Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 6(6): e20855, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21695261

RESUMO

BACKGROUND: Most ion channels are regulated by phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)) in the cell membrane by diverse mechanisms. Important molecular tools to study ion channel regulation by PtdIns(4,5)P(2) in living cells have been developed in the past. These include fluorescent PH-domains as sensors for Förster resonance energy transfer (FRET), to monitor changes in plasma membrane(.) For controlled and reversible depletion of PtdIns(4,5)P(2), voltage-sensing phosphoinositide phosphatases (VSD) have been demonstrated as a superior tool, since they are independent of cellular signaling pathways. Combining these methods in intact cells requires multiple transfections. We used self-cleaving viral 2A-peptide sequences for adenovirus driven expression of the PH-domain of phospholipase-Cδ1 (PLCδ1) fused to ECFP and EYFP respectively and Ciona intestinalis VSP (Ci-VSP), from a single open reading frame (ORF) in adult rat cardiac myocytes. METHODS AND RESULTS: Expression and correct targeting of ECFP-PH-PLCδ1(,) EYFP-PH-PLCδ1, and Ci-VSP from a single tricistronic vector containing 2A-peptide sequences first was demonstrated in HEK293 cells by voltage-controlled FRET measurements and Western blotting. Adult rat cardiac myocytes expressed Ci-VSP and the two fluorescent PH-domains within 4 days after gene transfer using the vector integrated into an adenoviral construct. Activation of Ci-VSP by depolarization resulted in rapid changes in FRET ratio indicating depletion of PtdIns(4,5)P(2) in the plasma membrane. This was paralleled by inhibition of endogenous G protein activated K(+) (GIRK) current. By comparing changes in FRET and current, a component of GIRK inhibition by adrenergic receptors unrelated to depletion of PtdIns(4,5)P(2) was identified. CONCLUSIONS: Expression of a FRET sensor pair and Ci-VSP from a single ORF provides a useful approach to study regulation of ion channels by phosphoinositides in cell lines and transfection-resistant postmitotic cells. Generally, adenoviral constructs containing self-cleaving 2A-peptide sequences are highly suited for simultaneous transfer of multiple genes in adult cardiac myocytes.


Assuntos
Membrana Celular/metabolismo , Técnicas Genéticas , Fosfatidilinositol 4,5-Difosfato/metabolismo , Adenoviridae/genética , Animais , Ciona intestinalis/enzimologia , DNA Complementar/genética , Transferência Ressonante de Energia de Fluorescência , Vetores Genéticos/genética , Células HEK293 , Átrios do Coração/citologia , Homeostase , Humanos , Miócitos Cardíacos/metabolismo , Fases de Leitura Aberta/genética , Fosfolipase C delta/química , Fosfolipase C delta/genética , Fosfolipase C delta/metabolismo , Monoéster Fosfórico Hidrolases/genética , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
2.
Pflugers Arch ; 461(1): 165-76, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21061016

RESUMO

G protein-activated K(+) channels composed of Kir3 (GIRK) subunits contribute to regulation of heart rate and excitability. Opening of these channels in myocytes is increased by binding of G(ßγ) upon activation of muscarinic M(2) receptors (M(2)-R) or A(1) adenosine receptors (A(1)-R). It has been shown that saturating activation of A(1)-R resulted in a smaller GIRK current than activation of M(2)-R. Adenovirus-driven overexpression of the A(1)-R caused an increase in current induced by adenosine (I(K(Ado))), whereas the M(2)-R-activated current (I(K(ACh))) was reduced. Here, we sought to get deeper insight into the mechanism causing this negative crosstalk. GIRK current in cultured rat atrial myocytes was recorded in whole cell mode. Adenovirus-driven RNA interference targeting the M(2)-R resulted in a reduction in I(K(ACh)) without affecting I(K(Ado)), arguing against a competition of the two receptors for common signaling complexes. The negative effect of A(1)-R overexpression on I(K(ACh)) was reduced by the A(1)-R antagonist DPCPX and augmented by the agonist chloro-N6-cyclopentyladenosin (CCPA). In native myocytes incubation with either CCPA or the muscarinic agonist carbachol resulted in reduction in I(K(ACh)) and I(K(Ado)), suggesting common pathways of A(1)-R and M(2)-R downregulation. In the absence of agonist, inhibition of adenosine deaminase by EHNA or exposure to AMP, less to ADP, but not ATP resulted in reduction of I(K(ACh)) and I(K(Ado)). Our data indicate that atrial myocytes generate adenosine from extracellular AMP, which activates A(1)-R in an autocrine fashion. Chronic activation of A(1)-R causes parallel downregulation of both A(1)-R and M(2)-R.


