characterization. The major functional aspects of these cells, including excitation-contraction coupling and receptor-mediated signaling, remained intact, but the contraction kinetics were significantly slowed. Furthermore, gene delivery via recombinant adenoviral infection was highly efficient and reproducible. In adult beta(1)/beta(2)-adrenergic receptor (AR) double-knockout mouse myocytes, adenovirus-directed expression of either beta(1)- or beta(2)-AR, which occurred in 100% of cells, rescued the functional response to beta-AR agonist stimulation. These techniques will permit novel experimental settings for cellular genetic physiology.


Subject(s)
Adenoviridae , Gene Transfer Techniques , Myocardium/cytology , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Transfection/methods , Animals , Calcium/metabolism , Cell Culture Techniques/methods , Cell Membrane/physiology , Cells, Cultured , Female , Heart Ventricles , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Mice, Knockout , Myocardium/metabolism , Receptors, Adrenergic, beta-1/deficiency , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/deficiency , Receptors, Adrenergic, beta-2/physiology
13.
J Biol Chem ; 275(29): 22532-6, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10825152

ABSTRACT

Both Ser(16) and Thr(17) of phospholamban (PLB) are phosphorylated, respectively, by cAMP-dependent protein kinase (PKA) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). PLB phosphorylation relieves cardiac sarcoplasmic reticulum Ca(2+) pump from inhibition by PLB. Previous studies have suggested that phosphorylation of Ser(16) by PKA is a prerequisite for Thr(17) phosphorylation by CaMKII and is essential to the relaxant effect of beta-adrenergic stimulation. To determine the role of Thr(17) PLB phosphorylation, we investigated the dual-site phosphorylation of PLB in isolated adult rat cardiac myocytes in response to beta(1)-adrenergic stimulation or electrical field stimulation (0. 1-3 Hz) or both. A beta(1)-adrenergic agonist, norepinephrine (10(-9)-10(-6) m), in the presence of an alpha(1)-adrenergic antagonist, prazosin (10(-6) m), selectively increases the PKA-dependent phosphorylation of PLB at Ser(16) in quiescent myocytes. In contrast, electrical pacing induces an opposite phosphorylation pattern, selectively enhancing the CaMKII-mediated Thr(17) PLB phosphorylation in a frequency-dependent manner. When combined, electric stimulation (2 Hz) and beta(1)-adrenergic stimulation lead to dual phosphorylation of PLB and exert a synergistic effect on phosphorylation of Thr(17) but not Ser(16). Frequency-dependent Thr(17) phosphorylation is closely correlated with a decrease in 50% relaxation time (t(50)) of cell contraction, which is independent of, but additive to, the relaxant effect of Ser(16) phosphorylation, resulting in hastened contractile relaxation at high stimulation frequencies. Thus, we conclude that in intact cardiac myocytes, phosphorylation of PLB at Thr(17) occurs in the absence of prior Ser(16) phosphorylation, and that frequencydependent Thr(17) PLB phosphorylation may provide an intrinsic mechanism for cardiac myocytes to adapt to a sudden change of heart rate.


Subject(s)
Calcium-Binding Proteins/metabolism , Myocardium/metabolism , Adenosine Triphosphatases/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Phosphorylation , Rats , Serine , Threonine
14.
J Biol Chem ; 275(28): 21773-9, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10787424

