Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 587(Pt 11): 2599-612, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19403597

RESUMO

Activation of myosin light chain kinase (MLCK) and other kinases was studied in the arteries of transgenic mice that express an optical fluorescence resonance energy transfer (FRET) MLCK activity biosensor. Binding of Ca(2+)/calmodulin (Ca(2+)/CaM) induces an increase in MLCK activity and a change in FRET. After exposure to high external [K(+)], intracellular [Ca(2+)] (fura-2 ratio or fluo-4 fluorescence) and MLCK activity both increased rapidly to an initial peak and then declined, rapidly at first and then very slowly. After an initial peak ('phasic') force was constant or increased slowly (termed 'tonic' force). Inhibition of rho-kinase (Y-27632) decreased tonic force more than phasic, but had little effect on [Ca(2+)] and MLCK activation. Inhibition of PKCalpha and PKCbeta with Gö6976 had no effect. KN-93, an inhibitor of CaMK II, markedly reduced force, MLCK FRET and [Ca(2+)]. Applied during tonic force, forskolin caused a rapid decrease in MLCK FRET ratio and force, but no change in Ca(2+), suggesting a cAMP mediated decrease in affinity of MLCK for Ca(2+)/CaM. However, receptor (beta-adrenergic) activated increases in cAMP during KCl were ineffective in causing relaxation, changes in [Ca(2+)], or MLCK FRET. At the same tonic force, MLCK FRET ratio activated by alpha(1)-adrenoceptors was approximately 60% of that activated by KCl. In conclusion, MLCK activity of arterial smooth muscle during KCl-induced contraction is determined primarily by Ca(2+)/CaM. Rho-kinase is activated, by unknown mechanisms, and increases 'Ca(2+) sensitivity' significantly. Forskolin mediated increases in cAMP, but not receptor mediated increases in cAMP cause a rapid decrease in the affinity of MLCK for Ca(2+)/CaM.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Artérias Mesentéricas/enzimologia , Quinase de Cadeia Leve de Miosina/metabolismo , Vasoconstrição , Adenilil Ciclases/metabolismo , Animais , Técnicas Biossensoriais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Transferência Ressonante de Energia de Fluorescência , Cinética , Artérias Mesentéricas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fosforilação , Potássio/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores Adrenérgicos beta 1/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 291(6): H3106-13, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16920810

RESUMO

Brief, spatially localized Ca(2+) transients occur in the smooth muscle adjacent to perivascular nerves of small arteries during neurogenic contractions. We named these "junctional Ca(2+) transients" (jCaTs) and postulated that they arose from Ca(2+) entering smooth muscle cells through P2X(1) receptors activated by neurally released ATP. Nevertheless, the lack of potent, subtype-selective P2X-receptor antagonists made determining the exact molecular identity of the channels difficult. Here we used small, pressurized mesenteric arteries from P2X(1)-receptor-deficient mice (KO) to test the hypothesis that jCaTs arise from Ca(2+) entering the smooth muscle cell via P2X(1) receptors. In wild-type (WT) arteries, confocal microscopy of fluo-4 fluorescence during electrical field stimulation (EFS) of perivascular sympathetic nerves revealed jCaTs in the smooth muscle cells adjacent to the perivascular nerves, similar to those reported previously in rat arteries, and alpha-latrotoxin (2.5 nM) markedly increased the frequency of "spontaneous" jCaTs. In the KO arteries, however, neither EFS nor alpha-latrotoxin elicited any jCaTs. A potent P2X-receptor agonist, alpha,beta-methylene ATP (10.0 microM), elicited strong contractions and increased intracellular Ca(2+) concentration in WT arteries but elicited neither in KO arteries. A biphasic vasoconstriction in response to EFS was observed in WT arteries. In KO arteries, however, the initial rapid, transient component of the biphasic vasoconstriction was absent. The data support the hypothesis that jCaTs represent Ca(2+) that enters the smooth muscle cells through P2X(1) receptors activated by neurally released ATP and that this Ca(2+) is involved in the initial rapid component of the sympathetic neurogenic contraction.