Assuntos
Comunicação Autócrina , Miócitos Cardíacos/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor Muscarínico M2/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Adenosina Desaminase/metabolismo , Inibidores de Adenosina Desaminase/farmacologia , Monofosfato de Adenosina/metabolismo , Animais , Carbacol/farmacologia , Células Cultivadas , Regulação para Baixo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Átrios do Coração , Agonistas Muscarínicos/farmacologia , Técnicas de Patch-Clamp , Interferência de RNA , Ratos , Receptor Cross-Talk , Receptor A1 de Adenosina/genética , Receptor Muscarínico M2/genética , Xantinas/farmacologia
3.
J Biol Chem ; 286(1): 290-8, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21041301

RESUMO

A large conductance (∼300 picosiemens) channel (LCC) of unknown molecular identity, activated by Ca(2+) release from the sarcoplasmic reticulum, particularly when augmented by caffeine, has been described previously in isolated cardiac myocytes. A potential candidate for this channel is pannexin 1 (Panx1), which has been shown to form large ion channels when expressed in Xenopus oocytes and mammalian cells. Panx1 function is implicated in ATP-mediated auto-/paracrine signaling, and a crucial role in several cell death pathways has been suggested. Here, we demonstrate that after culturing for 4 days LCC activity is no longer detected in myocytes but can be rescued by adenoviral gene transfer of Panx1. Endogenous LCCs and those related to expression of Panx1 share key pharmacological properties previously used for identifying and characterizing Panx1 channels. These data demonstrate that Panx1 constitutes the LCC of cardiac myocytes. Sporadic openings of single Panx1 channels in the absence of Ca(2+) release can trigger action potentials, suggesting that Panx1 channels potentially promote arrhythmogenic activities.


Assuntos
Conexinas/metabolismo , Canais Iônicos/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Potenciais de Ação , Trifosfato de Adenosina/metabolismo , Adenoviridae/genética , Animais , Fenômenos Biomecânicos , Conexinas/genética , Feminino , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Masculino , Miócitos Cardíacos/citologia , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Wistar , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
4.
Methods Mol Biol ; 515: 107-23, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378115

RESUMO

RNA interference (RNAi) represents the most frequently utilized technique to analyze proteins by loss of function assays. Protein synthesis is impaired by sequence-specific degradation of mRNA, which is triggered by short (19-28 nt) silencing RNAs (siRNA). Efficient gene silencing using RNAi has been demonstrated in numerous cell lines and primary cultured cells. Incorporation of siRNA into terminally differentiated mammalian cells, such as adult cardiac myocytes is limited by their resistance to standard transfection protocols. Viral delivery of short-hairpin RNA (shRNA) overcomes these limitations and allows efficient gene silencing in these cells. This chapter describes the generation and characterization of recombinant siRNA-encoding adenoviruses and their application to adult cardiac myocytes, which represent a standard experimental model in research related to cardiac physiology and pathophysiology. Feasibility of this approach is demonstrated by effective ablation (>80%) of both, a transgene encoding for eGFP and the endogenous muscarinic M(2) acetylcholine receptor.


Assuntos
Adenoviridae/genética , Diferenciação Celular , Técnicas de Transferência de Genes , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Interferência de RNA , Adenoviridae/isolamento & purificação , Animais , Células Cultivadas , Clonagem Molecular , Vetores Genéticos/genética , Humanos , Ratos , Vírion/genética , Vírion/isolamento & purificação
5.
Cell Physiol Biochem ; 21(4): 259-68, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18441514