ABSTRACT

Cardiac-specific overexpression of the human beta(2)-adrenergic receptor (AR) in transgenic mice (TG4) enhances basal cardiac function due to ligand-independent spontaneous beta(2)-AR activation. However, agonist-mediated stimulation of either beta(1)-AR or beta(2)-AR fails to further enhance contractility in TG4 ventricular myocytes. Although the lack of beta(2)-AR response has been ascribed to an efficient coupling of the receptor to pertussis toxin-sensitive G(i) proteins in addition to G(s), the contractile response to beta(1)-AR stimulation by norepinephrine and an alpha(1)-adrenergic antagonist prazosin is not restored by pertussis toxin treatment despite a G(i) protein elevation of 1.7-fold in TG4 hearts. Since beta-adrenergic receptor kinase, betaARK1, activity remains unaltered, the unresponsiveness of beta(1)-AR is not caused by betaARK1-mediated receptor desensitization. In contrast, pre-incubation of cells with anti-adrenergic reagents such as muscarinic receptor agonist, carbachol (10(-5)m), or a beta(2)-AR inverse agonist, ICI 118,551 (5 x 10(-7)m), to abolish spontaneous beta(2)-AR signaling, both reduce the base-line cAMP and contractility and, surprisingly, restore the beta(1)-AR contractile response. The "rescued" contractile response is completely reversed by a beta(1)-AR antagonist, CGP 20712A. Furthermore, these results from the transgenic animals are corroborated by in vitro acute gene manipulation in cultured wild type adult mouse ventricular myocytes. Adenovirus-directed overexpression of the human beta(2)-AR results in elevated base-line cAMP and contraction associated with a marked attenuation of beta(1)-AR response; carbachol pretreatment fully revives the diminished beta(1)-AR contractile response. Thus, we conclude that constitutive beta(2)-AR activation induces a heterologous desensitization of beta(1)-ARs independent of betaARK1 and G(i) proteins; suppression of the constitutive beta(2)-AR signaling by either a beta(2)-AR inverse agonist or stimulation of the muscarinic receptor rescues the beta(1)-ARs from desensitization, permitting agonist-induced contractile response.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Heart/physiology , Myocardial Contraction/drug effects , Myocardium/cytology , Propanolamines/pharmacology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Animals , Carbachol/pharmacology , Cells, Cultured , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , G-Protein-Coupled Receptor Kinase 2 , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Heart Ventricles , Humans , Mice , Norepinephrine/pharmacology , Pertussis Toxin , Prazosin/pharmacology , Receptors, Adrenergic, beta-2/genetics , Recombinant Proteins/metabolism , Transfection , Virulence Factors, Bordetella/pharmacology , beta-Adrenergic Receptor Kinases
15.
Circ Res ; 87(12): 1172-9, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11110775

ABSTRACT

Recent studies have shown that chronic beta-adrenergic receptor (beta-AR) stimulation alters cardiac myocyte survival in a receptor subtype-specific manner. We examined the effect of selective beta(1)- and beta(2)-AR subtype stimulation on apoptosis induced by hypoxia or H(2)O(2) in rat neonatal cardiac myocytes. Although neither beta(1)- nor beta(2)-AR stimulation had any significant effect on the basal level of apoptosis, selective beta(2)-AR stimulation protected myocytes from apoptosis. beta(2)-AR stimulation markedly increased mitogen-activated protein kinase/extracellular signal-regulated protein kinase (MAPK/ERK) activation as well as phosphatidylinositol-3'-kinase (PI-3K) activity and Akt/protein kinase B phosphorylation. beta(1)-AR stimulation also markedly increased MAPK/ERK activation but only minimally activated PI-3K and Akt. Pretreatment with pertussis toxin blocked beta(2)-AR-mediated protection from apoptosis as well as the beta(2)-AR-stimulated changes in MAPK/ERK, PI-3K, and Akt/protein kinase B. The selective PI-3K inhibitor, LY 294002, also blocked beta(2)-AR-mediated protection, whereas inhibition of MAPK/ERK activation at an inhibitor concentration that blocked agonist-induced activation but not the basal level of activation had no effect on beta(2)-AR-mediated protection. These findings demonstrate that beta(2)-ARs activate a PI-3K-dependent, pertussis toxin-sensitive signaling pathway in cardiac myocytes that is required for protection from apoptosis-inducing stimuli often associated with ischemic stress.