Assuntos
Cálcio/metabolismo , Artérias Mesentéricas/inervação , Artérias Mesentéricas/metabolismo , Músculo Liso Vascular/inervação , Músculo Liso Vascular/metabolismo , Receptores Purinérgicos P2/metabolismo , Sistema Nervoso Simpático/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Estimulação Elétrica , Camundongos , Camundongos Transgênicos , Junção Neuromuscular/fisiologia , Prazosina/farmacologia , Agonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2X , Venenos de Aranha/farmacologia , Vasoconstrição/fisiologia
3.
Rev Physiol Biochem Pharmacol ; 150: 91-139, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12884052

RESUMO

Our goal in this review is to provide a comprehensive, integrated view of the numerous signaling pathways that are activated by alpha(1)-adrenoceptors and control actin-myosin interactions (i.e., crossbridge cycling and force generation) in mammalian arterial smooth muscle. These signaling pathways may be categorized broadly as leading either to thick (myosin) filament regulation or to thin (actin) filament regulation. Thick filament regulation encompasses both "Ca(2+) activation" and "Ca(2+)-sensitization" as it involves both activation of myosin light chain kinase (MLCK) by Ca(2+)-calmodulin and regulation of myosin light chain phosphatase (MLCP) activity. With respect to Ca(2+) activation, adrenergically induced Ca(2+) transients in individual smooth muscle cells of intact arteries are now being shown by high resolution imaging to be sarcoplasmic reticulum-dependent asynchronous propagating Ca(2+) waves. These waves differ from the spatially uniform increases in [Ca(2+)] previously assumed. Similarly, imaging during adrenergic activation has revealed the dynamic translocation, to membranes and other subcellular sites, of protein kinases (e.g., Ca(2+)-activated protein kinases, PKCs) that are involved in regulation of MLCP and thus in "Ca(2+) sensitization" of contraction. Thin filament regulation includes the possible disinhibition of actin-myosin interactions by phosphorylation of CaD, possibly by mitogen-activated protein (MAP) kinases that are also translocated during adrenergic activation. An hypothesis for the mechanisms of adrenergic activation of small arteries is advanced. This involves asynchronous Ca(2+) waves in individual SMC, synchronous Ca(2+) oscillations (at high levels of adrenergic activation), Ca(2+) sparks, "Ca(2+)-sensitization" by PKC and Rho-associated kinase (ROK), and thin filament mechanisms.


Assuntos
Contração Muscular , Músculo Liso Vascular/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Transdução de Sinais , Actinas/metabolismo , Animais , Cálcio/metabolismo , Calmodulina/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Modelos Anatômicos , Modelos Biológicos , Músculo Liso Vascular/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Miosinas/metabolismo , Retículo Sarcoplasmático/metabolismo , Fatores de Tempo
4.
Biophys J ; 81(5): 2647-59, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606278

RESUMO

The Na(+) current component I(Ca(TTX)) is functionally distinct from the main body of Na(+) current, I(Na). It was proposed that I(Ca(TTX)) channels are I(Na) channels that were altered by bathing media containing Ca(2+), but no, or very little, Na(+). It is known that Na(+)-free conditions are not required to demonstrate I(Ca(TTX).) We show here that Ca(2+) is also not required. Whole-cell, tetrodotoxin-blockable currents from fresh adult rat ventricular cells in 65 mm Cs(+) and no Ca(2+) were compared to those in 3 mM Ca(2+) and no Cs(+) (i.e., I(Ca(TTX))). I(Ca(TTX)) parameters were shifted to more positive voltages than those for Cs(+). The Cs(+) conductance-voltage curve slope factor (mean, -4.68 mV; range, -3.63 to -5.72 mV, eight cells) is indistinguishable from that reported for I(Ca(TTX)) (mean, -4.49 mV; range, -3.95 to -5.49 mV). Cs(+) current and I(Ca(TTX)) time courses were superimposable after accounting for the voltage shift. Inactivation time constants as functions of potential for the Cs(+) current and I(Ca(TTX)) also superimposed after voltage shifting, as did the inactivation curves. Neither of the proposed conditions for conversion of I(Na) into I(Ca(TTX)) channels is required to demonstrate I(Ca(TTX)). Moreover, we find that cardiac Na(+) (H1) channels expressed heterologously in HEK 293 cells are not converted to I(Ca(TTX)) channels by Na(+)-free, Ca(2+)-containing bathing media. The gating properties of the Na(+) current through H1 and those of Ca(2+) current through H1 are identical. All observations are consistent with two non-interconvertable Na(+) channel populations: a larger that expresses little Ca(2+) permeability and a smaller that is appreciably Ca(2+)-permeable.