RESUMO

Inwardly-rectifying K+ channel subunits are not homogenously expressed in different cardiac tissues. In ventricular myocytes (VM) the background current-voltage relation is dominated by I(K1), carried by channels composed of Kir2.x subunits, which is less important in atrial myocytes (AM). On the other hand in AM a large G protein gated current carried by Kir3.1/3.4 complexes can be activated by stimulation of muscarinic M(2) receptors (I(K(ACh))), which is assumed to be marginal in VM. Recent evidence suggests that total current carried by cardiac inward-rectifiers (I(K(ATP)), I(K(ACh)), I(K1)) in both, AM and VM is limited, due to K+ accumulation/depletion. This lead us to hypothesize that in conventional whole celI recordings I(K(ACh)) in VM is underestimated as a consequence of constitutive I(K1). In that case a reduction in density of I(K1) should be paralleled by an increase in density of I(K(ACh)). Three different experimental strategies have been used to test for this hypothesis: (i) Adenovirus-driven expression of a dominant-negative mutant of Kir2.1, one of the subunits supposed to form I(K1) channels, in VM caused a reduction in I(K1)-density by about 80 %. In those cells I(K(ACh)) was increased about 4 fold. (ii) A comparable increase in I(K(ACh)) was observed upon reduction of I(K1) caused by adenovirus-mediated RNA interference.(iii) Ba2+ in a concentration of 2 microM blocks I(K1) in VM by about 60 % without affecting atrial I(K(ACh)). The reduction in I(K1) by 2 microM Ba2+ is paralleled by a reversible increase in I(K(ACh)) by about 100%. These data demonstrate that the increase in K+ conductance underlying ventricular I(K(ACh)) is largely underestimated, suggesting that it might be of greater physiological relevance than previously thought.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Artérias/metabolismo , Sobrevivência Celular , Células Cultivadas , Eletrofisiologia , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , RNA Interferente Pequeno/genética , Ratos
6.
J Physiol ; 586(8): 2049-60, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18276732

RESUMO

The effect of beta-adrenergic stimulation on endogenous G-protein-activated K(+) (GIRK) current has been investigated in atrial myocytes from hearts of adult rats. Beta-adrenergic stimulation (10 microm isoprenaline, Iso) had no effect on activation kinetics, peak current or steady-state current but resulted in slowing of deactivation upon washout of acetylcholine (ACh), the time constant (tau(d)) being increased by a factor of about 2.5. The effect of Iso could be mimicked by inclusion of cAMP (500 microm) in the filling solution of the patch clamp pipette. The Iso-induced increase in tau(d) was blocked by the selective beta(1) receptor antagonist CGP-20112A (2 microm) and by the PKA inhibitor H9 (100 microm included in the pipette solution). A candidate for mediating these effects is RGS10, one of the regulators of G-protein signalling (RGS) species expressed in cardiac myocytes. Overexpression of RGS10 by adenoviral gene transfer resulted in a reduction in tau(d) of 60%. Sensitivity of tau(d) to Iso remained in cells overexpressing RGS10. Overexpression of RGS4 caused a comparable reduction in tau(d), which became insensitive to Iso. Expression of an RGS10 carrying a mutation (RGS10-S168A), which deletes a PKA phosphorylation site, caused a decrease in tau(d) comparable to overexpression of wild-type RGS10. Sensitivity of tau(d) to Iso was lost in RGS10-S168A-expressing myocytes. Silencing of RGS10 by means of adenovirus-mediated transcription of a short hairpin RNA did not affect basal tau(d) but removed sensitivity to Iso. These data suggest that endogenous RGS10 has GTPase-activating protein (GAP) activity on the G-protein species that mediates activation of atrial GIRK channels. Moreover, RGS10, via PKA-dependent phosphorylation, enables a crosstalk between beta-adrenergic and muscarinic cholinergic signalling.


Assuntos
Função Atrial/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/fisiologia , Potássio/metabolismo , Proteínas RGS/metabolismo , Animais , Células Cultivadas , Feminino , Átrios do Coração/citologia , Masculino , Ratos , Ratos Endogâmicos WKY
7.
J Physiol ; 585(Pt 1): 3-13, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17884923