Subject(s)
Apoptosis/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Adrenergic, beta-2/physiology , Cells, Cultured , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Myocardium/enzymology , Pertussis Toxin , Receptors, Adrenergic, beta-1/physiology , Signal Transduction , Virulence Factors, Bordetella/pharmacology
16.
J Physiol ; 521 Pt 2: 351-61, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10581307

ABSTRACT

1. Transgenic overexpression of the beta2-adrenergic receptor (beta2AR) in mouse heart augments baseline cardiac function in a ligand-independent manner, due to the presence of spontaneously active beta2AR (beta2AR*). This study aims to elucidate the mechanism of beta2AR*-mediated modulation of cardiac excitation-contraction (EC) coupling. 2. Confocal imaging was used to analyse Ca2+ sparks and spatially resolve Ca2+ transients in single ventricular myocytes from transgenic (TG4) and non-transgenic (NTG) littermates. Whole-cell voltage- and current-clamp techniques were used to record L-type Ca2+ currents (ICa) and action potentials, respectively. 3. In the absence of any beta2AR ligand, TG4 myocytes had greater contraction amplitudes, larger Ca2+ transients and faster relaxation times than did NTG cells. 4. The action potentials of TG4 and NTG myocytes were similar, except for a prolonged end-stage repolarization in TG4 cells; the ICa density and kinetics were nearly identical. The relationship between peak Ca2+ and contraction, which reflects myofilament Ca2+ sensitivity, was similar. 5. In TG4 cells, the frequency of Ca2+ sparks (spontaneous or evoked at -40 mV) was 2-7 times greater, despite the absence of change in the resting Ca2+, sarcoplasmic reticulum (SR) Ca2+ content, and ICa. Individual sparks were brighter, broader and lasted longer, leading to a 2.3-fold greater signal mass. Thus, changes in both spark frequency and size underlie the greater Ca2+ transient in TG4 cells. 6. The inverse agonist ICI 118,551 (ICI, 5 x 10-7 M), which blocks spontaneous beta2AR activation, reversed the aforementioned beta2AR* effects on cardiac EC coupling without affecting the sarcolemmal ICa. However, ICI failed to detect significant constitutive beta2AR activity in NTG cells. 7. We conclude that beta2AR*-mediated signalling enhances SR release channel activity and Ca2+-induced Ca2+ release in TG4 cardiac myocytes, and that beta2AR* enhances EC coupling by reinforcing SR Ca2+ cycling (release and reuptake), but bypassing the sarcolemmal ICa.


Subject(s)
Heart/physiology , Myocardial Contraction/physiology , Receptors, Adrenergic, beta-2/physiology , Sarcoplasmic Reticulum/physiology , Signal Transduction/physiology , Action Potentials/physiology , Aniline Compounds , Animals , Calcium/metabolism , Fluorescent Dyes , Heart Ventricles/chemistry , Heart Ventricles/cytology , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/metabolism , Myocardium/chemistry , Myocardium/cytology , Myocardium/metabolism , Ventricular Function , Xanthenes
17.
Circ Res ; 85(11): 1092-100, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10571541

ABSTRACT

Recent studies have added complexities to the conceptual framework of cardiac beta-adrenergic receptor (beta-AR) signal transduction. Whereas the classical linear G(s)-adenylyl cyclase-cAMP-protein kinase A (PKA) signaling cascade has been corroborated for beta(1)-AR stimulation, the beta(2)-AR signaling pathway bifurcates at the very first postreceptor step, the G protein level. In addition to G(s), beta(2)-AR couples to pertussis toxin-sensitive G(i) proteins, G(i2) and G(i3). The coupling of beta(2)-AR to G(i) proteins mediates, to a large extent, the differential actions of the beta-AR subtypes on cardiac Ca(2+) handling, contractility, cAMP accumulation, and PKA-mediated protein phosphorylation. The extent of G(i) coupling in ventricular myocytes appears to be the basis of the substantial species-to-species diversity in beta(2)-AR-mediated cardiac responses. There is an apparent dissociation of beta(2)-AR-induced augmentations of the intracellular Ca(2+) (Ca(i)) transient and contractility from cAMP production and PKA-dependent cytoplasmic protein phosphorylation. This can be largely explained by G(i)-dependent functional compartmentalization of the beta(2)-AR-directed cAMP/PKA signaling to the sarcolemmal microdomain. This compartmentalization allows the common second messenger, cAMP, to perform selective functions during beta-AR subtype stimulation. Emerging evidence also points to distinctly different roles of these beta-AR subtypes in modulating noncontractile cellular processes. These recent findings not only reveal the diversity and specificity of beta-AR and G protein interactions but also provide new insights for understanding the differential regulation and functionality of beta-AR subtypes in healthy and diseased hearts.