Assuntos
Canais de Cálcio/metabolismo , Césio/antagonistas & inibidores , Césio/metabolismo , Miocárdio/metabolismo , Canais de Sódio/metabolismo , Tetrodotoxina/farmacologia , Animais , Canais de Cálcio/efeitos dos fármacos , Linhagem Celular/citologia , Membrana Celular/metabolismo , Eletrofisiologia/métodos , Ventrículos do Coração/citologia , Humanos , Potenciais da Membrana/fisiologia , Miocárdio/citologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Transfecção/métodos
5.
Cell Calcium ; 29(5): 327-34, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11292389

RESUMO

Confocal laser scanning microscopy and Fluo-4 were used to visualize Ca(2+) transients within individual smooth muscle cells (SMC) of rat resistance arteries during alpha(1)-adrenoceptor activation. The typical spatio-temporal pattern of [Ca(2+)] in an artery after exposure to a maximally effective concentration of phenylephrine (PE, 10.0 microM) was a large, brief, relatively homogeneous Ca(2+) transient, followed by Ca(2+) waves, which then declined in frequency over the course of 5 min and which were asynchronous in different SMC. Concentration-Effect (CE) curves relating the concentration of PE (range: 0.1 microM to 10.0 microM) to the effects (fraction of cells producing at least one Ca(2+) wave, and number of Ca(2+) waves during 5 min) had EC(50) values of approximately 0.5 microM and approximately 1.0 microM respectively. The initial Ca(2+) transient and the subsequent Ca(2+) waves were abolished in the presence of caffeine (10.0 mM). A repeated exposure to PE, 1.5 min after the first had ended, elicited fewer Ca(2+) waves in fewer cells than did the initial exposure. Caffeine-sensitive Ca(2+) stores were not depleted at this time, however, as caffeine alone was capable of inducing a large release of Ca(2+)1.5 min after PE. In summary, the mechanism of a graded response to graded alpha(1)-adrenoceptor activation is the progressive 'recruitment' of individual SMC, which then respond in 'all or none' fashion (viz. asynchronous Ca(2+) waves). Ca(2+) signaling continues in the arterial wall throughout the time-course (at least 5 min) of activation of alpha(1)-adrenoceptors. The fact that the Ca(2+) waves are asynchronous accounts for the previously reported fall in 'arterial wall [Ca(2+)]' (i.e. spatial average [Ca(2+)] over all cells).


Assuntos
Sinalização do Cálcio/fisiologia , Músculo Liso Vascular/fisiologia , Receptores Adrenérgicos alfa 1/metabolismo , Agonistas alfa-Adrenérgicos/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Artérias/fisiologia , Cafeína/farmacologia , Relação Dose-Resposta a Droga , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Fenilefrina/metabolismo , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley
6.
Am J Physiol Heart Circ Physiol ; 280(5): H2399-405, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11299247

RESUMO

Confocal laser scanning microscopy and fluo 4 were used to visualize local and whole cell Ca(2+) transients within individual smooth muscle cells (SMC) of intact, pressurized rat mesenteric small arteries during activation of alpha1-adrenoceptors. A method was developed to record the Ca(2+) transients within individual SMC during the changes in arterial diameter. Three distinct types of "Ca(2+) signals" were influenced by adrenergic activation (agonist: phenylephrine). First, asynchronous Ca(2+) transients were elicited by low levels of adrenergic stimulation. These propagated from a point of origin and then filled the cell. Second, synchronous, spatially uniform Ca(2+) transients, not reported previously, occurred at higher levels of adrenergic stimulation and continued for long periods during oscillatory vasomotion. Finally, Ca(2+) sparks slowly decreased in frequency of occurrence during exposure to adrenergic agonists. Thus adrenergic activation causes a decrease in the frequency of Ca(2+) sparks and an increase in the frequency of asynchronous wavelike Ca(2+) transients, both of which should tend to decrease arterial diameter. Oscillatory vasomotion is associated with spatially uniform synchronous oscillations of cellular [Ca(2+)] and may have a different mechanism than the asynchronous, propagating Ca(2+) transients.


Assuntos
Cálcio/metabolismo , Músculo Liso Vascular/metabolismo , Receptores Purinérgicos P1/metabolismo , Resistência Vascular/fisiologia , Animais , Temperatura Corporal , Masculino , Artérias Mesentéricas/metabolismo , Fenilefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasoconstritores/farmacologia
7.
Biophys J ; 80(1): 88-102, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11159385

RESUMO

Previous models of cardiac Ca2+ sparks have assumed that Ca2+ currents through the Ca2+ release units (CRUs) were approximately 1-2 pA, producing sparks with peak fluorescence ratio (F/F(0)) of approximately 2.0 and a full-width at half maximum (FWHM) of approximately 1 microm. Here, we present actual Ca2+ sparks with peak F/F(0) of >6 and a FWHM of approximately 2 microm, and a mathematical model of such sparks, the main feature of which is a much larger underlying Ca2+ current. Assuming infinite reaction rates and no endogenous buffers, we obtain a lower bound of approximately 11 pA needed to generate a Ca2+ spark with FWHM of 2 microm. Under realistic conditions, the CRU current must be approximately 20 pA to generate a 2- microm Ca2+)spark. For currents > or =5 pA, the computed spark amplitudes (F/F(0)) are large (approximately 6-12 depending on buffer model). We considered several factors that might produce sparks with FWHM approximately 2 microm without using large currents. Possible protein-dye interactions increased the FWHM slightly. Hypothetical Ca2+ "quarks" had little effect, as did blurring of sparks by the confocal microscope. A clusters of CRUs, each producing 10 pA simultaneously, can produce sparks with FWHM approximately 2 microm. We conclude that cardiac Ca2+ sparks are significantly larger in peak amplitude than previously thought, that such large Ca2+ sparks are consistent with the measured FWHM of approximately 2 microm, and that the underlying Ca2+ current is in the range of 10-20 pA.