RESUMO

Apart from gating by interaction with betagamma subunits from heterotrimeric G proteins upon stimulation of appropriate receptors, Kir.3 channels have been shown to be gated by intracellular Na+. However, no information is available on how Na+-dependent gating affects endogenous Kir3.1/Kir3.4 channels in mammalian atrial myocytes. We therefore studied how loading of adult atrial myocytes from rat hearts via the patch pipette filling solution with different concentrations of Na+ ([Na+]pip) affects Kir3 current. Surprisingly, in a range between 0 and 60 mm, Na+ neither had an effect on basal inward-rectifier current nor on the current activated by acetylcholine. Overexpression of Kir3.4 in adult atrial myocytes forced by adenoviral gene transfer results in formation of functional homomeric channels that interact with betagamma subunits upon activation of endogenous muscarinic receptors. These channels are activated at [Na+]pip >or= 15 mm, resulting in a receptor-independent basal inward rectifier current (I bir). I bir was neither affected by pertussis toxin nor by GDP-beta-S, suggesting G-protein-independent activation. PIP(2) depletion via endogenous PLC-coupled alpha1 adrenergic receptors causes inhibition of endogenous Kir3.1/3.4 channel currents by about 75%. In contrast, inhibition of Na+-activated I bir amounts to < 20%. The effect of the Kir3 channel blocker tertiapin-Q can be described using an IC50 of 12 nm (endogenous I K(ACh)) and 0.61 nm (I bir). These data clearly identify I bir as a homotetrameric Kir3.4 channel current with novel properties of regulation and pharmacology. Ibir shares some properties with a basal current recently described in atrial myocytes from an animal model of atrial fibrillation (AF) and AF patients.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Átrios do Coração/metabolismo , Ativação do Canal Iônico/fisiologia , Miócitos Cardíacos/metabolismo , Sódio/fisiologia , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Fibrilação Atrial/metabolismo , Fibrilação Atrial/patologia , Fibrilação Atrial/fisiopatologia , Venenos de Abelha/farmacologia , Células Cultivadas , Colinérgicos/farmacologia , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Proteínas de Ligação ao GTP/fisiologia , Átrios do Coração/patologia , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ratos , Ratos Endogâmicos WKY , Sódio/farmacologia
8.
J Muscle Res Cell Motil ; 27(5-7): 413-21, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16874447

RESUMO

RNA interference (RNAi) by short double stranded RNA (siRNA) represents an efficient and frequently used tool for gene silencing to study gene function. Whereas efficient ablation of genes has been demonstrated in neonatal cardiac myocytes, thus far information on successful application of this technique in adult cardiac myocytes (ACM), a standard experimental model in cardiac physiology and pathophysiology, is sparse. Here we demonstrate efficient ablation of a transgene encoding for enhanced green fluorescent protein (EGFP) and a cell specific endogenous gene encoding for an inward-rectifier channel subunit (Kir2.1) in ACM in vitro using adenovirus driven transcription of siRNA hairpins. EGFP fluorescence and density of background inward rectifier current (IK1) were reduced by > 90% within about 6-8 days after transformation with the corresponding virus. In Kir2.1-silenced myocytes resting membrane potential was significantly reduced. Survival of these cells in culture was compromised, presumably due to Ca2+ -overload caused by the depolarization. The sequence-specific knockdowns of EGFP and Kir2.1 were confirmed on the RNA level using real-time RT-PCR. In Kir2.1-silenced myocytes density of transient outward current, carried predominantly by Kv4.x subunits remained unaffected. This communication for the first time demonstrates proof of principle of efficient RNA interference using adenovirus-based vectors and demonstrates its large potential in phenotyping of ACM.


Assuntos
Adenoviridae/genética , Miócitos Cardíacos/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Interferência de RNA , Animais , Células Cultivadas , Regulação para Baixo , Inativação Gênica , Ventrículos do Coração/citologia , Potenciais da Membrana/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , RNA Interferente Pequeno , Ratos , Fatores de Tempo , Transformação Genética
9.
J Physiol ; 561(Pt 2): 471-83, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15459243