Subject(s)
Heart/physiology , Muscle Proteins/physiology , Myocardial Contraction/physiology , Myocardium/metabolism , Receptors, Adrenergic, beta-2/physiology , Signal Transduction/physiology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Calcium/physiology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Dogs , GTP-Binding Proteins/physiology , Heart/drug effects , Heart Failure/metabolism , Humans , Hydrogen-Ion Concentration , Mice , Mice, Transgenic , Muscle Proteins/drug effects , Myocardial Contraction/drug effects , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/classification , Receptors, Adrenergic, beta-2/drug effects , Signal Transduction/drug effects , Species Specificity
18.
Mol Pharmacol ; 56(3): 485-93, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10462536

ABSTRACT

A receptor can be activated either by specific ligand-directed changes in conformation or by intrinsic, spontaneous conformational change. In the beta(2)-adrenergic receptor (AR) overexpression transgenic (TG4) murine heart, spontaneously activated beta(2)AR (beta(2)-R*) in the absence of ligands has been evidenced by elevated basal adenylyl cyclase activity and cardiac function. In the present study, we determined whether the signaling mediated by beta(2)-R* differs from that of a ligand-elicited beta(2)AR activation (beta(2)-LR*). In ventricular myocytes from TG4 mice, the properties of L-type Ca(2+) current (I(Ca)), a major effector of beta(2)-LR* signaling, was unaltered, despite a 2.5-fold increase in the basal cAMP level and a 1.9-fold increase in baseline contraction amplitude as compared with that of wild-type (WT) cells. Although the contractile response to beta(2)-R* in TG4 cells was abolished by a beta(2)AR inverse agonist, ICI118,551 (5 x 10(-7) M), or an inhibitory cAMP analog, Rp-CPT-cAMPS (10(-4) M), no change was detected in the simultaneously recorded I(Ca). These results suggest that the increase in basal cAMP due to beta(2)-R*, while increasing contraction amplitude, does not affect I(Ca) characteristics. In contrast, the beta(2)AR agonist, zinterol elicited a substantial augmentation of I(Ca) in both TG4 and WT cells (pertussis toxin-treated), indicating that L-type Ca(2+) channel in these cells can respond to ligand-directed signaling. Furthermore, forskolin, an adenylyl cyclase activator, elicited similar dose-dependent increase in I(Ca) amplitude in WT and TG4 cells, suggesting that the sensitivity of L-type Ca(2+) channel to cAMP-dependent modulation remains intact in TG4 cells. Thus, we conclude that beta(2)-R* bypasses I(Ca) to modulate contraction, and that beta(2)-LR* and beta(2)-R* exhibit different intracellular signaling and target protein specificity.


Subject(s)
Calcium Channels/metabolism , Myocardium/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Adenylate Cyclase Toxin , Animals , Calcium Channels, L-Type , Heart Ventricles/drug effects , In Vitro Techniques , Mice , Myocardial Contraction/drug effects , Pertussis Toxin , Protein Conformation , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/physiology , Ventricular Function , Virulence Factors, Bordetella/pharmacology
19.
J Biol Chem ; 274(31): 22048-52, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419531