Assuntos
Sinalização do Cálcio/fisiologia , Miocárdio/metabolismo , Animais , Fenômenos Biofísicos , Biofísica , Técnicas In Vitro , Matemática , Microscopia Confocal , Modelos Cardiovasculares , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
8.
Biophys J ; 80(1): 103-20, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11159386

RESUMO

We present a model that provides a unified framework for studying Ca2+ sparks and Ca2+ waves in cardiac cells. The model is novel in combining 1) use of large currents (approximately 20 pA) through the Ca2+ release units (CRUs) of the sarcoplasmic reticulum (SR); 2) stochastic Ca2+ release (or firing) of CRUs; 3) discrete, asymmetric distribution of CRUs along the longitudinal (separation distance of 2 microm) and transverse (separated by 0.4-0.8 microm) directions of the cell; and 4) anisotropic diffusion of Ca2+ and fluorescent indicator to study the evolution of Ca2+ waves from Ca2+ sparks. The model mimics the important features of Ca2+ sparks and Ca2+ waves in terms of the spontaneous spark rate, the Ca2+ wave velocity, and the pattern of wave propagation. Importantly, these features are reproduced when using experimentally measured values for the CRU Ca2+ sensitivity (approximately 15 microM). Stochastic control of CRU firing is important because it imposes constraints on the Ca2+ sensitivity of the CRU. Even with moderate (approximately 5 microM) Ca2+ sensitivity the very high spontaneous spark rate triggers numerous Ca2+ waves. In contrast, a single Ca2+ wave with arbitrarily large velocity can exist in a deterministic model when the CRU Ca2+ sensitivity is sufficiently high. The combination of low CRU Ca2+ sensitivity (approximately 15 microM), high cytosolic Ca2+ buffering capacity, and the spatial separation of CRUs help control the inherent instability of SR Ca2+ release. This allows Ca2+ waves to form and propagate given a sufficiently large initiation region, but prevents a single spark or a small group of sparks from triggering a wave.


Assuntos
Sinalização do Cálcio/fisiologia , Modelos Cardiovasculares , Miocárdio/metabolismo , Animais , Fenômenos Biofísicos , Biofísica , Soluções Tampão , Técnicas In Vitro , Cinética , Modelos Lineares , Miocárdio/citologia , Dinâmica não Linear , Retículo Sarcoplasmático/metabolismo , Processos Estocásticos
9.
Am J Physiol Heart Circ Physiol ; 278(6): H2150-6, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10843915

RESUMO

We describe a custom one-photon (confocal) and two-photon all-digital (photon counting) laser scanning microscope. The confocal component uses two avalanche photodiodes (APDs) as the fluorescence detector to achieve high sensitivity and to overcome the limited photon counting rate of a single APD ( approximately 5 MHz). The confocal component is approximately nine times more efficient than our commercial confocal microscope (fluorophore fluo 4). Switching from one-photon to two-photon excitation mode (Ti:sapphire laser) is accomplished by moving a single mirror beneath the objective lens. The pulse from the Ti:sapphire laser is 109 fs in duration at the specimen plane, and average power is approximately 5 mW. Two-photon excited fluorescence is detected by a fast photomultiplier tube. With a x63 1.4 NA oil-immersion objective, the resolution of the confocal system is 0.25 microm laterally and 0.52 microm axially. For the two-photon system, the corresponding values are 0.28 and 0.82 microm. The system is advantageous when excitation intensity must be limited, when fluorescence is low, or when thick, scattering specimens are being studied (with two-photon excitation).