RESUMO

We have investigated the acute desensitization of acetylcholine-activated GIRK current (I(K(ACh))) in cultured adult rat atrial myocytes. Acute desensitization of I(K(ACh)) is observed as a partial relaxation of current with a half-time of < 5 s when muscarinic M2 receptors are stimulated by a high concentration (> 2 micromol l(-1)) of ACh. Under this condition experimental manoeuvres that cause a decrease in the amplitude of I(K(ACh)), such as partial block of M2 receptors by atropine, intracellular loading with GDP-beta-S, or exposure to Ba2+, caused a reduction in desensitization. Acute desensitization was also identified as a decrease in current amplitude and a blunting of the response to saturating [ACh] (20 micromol l(-1)) when the current had been partially activated by a low concentration of ACh or by stimulation of adenosine A1 receptors. A reduction in current analogous to acute desensitization was observed when ATP-dependent K+ current (I(K(ATP))) was activated either by mitochondrial uncoupling using 2,4-dinitrophenole (DNP) or by the channel opener rilmakalim. Adenovirus-driven overexpression of Kir2.1, a subunit of constitutively active inwardly rectifying K+ channels, resulted in a large Ba2+-sensitive background K+ current and a dramatic reduction of ACh-activated current. Adenovirus-driven overexpression of GIRK4 (Kir3.4) subunits resulted in an increased agonist-independent GIRK current paralleled by a reduction in I(K(ACh)) and removal of the desensitizing component. These data indicate that acute desensitization depends on K+ current flow, independent of the K+ channel species, suggesting that it reflects a reduction in electrochemical driving force rather than a bona fide signalling mechanism. This is supported by the observation that desensitization is paralleled by a significant negative shift in reversal potential of I(K(ACh)). Since the ACh-induced hyperpolarization shows comparable desensitization properties as I(K(ACh)), this novel current-dependent desensitization is a physiologically relevant process, shaping the time course of parasympathetic bradycardia.


Assuntos
Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Potássio/fisiologia , Acetilcolina/farmacologia , Animais , Células Cultivadas , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos
10.
J Physiol ; 561(Pt 2): 459-69, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15459245

RESUMO

In this study we have investigated the voltage dependence of ATP-dependent K+ current (I(K(ATP))) in atrial and ventricular myocytes from hearts of adult rats and in CHO cells expressing Kir6.2 and SUR2A. The current-voltage relation of 2,4-dinitrophenole (DNP) -induced I(K(ATP)) in atrial myocytes and expressed current in CHO cells was linear in a voltage range between 0 and -100 mV. In ventricular myocytes, the background current-voltage relation of which is dominated by a large constitutive inward rectifier (I(K1)), the slope conductance of I(K(ATP)) was reduced at membrane potentials negative to E(K) (around -50 mV), resulting in an outwardly rectifying I-V relation. Overexpression of Kir2.1 by adenoviral gene transfer, a subunit contributing to I(K1) channels, in atrial myocytes resulted in a large I(K1)-like background current. The I-V relation of I(K(ATP)) in these cells showed a reduced slope conductance negative to E(K) similar to ventricular myocytes. In atrial myocytes with an increased background inward-rectifier current through Kir3.1/Kir3.4 channels (I(K(ACh))), irreversibly activated by intracellular loading with GTP-gamma-S, the I-V relation of I(K(ATP)) showed a reduced slope negative to E(K), as in ventricular myocytes and atrial myocytes overexpressing Kir2.1. It is concluded that the voltage dependencies of membrane currents are not only dependent on the molecular composition of the charge-carrying channel complexes but can be affected by the activity of other ion channel species. We suggest that the interference between inward I(K(ATP)) and other inward rectifier currents in cardiac myocytes reflects steady-state changes in K+ driving force due to inward K+ current.


Assuntos
Acetilcolina/farmacologia , Trifosfato de Adenosina/fisiologia , Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Transportadores de Cassetes de Ligação de ATP/agonistas , Transportadores de Cassetes de Ligação de ATP/fisiologia , Animais , Células CHO , Células Cultivadas , Cricetinae , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Pinacidil/farmacologia , Canais de Potássio/agonistas , Canais de Potássio/fisiologia , Canais de Potássio Cálcio-Ativados/agonistas , Canais de Potássio Cálcio-Ativados/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/agonistas , Ratos , Receptores de Droga/agonistas , Receptores de Droga/fisiologia , Receptores de Sulfonilureias
11.
Biochim Biophys Acta ; 1642(1-2): 67-77, 2003 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-12972295