ABSTRACT

In contrast to beta(1)-adrenoreceptor (beta(1)-AR) signaling, beta(2)-AR stimulation in cardiomyocytes augments L-type Ca(2+) current in a cAMP-dependent protein kinase (PKA)-dependent manner but fails to phosphorylate phospholamban, indicating that the beta(2)-AR-induced cAMP/PKA signaling is highly localized. Here we show that inhibition of G(i) proteins with pertussis toxin (PTX) permits a full phospholamban phosphorylation and a de novo relaxant effect following beta(2)-AR stimulation, converting the localized beta(2)-AR signaling to a global signaling mode similar to that of beta(1)-AR. Thus, beta(2)-AR-mediated G(i) activation constricts the cAMP signaling to the sarcolemma. PTX treatment did not significantly affect the beta(2)-AR-stimulated PKA activation. Similar to G(i) inhibition, a protein phosphatase inhibitor, calyculin A (3 x 10(-8) M), selectively enhanced the beta(2)-AR but not beta(1)-AR-mediated contractile response. Furthermore, PTX and calyculin A treatment had a non-additive potentiating effect on the beta(2)-AR-mediated positive inotropic response. These results suggest that the interaction of the beta(2)-AR-coupled G(i) and G(s) signaling affects the local balance of protein kinase and phosphatase activities. Thus, the additional coupling of beta(2)-AR to G(i) proteins is a key factor causing the compartmentalization of beta(2)-AR-induced cAMP signaling.


Subject(s)
Ethanolamines/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heart/physiology , Myocardium/cytology , Receptors, Adrenergic, beta-2/physiology , Signal Transduction/physiology , Adrenergic beta-2 Receptor Agonists , Adrenergic beta-2 Receptor Antagonists , Animals , Calcium Channels/physiology , Calcium Channels, L-Type , Calcium-Binding Proteins/metabolism , Cell Size , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart/drug effects , Heart Ventricles , Marine Toxins , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Oxazoles/pharmacology , Pertussis Toxin , Phosphorylation , Propanolamines/pharmacology , Rats , Signal Transduction/drug effects , Thionucleotides/pharmacology , Vasoconstrictor Agents/pharmacology , Virulence Factors, Bordetella/pharmacology
20.
Circulation ; 99(18): 2458-65, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318670

ABSTRACT

BACKGROUND: Recent studies of beta-adrenergic receptor (beta-AR) subtype signaling in in vitro preparations have raised doubts as to whether the cAMP/protein kinase A (PKA) signaling is activated in the same manner in response to beta2-AR versus beta1-AR stimulation. METHODS AND RESULTS: The present study compared, in the intact dog, the magnitude and characteristics of chronotropic, inotropic, and lusitropic effects of cAMP accumulation, PKA activation, and PKA-dependent phosphorylation of key effector proteins in response to beta-AR subtype stimulation. In addition, many of these parameters and L-type Ca2+ current (ICa) were also measured in single canine ventricular myocytes. The results indicate that although the cAMP/PKA-dependent phosphorylation cascade activated by beta1-AR stimulation could explain the resultant modulation of cardiac function, substantial beta2-AR-mediated chronotropic, inotropic, and lusitropic responses occurred in the absence of PKA activation and phosphorylation of nonsarcolemmal proteins, including phospholamban, troponin I, C protein, and glycogen phosphorylase kinase. However, in single canine myocytes, we found that beta2-AR-stimulated increases in both ICa and contraction were abolished by PKA inhibition. Thus, the beta2-AR-directed cAMP/PKA signaling modulates sarcolemmal L-type Ca2+ channels but does not regulate PKA-dependent phosphorylation of cytoplasmic proteins. CONCLUSIONS: These results indicate that the dissociation of beta2-AR signaling from cAMP regulatory systems is only apparent and that beta2-AR-stimulated cAMP/PKA signaling is uncoupled from phosphorylation of nonsarcolemmal regulatory proteins involved in excitation-contraction coupling.


Subject(s)
Cyclic AMP/physiology , Heart/drug effects , Myocardial Contraction/drug effects , Myocardium/metabolism , Protein Processing, Post-Translational , Receptors, Adrenergic, beta-2/physiology , Second Messenger Systems/physiology , Sulfonamides , Actin Cytoskeleton/metabolism , Adrenergic Agents/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cardiotonic Agents/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Dogs , Imidazoles/pharmacology , Isoquinolines/pharmacology , Muscle Relaxation/drug effects , Norepinephrine/pharmacology , Phosphorylase Kinase/metabolism , Phosphorylase a/metabolism , Phosphorylase b/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Receptors, Adrenergic, beta-1/drug effects , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/drug effects , Reserpine/pharmacology , Sarcolemma/metabolism , Second Messenger Systems/drug effects , Thionucleotides/pharmacology , Troponin C/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...