Assuntos
Computadores , Microscopia Confocal/instrumentação , Fótons , Animais , Cálcio/metabolismo , Calibragem , Desenho de Equipamento , Fluorescência , Artérias Mesentéricas/citologia , Microscopia Confocal/normas , Miocárdio/citologia , Miocárdio/metabolismo , Pólen/fisiologia , Ratos , Sensibilidade e Especificidade , Raios Ultravioleta
11.
Biophys J ; 77(2): 808-16, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10423427

RESUMO

To study the function and regulation of the cardiac ryanodine receptor (RyR2) Ca(2+) release channel, we expressed the RyR2 proteins in a Chinese hamster ovary (CHO) cell line, and assayed its function by single channel current recording and confocal imaging of intracellular Ca(2+) ([Ca(2+)](i)). The 16-kb cDNA encoding the full-length RyR2 was introduced into CHO cells using lipofectAmine and electroporation methods. Incorporation of microsomal membrane vesicles isolated from these transfected cells into lipid bilayer membrane resulted in single Ca(2+) release channel activities similar to those of the native Ca(2+) release channels from rabbit cardiac muscle SR membranes, both in terms of gating kinetics, conductance, and ryanodine modification. The expressed RyR2 channels were found to exhibit more frequent transitions to subconductance states than the native RyR2 channels and RyR1 expressed in CHO cells. Caffeine, an exogenous activator of RyR, induced release of [Ca(2+)](i) from these cells. Confocal imaging of cells expressing RyR2 did not detect spontaneous or caffeine-induced local Ca(2+) release events (i.e., "Ca(2+) sparks") typically seen in cardiac muscle. Our data show that the RyR2 expressed in CHO cells forms functional Ca(2+) release channels. Furthermore, the lack of localized Ca(2+) release events in these cells suggests that Ca(2+) sparks observed in cardiac muscle may involve cooperative gating of a group of Ca(2+) release channels and/or their interaction with muscle-specific proteins.


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Fenômenos Biofísicos , Biofísica , Células CHO , Sinalização do Cálcio , Cricetinae , Eletroquímica , Expressão Gênica , Potenciais da Membrana , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
J Physiol ; 518 ( Pt 3): 815-24, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10420017

RESUMO

1. Confocal laser scanning microscopy was used to visualize Ca2+ transients in the vascular smooth muscle cells (VSMC) of intact, pressurized rat mesenteric resistance arteries loaded with fluorescent calcium indicators. Vasoconstriction was assessed by measuring inner arterial diameter. All arteries were studied at 70 mmHg intralumenal pressure and 37 C. 2. In the control condition of myogenic tone the arteries were constricted to 62 % (n = 10) of their passive diameter (p.d.). The [Ca2+]i in most VSMC of these arteries was constant over time. In a small percentage (< 10 %) of cells in each artery, [Ca2+]i oscillated regularly. Local calcium transients (Ca2+ sparks) were observed in five arteries studied with confocal linescan imaging. 3. Activation of alpha-adrenoceptors by phenylephrine (PE, 1.0 microM) induced further vasoconstriction of pressurized arteries (to 27 % of p.d.). In this condition, [Ca2+]i oscillations were prominent in a large percentage (83 %) of the VSMC. The Ca2+ oscillations ranged in frequency from 4 to 22 min-1, and were usually asynchronous between cells. 4. High [KCl]o (65 mM) induced nearly comparable vasoconstriction to PE (37 % of p.d.) but [Ca2+]i oscillated in only about 13 % of cells in each artery. 5. Block of L-type Ca2+ channels (with nifedipine) in arteries activated by PE caused nearly full vasodilatation, but did not abolish the Ca2+ oscillations. Subsequent block of the sarcoplasmic reticulum Ca2+ pump (with cyclopiazonic acid) abolished Ca2+ oscillations in all cells. 6. We conclude that Ca2+ entering VSMC via L-type Ca2+ channels has an obligatory role in force development, both in myogenic tone and during alpha1-adrenoceptor activation. The oscillatory pattern of [Ca2+]i that persists in the absence of Ca2+ entry via L-type Ca2+ channels is ineffective in activating contraction.


Assuntos
Artérias/fisiologia , Sinalização do Cálcio/fisiologia , Músculo Liso Vascular/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Artérias/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo L , Sinalização do Cálcio/efeitos dos fármacos , Corantes Fluorescentes , Técnicas In Vitro , Indóis/farmacologia , Masculino , Microscopia Confocal , Tono Muscular/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Nifedipino/farmacologia , Fenilefrina/farmacologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Estimulação Química , Resistência Vascular/efeitos dos fármacos , Resistência Vascular/fisiologia , Vasoconstrição/fisiologia , Vasodilatadores/farmacologia
13.
Circ Res ; 84(4): 424-34, 1999 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-10066677