RESUMO

The predominant histamine receptor subtype in the supraventricular and ventricular tissue of various mammalian species is the H2 receptor (H2-R) subtype, which is known to couple to stimulatory G proteins (Gs), i.e. the major effects of this autacoid are an increase in sinus rate and in force of contraction. To investigate histamine effects in H2-R-transfected rat atrial myocytes, endogenous GIRK currents and L-type Ca2+ currents were used as functional assays. In H2-R-transfected myocytes, exposure to His resulted in a reversible augmentation of L-type Ca2+ currents, consistent with the established coupling of this receptor to the Gs-cAMP-PKA signalling pathway. Mammalian K+ channels composed of GIRK (Kir3.x) subunits are directly controlled by interaction with betagamma subunits released from G proteins, which couple to seven-helix receptors. In mock-transfected atrial cardiomyocytes, activation of muscarinic K+ channels (IK(ACh)) was limited to Gi-coupled receptors (M2R, A1R). In H2-R-overexpressing cells, histamine activated IK(ACh) via Gs-derived betagamma subunits since the histamine-induced current was insensitive to pertussis toxin. These data indicate that overexpression of Gs-coupled H2-R results in a loss of target specificity due to an increased agonist-induced release of Gs-derived betagamma subunits. When IK(ACh) was maximally activated by GTP-gamma-S, histamine induced an irreversible inhibition of the inward current in a fraction of H2-R-transfected cells. This inhibition is supposed to be mediated via a G(q/11)-PLC-mediated depletion of PIP2, suggesting a partial coupling of overexpressed H2-R to G(q/11). Dual coupling of H2-Rs to Gs and Gq is demonstrated for the first time in cardiac myocytes. It represents a novel mechanism to augment positive inotropic effects by activating two different signalling pathways via one type of histamine receptor. Activation of the Gs-cAMP-PKA pathway promotes Ca2+ influx through phosphorylation of L-type Ca2+ channels. Simultaneous activation of Gq-signalling pathways might result in phosphoinositide turnover and Ca2+ release from intracellular stores, thereby augmenting H2-induced increases in [Ca2+]i.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Átrios do Coração/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização , Receptores Histamínicos H2/metabolismo , Animais , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Subunidades alfa Gs de Proteínas de Ligação ao GTP/efeitos dos fármacos , Átrios do Coração/efeitos dos fármacos , Proteínas Heterotriméricas de Ligação ao GTP/efeitos dos fármacos , Histamina/metabolismo , Histamina/farmacologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/metabolismo , Ratos , Receptor Muscarínico M2 , Receptores Histamínicos H2/efeitos dos fármacos , Receptores Histamínicos H2/genética , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transfecção
12.
J Physiol ; 550(Pt 3): 707-17, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12815176

RESUMO

G protein-activated inwardly rectifying K+ (GIRK) channels, important regulators of membrane excitability in the heart and central nervous system, are activated by interaction with betagamma subunits from heterotrimeric G proteins upon receptor stimulation. In atrial myocytes various endogenous receptors couple to GIRK channels, including the canonical muscarinic M2 receptor (M2AChR) and the A1 adenosine receptor (A1AdoR). Saturating stimulation of A1AdoR in atrial myocytes activates only a fraction of the GIRK current that is activated via M2AChR, which reflects a lower density of A1AdoR. In the present study A1AdoR were overexpressed by means of adenovirus-mediated gene transfer using green fluorescent protein (GFP) as the reporter. Confirmatory to a previous study, this resulted in an increased sensitivity of macroscopic GIRK current (ACh-activated K+ current (IK(ACh))) to stimulation by Ado. However, in the majority of GFP-positive myocytes, exposure to Ado at concentrations > or =1 microM resulted in activation of IK(ACh) followed by a rapid inhibition. In those cells a rebound activation of current was recorded upon washout of Ado. The inhibitory component could be recorded in isolation when IK(ACh) was activated by M2AChR-stimulation and brief pulses of Ado were superimposed. In myocytes loaded with GTP-gamma-S, IK(ACh), irreversibly activated by brief exposure to agonist, was still reversibly inhibited by Ado, suggesting that inhibition is independent of G protein cycling. In myocytes co-transfected with adenoviral vectors encoding A1AdoR and GIRK4 subunit, no inhibition of GIRK current by Ado was observed. As acute desensitization of atrial GIRK current, which is reminiscent of the inhibition described here, has been shown to be absent in myocytes overexpressing GIRK4, this suggests that acute desensitization and the novel inhibition might share a common pathway whose target is the GIRK channel complex or its GIRK1 subunit.