RESUMO

To investigate the cellular mechanisms for altered Ca2+ homeostasis and contractility in cardiac hypertrophy, we measured whole-cell L-type Ca2+ currents (ICa,L), whole-cell Ca2+ transients ([Ca2+]i), and Ca2+ sparks in ventricular cells from 6-month-old spontaneously hypertensive rats (SHRs) and from age- and sex-matched Wistar-Kyoto and Sprague-Dawley control rats. By echocardiography, SHR hearts had cardiac hypertrophy and enhanced contractility (increased fractional shortening) and no signs of heart failure. SHR cells had a voltage-dependent increase in peak [Ca2+]i amplitude (at 0 mV, 1330+/-62 nmol/L [SHRs] versus 836+/-48 nmol/L [controls], P<0.05) that was not associated with changes in ICa,L density or kinetics, resting [Ca2+]i, or Ca2+ content of the sarcoplasmic reticulum (SR). SHR cells had increased time of relaxation. Ca2+ sparks from SHR cells had larger average amplitudes (173+/-192 nmol/L [SHRs] versus 109+/-64 nmol/L [control]; P<0.05), which was due to redistribution of Ca2+ sparks to a larger amplitude population. This change in Ca2+ spark amplitude distribution was not associated with any change in the density of ryanodine receptors, calsequestrin, junctin, triadin 1, Ca2+-ATPase, or phospholamban. Therefore, SHRs with cardiac hypertrophy have increased contractility, [Ca2+]i amplitude, time to relaxation, and average Ca2+ spark amplitude ("big sparks"). Importantly, big sparks occurred without alteration in the trigger for SR Ca2+ release (ICa,L), SR Ca2+ content, or the expression of several SR Ca2+-cycling proteins. Thus, cardiac hypertrophy in SHRs is linked with an alteration in the coupling of Ca2+ entry through L-type Ca2+ channels and the release of Ca2+ from the SR, leading to big sparks and enhanced contractility. Alterations in the microdomain between L-type Ca2+ channels and SR Ca2+ release channels may underlie the changes in Ca2+ homeostasis observed in cardiac hypertrophy. Modulation of SR Ca2+ release may provide a new therapeutic strategy for cardiac hypertrophy and for its progression to heart failure and sudden death.


Assuntos
Cardiomegalia/fisiopatologia , Contração Miocárdica/fisiologia , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Canais de Cálcio/fisiologia , Cardiomegalia/patologia , Condutividade Elétrica , Coração/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Concentração Osmolar , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
14.
J Physiol ; 512 ( Pt 3): 669-76, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9769412

RESUMO

1. Confocal laser scanning microscopy was used to visualize intercellular transmission of Ca2+ waves in intact rat ventricular trabeculae micro-injected with the calcium indicator fluo-3. 2. Ca2+ waves usually failed to be transmitted from cell to cell. At identified individual end-to-end cell contacts, successful transmission interspersed with failure, which sometimes occurred despite an apparent small spritz of Ca2+ between cells. The probability of cell to cell transmission (Ptran) was 0.13. 3. Ca2+ waves arose preferentially near junctions of connected cells, where connexin-43 was found, but randomly in enzymatically disconnected heart cells. 4. beta-Adrenergic stimulation significantly increased Ptran (to 0.22) and heptanol, an uncoupler of gap junction channels, significantly decreased it (to 0.045). 5. In regions of high [Ca2+]i due to damage, wave frequency decreased markedly with each cell-cell junction passed. 6. The Ca2+ permeability of cardiac gap junctions may be regulated, and the low ability of cardiac gap junctions to transmit Ca2+ may help control the spread of Ca2+ from damaged regions.


Assuntos
Cálcio/metabolismo , Miocárdio/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Compostos de Anilina , Animais , Comunicação Celular/fisiologia , Conexinas/metabolismo , Corantes Fluorescentes , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Coração/fisiologia , Processamento de Imagem Assistida por Computador , Técnicas In Vitro , Microscopia Confocal , Ratos , Ratos Endogâmicos BN , Desacopladores/farmacologia , Xantenos
15.
Biophys J ; 75(3): 1144-62, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9726917

RESUMO

A difficulty of using confocal microscopy to study Ca2+ sparks is the uncertainty of the linescan position with respect to the source of Ca2+ release. Random placement of the linescan is expected to result in a broad distribution of measured Ca2+ spark amplitudes (a) even if all Ca2+ sparks were generated identically. Thus variations in Ca2+ spark amplitude due to positional differences between confocal linescans and Ca2+ release site are intertwined with variations due to intrinsic differences in Ca2+ release properties. To separate these two sources of variations on the Ca2+ spark amplitude, we determined the effect changes of channel current or channel open time--collectively called the source strength, alpha--had on the measured Ca2+ spark amplitude histogram, N(a). This was done by 1) simulating Ca2+ release, Ca2+ and fluo-3 diffusion, and Ca2+ binding reactions; 2) simulation of image formation of the Ca2+ spark by a confocal microscope; and 3) using a novel automatic Ca2+ spark detector. From these results we derived an integral equation relating the probability density function of source strengths, f alpha (alpha), to N(a), which takes into account random positional variations between the source and linescan. In the special, but important, case that the spatial distribution of Ca(2+)-bound fluo-3 is Gaussian, we show the following: 1) variations of Ca2+ spark amplitude due to positional or intrinsic differences can be separated, and 2) f alpha (alpha) can, in principle, be calculated from the Ca2+ spark amplitude histogram since N(a) is the sum of shifted hyperbolas, where the magnitudes of the shifts and weights depend on f alpha (alpha). In particular, if all Ca2+ sparks were generated identically, then the plot of 1/N(a) against a will be a straight line. Multiple populations of channels carrying distinct currents are revealed by discontinuities in the 1/N(a) plot. 3) Although the inverse relationship between Ca2+ spark amplitude and decay time might be used to distinguish Ca2+ sparks from different channel populations, noise can render the measured decay times meaningless for small amplitude Ca2+ sparks.


Assuntos
Cálcio/metabolismo , Modelos Biológicos , Compostos de Anilina , Animais , Fenômenos Biofísicos , Biofísica , Canais de Cálcio/metabolismo , Corantes Fluorescentes , Microscopia Confocal/métodos , Microscopia Confocal/estatística & dados numéricos , Modelos Cardiovasculares , Miocárdio/citologia , Miocárdio/metabolismo , Retículo Sarcoplasmático/metabolismo , Xantenos
17.
Circ Res ; 82(4): 424-9, 1998 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-9506702

RESUMO

The goal of this study was to examine the relationship between Ca2+ entry through L-type Ca2+ channels and local [Ca2+]i transients (Ca2+ sparks) in single rat cardiac ventricular cells. L-type Ca2+ channels were activated by depolarization of cell-attached membrane patches, and [Ca2+]i was measured simultaneously as fluo 3 fluorescence using laser scanning confocal microscopy. Patch depolarization with Ca2+ as the charge carrier (10 or 110 mmol.L(-1)) significantly increased the probability of the occurrence of Ca2+ sparks (Ca2+ spark rate) only in the volume of cytoplasm located immediately beneath the membrane patch (basal Ca2+ spark rate, 119 Ca2+ sparks.cell(-1).s(-1); patch depolarization Ca2+ spark rate, 610 Ca2+ sparks.cell(-1).s(-1); P<.005). With Ba2+ in the pipette solution (10 mmol.L(-1)), patch depolarization was not associated with an increased Ca2+ spark rate at the position of the pipette or at any other sites distant from the pipette. Therefore, Ca2+ entry and not voltage per se was a necessary event for the occurrence of Ca2+ sparks. Under identical experimental conditions, patch depolarization experiments opened single L-type Ca2+ channels with a single-channel conductance of 19 pS with Ba2+ as the charge carrier. Although single-channel openings could not be resolved when Ca2+ was the charge carrier, ensemble averages yielded an inward current of up to 0.75 pA. The results suggest that voltage-activated Ca2+ entry through one or a small number of L type Ca2+ channels triggers the release of Ca2+ only from the sarcoplasmic reticulum in direct proximity to those L-type Ca2+ channels. The relatively low probability of triggering Ca2+ sparks may have resulted from some alteration of excitation-contraction coupling associated with the technique of the cell-attached patch clamp.


Assuntos
Cálcio/fisiologia , Coração/fisiologia , Animais , Membrana Celular/fisiologia , Condutividade Elétrica , Ativação do Canal Iônico , Potenciais da Membrana , Microscopia Confocal , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sarcolema/fisiologia , Retículo Sarcoplasmático/fisiologia
18.
Circ Res ; 81(4): 462-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9314826