Assuntos
Adenosina/farmacologia , Adenoviridae/genética , Proteínas de Ligação ao GTP/fisiologia , Células Musculares/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/efeitos dos fármacos , Receptores Purinérgicos P1/biossíntese , Acetilcolina/metabolismo , Animais , Primers do DNA , Eletrofisiologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Técnicas de Transferência de Genes , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Técnicas In Vitro , Potenciais da Membrana/fisiologia , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Técnicas de Patch-Clamp , Canais de Potássio/genética , Ratos , Receptores Purinérgicos P1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Mol Cell Cardiol ; 35(1): 99-107, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12623304

RESUMO

A recently described reduction in sensitivity of G protein-activated inward-rectifying K(+) (GIRK) channels to stimulation of muscarinic M(2) receptors (M(2)AChR) in atrial myocytes overexpressing purinergic A(1) receptors (A(1)AdoR) was further investigated by heterologous expression of a 5-HT(1A) receptor (5-HT(1A)R) and by reducing the expression level of endogenous M(2)AChR receptors using antisense. In 5-HT(1A)R-expressing myocytes, in line with previous studies, sizable GIRK currents could be activated by 5-HT. In these cells, the mean current density and activation rate of M(2)AChR-activated current were significantly reduced, supporting the notion that signalling via this receptor is negatively regulated by other G protein-coupled receptors (GPCR) coupling to the same class (G(i/o)) of G proteins. To study if reducing M(2)AChR expression affects sensitivity of GIRK current to stimulation of A(1)AdoR, antisense oligodinucleotides (AsODN) against the M(2)AChR were used. Incubation of myocytes with M(2)AChR-specific AsODN resulted in a significant reduction in mean amplitude and activation rate of ACh-induced currents. This was paralleled by an increase in mean amplitude and activation rate of current activated by stimulation of A(1)AdoR. Plotting amplitudes of 5-HT- or Ado-induced currents from individual manipulated cells against the amplitude of ACh-induced current yielded a positive correlation between these data. Although difficult to interpret in mechanistic terms, this argues against a competition of receptors for a common pool of G(i/o). The mutual interaction between G(i/o)-coupled receptors depends on manipulation of the expression level, since long-term desensitization or down regulation of M(2)AChR by treatment with carbachol did not affect sensitivity of GIRK current to A(1)AdoR stimulation, despite a substantial reduction in amplitude and activation rate of M(2)AChR-activated currents. These data suggest a novel crosstalk between parallel receptors converging on the same class of G proteins.


Assuntos
Oligonucleotídeos Antissenso , Receptores Muscarínicos/genética , Receptores de Serotonina/genética , Transfecção , Animais , Comunicação Celular/genética , Comunicação Celular/fisiologia , Feminino , Masculino , Ratos , Receptor Muscarínico M2 , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Muscarínicos/metabolismo , Receptores de Serotonina/metabolismo , Receptores 5-HT1 de Serotonina
14.
FEBS Lett ; 529(2-3): 356-60, 2002 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-12372628

RESUMO

GIRK (G protein-activated inward-rectifying K(+) channel) channels, important regulators of membrane excitability in the heart and in the central nervous, are activated by interaction with betagamma subunits from heterotrimeric G proteins upon receptor stimulation. For activation interaction of the channel with phosphatidylinositol 4,5-bisphosphate (PtIns(4,5)P(2)) is conditional. Previous studies have provided evidence that in myocytes PtIns(4,5)P(2) levels relevant to GIRK channel regulation are under regulatory control of receptors activating phospholipase C. In the present study a phosphatidyl-4-phosphate 5-kinase was expressed in atrial myocytes by transient transfection. This did not affect basal properties of GIRK current activated by acetylcholine via M(2) receptors but completely abolished inhibition of guanosine triphosphate-gamma-S activated current by endothelin-1 or alpha-adrenergic agonists. We conclude that though PtIns(4,5)P(2) is conditional for channel gating, its normal level in the membrane is not limiting basal function of GIRK channels. Moreover, our data provide further evidence for a regulation of GIRK channels by alpha(1A) receptors and endothelin-A receptors, endogenously expressed in atrial myocytes, via depletion of PtIns(4,5)P(2).


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Endotelina-1/farmacologia , Átrios do Coração/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Bloqueadores dos Canais de Potássio , Transfecção , Animais , Feminino , Masculino , Ratos , Ratos Endogâmicos WKY
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...