RESUMO

The [Ca2+]i transient in heart is now thought to involve the recruitment and summation of discrete and independent "units" of Ca2+ release (Ca2+ "sparks") from the sarcoplasmic reticulum, each of which is controlled locally by single coassociated L-type Ca2+ channels ("local control theory of excitation-contraction coupling"). All prior studies on Ca2+ sparks, however, have been performed in single enzymatically dissociated heart cells under nonphysiological conditions. In order to understand the possible significance of Ca2+ sparks to normal working cardiac muscle, we used confocal microscopy to record Ca2+ sparks, spatially averaged [Ca2+]i transients and Ca2+ waves in individual cells of intact rat right ventricular trabeculae (composed of < 15 cells in cross section) microinjected with the Ca2+ indicator fluo 3 under physiological conditions ([Ca2+]o, 1 mmol/L; temperature, 33 +/- 1 degree C). Twitch force was recorded simultaneously. When stretched to optimal length (sarcomere length, 2.2 microns) and stimulated at 0.2 Hz, the trabeculae generated approximately equal to 700 micrograms of force per cell. Spatially averaged [Ca2+]i transients recorded from individual cells within a trabecula were similar to those recorded previously from single cells. The amplitude distribution of the peak ratio of Ca2+ sparks was bimodal, with maxima at ratios of 1.8 +/- 0.3 and 2.7 +/- 0.2 (mean +/- SD), respectively. The amplitude of the peak of Ca2+ sparks was approximately equal to 170 nmol/L. Ca2+ sparks occurred at a frequency of 12.0 +/- 0.8/s (mean +/- SEM) in line scans covering 94 sarcomeres. Ca2+ waves occurred randomly at a frequency of 0.57 +/- 0.08/s and propagated with a velocity of 29.5 +/- 1.7 microns/s. The extent of Ca2+ wave propagation was 3.9 +/- 0.3 sarcomere lengths (sarcomere length, 2.2 microns). Ca2+ sparks could be identified along the leading edge of the waves at intervals of 1.30 +/- 0.11 sarcomere length. Our observations suggest that (1) Ca2+ sparks, similar to those recorded in single cells, occur in trabeculae under physiological conditions and (2) coupling of Ca2+ spark generation between neighboring sites occurs and may lead to (3) the development of Ca2+ waves, which propagate under physiological conditions at a low velocity over limited distances. The results suggest that concepts of excitation-contraction coupling recently derived from isolated myocytes are applicable to intact cardiac trabeculae [corrected].


Assuntos
Cálcio/metabolismo , Miocárdio/metabolismo , Compostos de Anilina , Animais , Corantes Fluorescentes , Ventrículos do Coração , Microscopia Confocal , Contração Miocárdica , Ratos , Ratos Endogâmicos BN , Função Ventricular , Xantenos
19.
Cell Calcium ; 21(6): 441-52, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9223680

RESUMO

We describe the construction of a high-resolution confocal laser-scanning microscope, and illustrate its use for studying elementary Ca2+ signalling events in cells. An avalanche photodiode module and simple optical path provide a high efficiency system for detection of fluorescence signals, allowing use of a small confocal aperture giving near diffraction-limited spatial resolution (< 300 nm lateral and < 400 nm axial). When operated in line-scan mode, the maximum temporal resolution is 1 ms, and the associated computer software allows complete flexibility to record line-scans continuously for long (minutes) periods or to obtain any desired pixel resolution in x-y scans. An independent UV irradiation system permits simultaneous photolysis of caged compounds over either a uniform, wide field (arc lamp source) or at a tightly focussed spot (frequency-tripled Nd:YAG laser). The microscope thus provides a versatile tool for optical studies of dynamic cellular processes, as well as excellent resolution for morphological studies. The confocal scanner can be added to virtually any inverted microscope for a component cost that is only a small fraction of that of comparable commercial instruments, yet offers better performance and greater versatility.


Assuntos
Microscopia Confocal/instrumentação , Microscopia Confocal/métodos , Animais , Cálcio/análise , Cálcio/farmacocinética , Feminino , Processamento de Imagem Assistida por Computador/instrumentação , Processamento de Imagem Assistida por Computador/métodos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Oócitos/química , Oócitos/fisiologia , Óptica e Fotônica/instrumentação , Software , Xenopus
20.
J Physiol ; 505 ( Pt 2): 337-44, 1997 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-9423177

RESUMO

1. High spatial resolution confocal imaging was used to investigate spontaneous calcium release events (Ca2+ sparks) in isolated rat cardiac myocytes loaded with the fluorescent calcium indicator fluo-3. 2. Frequencies of sparks at different release sites varied widely, with a few sites showing sustained activities as great as 50 times the average. Sites with frequent sparks showed more rapid recovery of activity following Ca2+ waves and locally elevated basal [Ca2+]. 3. In addition to transient sparks, some sites showed bursts of rapid flurries of spark-like events, or apparently sustained Ca2+ liberation. Bursts remained localized to individual z-lines, but adjacent sites on the same z-line could be 'driven' by a bursting site to generate similar activity. 4. Individual sites showed long-term (tens of seconds) changes in 'modes' of activity, with abrupt transitions in frequencies of sparking, and between transient sparks and sustained bursts. These transitions were not associated with changes in the amplitude of the sparks. 5. We conclude that spontaneous sparks are not stereotyped events generated with uniform probability at all sites. Instead, the Ca2+ release event in variable, and some sites have a high probability of spark generation. Both factors show long-term changes at individual sites, raising the possibility that properties of fundamental Ca2+ release units may be subject to modulation.


Assuntos
Cálcio/metabolismo , Coração/fisiologia , Miocárdio/metabolismo , Compostos de Anilina , Animais , Corantes Fluorescentes , Ventrículos do Coração , Técnicas In Vitro , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Xantenos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA