Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 72018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29968564

RESUMEN

Genetically encoded fluorescent biosensors have revolutionized the study of signal transduction by enabling the real-time tracking of signaling activities in live cells. Investigating the interaction between signaling networks has become increasingly important to understanding complex cellular phenomena, necessitating an update of the biosensor toolkit to allow monitoring and perturbing multiple activities simultaneously in the same cell. We therefore developed a new class of fluorescent biosensors based on homo-FRET, deemed FLuorescence Anisotropy REporters (FLAREs), which combine the multiplexing ability of single-color sensors with a quantitative, ratiometric readout. Using an array of color variants, we were able to demonstrate multiplexed imaging of three activity reporters simultaneously in the same cell. We further demonstrate the compatibility of FLAREs for use with optogenetic tools as well as intravital two-photon imaging.


Asunto(s)
Técnicas Biosensibles , Polarización de Fluorescencia/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Colorantes Fluorescentes/metabolismo , Transducción de Señal , Análisis de la Célula Individual/métodos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Color , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/metabolismo , Citosol/ultraestructura , Transferencia Resonante de Energía de Fluorescencia/instrumentación , Colorantes Fluorescentes/síntesis química , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Transfección , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Proteína Fluorescente Roja
2.
Auton Neurosci ; 207: 59-66, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28781164

RESUMEN

We review the information that has been provided by optical imaging experiments directed at understanding the role and effects of sympathetic nerve activity (SNA) in the functioning of blood vessels. Earlier studies utilized electric field stimulation of nerve terminals (EFS) in isolated arteries and vascular tissues (ex vivo) to elicit SNA, but more recently, imaging studies have been conducted in vivo, enabling the study of SNA in truly physiological conditions. Ex vivo: In vascular smooth muscle cells (VSMC) of isolated arteries, the three sympathetic neurotransmitters, norepinephrine (NE), ATP and neuropeptide Y (NPY), elicit or modulate distinct patterns of Ca2+ signaling, as revealed by confocal imaging of exogenous fluorescent Ca2+ indicators. Purinergic junctional Ca2+ transients (jCaTs) arise from Ca2+ influx during excitatory junction potentials (eJPs), and are associated with the initial neurogenic contraction. Adrenergic Ca2+ waves and oscillations cause contraction while SNA-induced endothelial Ca2+ 'pulsars' cause relaxation. In vivo: optical biosensor mice, which express genetically encoded Ca2+ indicators (GECI's) specifically in smooth muscle, combined with non-invasive imaging techniques has enabled imaging SNA-induced Ca2+ signaling and arterial diameter in vivo. SNA induces Ca2+ oscillations in intact arteries. [Ca2+] of arterial smooth muscle cells increased in hypertension, in association with increased SNA. High resolution imaging has revealed local sympathetic, neurogenic Ca2+ signaling within smooth muscle and endothelial cells of the vasculature. The ongoing development of in vivo imaging together with an expanding availability of different biosensor animals promises to enable the further assessment of SNA and its effects in the vasculature of living animals.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Humanos , Neurogénesis/fisiología , Vasoconstricción/fisiología
3.
J Control Release ; 238: 139-148, 2016 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-27460683

RESUMEN

Therapeutic nanoparticles (NPs) approved for clinical use in solid tumor therapy provide only modest improvements in patient survival, in part due to physiological barriers that limit delivery of the particles throughout the entire tumor. Here, we explore the thresholds for NP size and surface poly(ethylene glycol) (PEG) density for penetration within tumor tissue extracellular matrix (ECM). We found that NPs as large as 62nm, but less than 110nm in diameter, diffused rapidly within a tumor ECM preparation (Matrigel) and breast tumor xenograft slices ex vivo. Studies of PEG-density revealed that increasing PEG density enhanced NP diffusion and that PEG density below a critical value led to adhesion of NP to ECM. Non-specific binding of NPs to tumor ECM components was assessed by surface plasmon resonance (SPR), which revealed excellent correlation with the particle diffusion results. Intravital microscopy of NP spread in breast tumor tissue confirmed a significant difference in tumor tissue penetration between the 62 and 110nm PEG-coated NPs, as well as between PEG-coated and uncoated NPs. SPR assays also revealed that Abraxane, an FDA-approved non-PEGylated NP formulation used for cancer therapy, binds to tumor ECM. Our results establish limitations on the size and surface PEG density parameters required to achieve uniform and broad dispersion within tumor tissue and highlight the utility of SPR as a high throughput method to screen NPs for tumor penetration.


Asunto(s)
Portadores de Fármacos/metabolismo , Nanopartículas/metabolismo , Neoplasias/metabolismo , Polietilenglicoles/metabolismo , Paclitaxel Unido a Albúmina/administración & dosificación , Paclitaxel Unido a Albúmina/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Mama/efectos de los fármacos , Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Colágeno/metabolismo , Difusión , Doxorrubicina/administración & dosificación , Doxorrubicina/análogos & derivados , Doxorrubicina/metabolismo , Portadores de Fármacos/análisis , Combinación de Medicamentos , Femenino , Humanos , Ácido Láctico/análisis , Ácido Láctico/metabolismo , Laminina/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/análisis , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Polietilenglicoles/administración & dosificación , Polietilenglicoles/análisis , Ácido Poliglicólico/análisis , Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Proteoglicanos/metabolismo , Propiedades de Superficie
4.
Am J Physiol Heart Circ Physiol ; 308(12): H1517-24, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25888510

RESUMEN

Myogenic tone is an intrinsic property of the vasculature that contributes to blood pressure control and tissue perfusion. Earlier investigations assigned a key role in myogenic tone to phospholipase C (PLC) and its products, inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). Here, we used the PLC inhibitor, U-73122, and two other, specific inhibitors of PLC subtypes (PI-PLC and PC-PLC) to delineate the role of PLC in myogenic tone of pressurized murine mesenteric arteries. U-73122 inhibited depolarization-induced contractions (high external K(+) concentration), thus confirming reports of nonspecific actions of U-73122 and its limited utility for studies of myogenic tone. Edelfosine, a specific inhibitor of PI-PLC, did not affect depolarization-induced contractions but modulated myogenic tone. Because PI-PLC produces IP3, we investigated the effect of blocking IP3 receptor-mediated Ca(2+) release on myogenic tone. Incubation of arteries with xestospongin C did not affect tone, consistent with the virtual absence of Ca(2+) waves in arteries with myogenic tone. D-609, an inhibitor of PC-PLC and sphingomyelin synthase, strongly inhibited myogenic tone and had no effect on depolarization-induced contraction. D-609 appeared to act by lowering cytoplasmic Ca(2+) concentration to levels below those that activate contraction. Importantly, incubation of pressurized arteries with a membrane-permeable analog of DAG induced vasoconstriction. The results therefore mandate a reexamination of the signaling pathways activated by the Bayliss mechanism. Our results suggest that PI-PLC and IP3 are not required in maintaining myogenic tone, but DAG, produced by PC-PLC and/or SM synthase, is likely through multiple mechanisms to increase Ca(2+) entry and promote vasoconstriction.


Asunto(s)
Músculo Liso Vascular/enzimología , Transducción de Señal , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Fosfolipasas de Tipo C/metabolismo , Resistencia Vascular , Vasoconstricción , Animales , Presión Arterial , Señalización del Calcio/efectos de los fármacos , Diglicéridos/metabolismo , Inhibidores Enzimáticos/farmacología , Arterias Mesentéricas/enzimología , Ratones , Músculo Liso Vascular/efectos de los fármacos , Fosfatidilcolinas/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/antagonistas & inhibidores , Fosfolipasas de Tipo C/antagonistas & inhibidores , Resistencia Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos
5.
Front Physiol ; 5: 387, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25339912

RESUMEN

UNLABELLED: Two-photon fluorescence microscopy and conscious, restrained optical biosensor mice were used to study smooth muscle Ca(2+) signaling in ear arterioles. Conscious mice were used in order to preserve normal mean arterial blood pressure (MAP) and sympathetic nerve activity (SNA). ExMLCK mice, which express a genetically-encoded smooth muscle-specific FRET-based Ca(2+) indicator, were equipped with blood pressure telemetry and immobilized for imaging. MAP was 101 ± 4 mmHg in conscious restrained mice, similar to the freely mobile state (107 ± 3 mmHg). Oscillatory vasomotion or irregular contractions were observed in most arterioles (71%), with the greatest oscillatory frequency observed at 0.25 s(-1). In a typical arteriole with an average diameter of ~35 µm, oscillatory vasomotion of a 5-6 µm magnitude was accompanied by nearly uniform [Ca(2+)] oscillations from ~0.1 to 0.5 µM, with maximum [Ca(2+)] occurring immediately before the rapid decrease in diameter. Very rapid, spatially uniform "Ca(2+) flashes" were also observed but not asynchronous propagating Ca(2+) waves. In contrast, vasomotion and dynamic Ca(2+) signals were rarely observed in ear arterioles of anesthetized exMLCK biosensor mice. Hexamethonium (30 µg/g BW, i.p.) caused a fall in MAP to 74 ± 4 mmHg, arteriolar vasodilation, and abolition of vasomotion and synchronous Ca(2+) transients. SUMMARY: MAP and heart rate (HR) were normal during high-resolution Ca(2+) imaging of conscious, restrained mice. SNA induced continuous vasomotion and irregular vasoconstrictions via spatially uniform Ca(2+) signaling within the arterial wall. FRET-based biosensor mice and two-photon imaging provided the first measurements of [Ca(2+)] in vascular smooth muscle cells in arterioles of conscious animals.

6.
Am J Physiol Heart Circ Physiol ; 307(2): H173-81, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24858846

RESUMEN

We used two-photon (2-p) Förster resonance energy transfer (FRET) microscopy to provide serial, noninvasive measurements of [Ca(2+)] in arterioles of living "biosensor" mice. These express a genetically encoded Ca(2+) indicator (GECI), either FRET-based exMLCK or intensity-based GCaMP2. The FRET ratios, Rmin and Rmax, required for in vivo Ca(2+) calibration of exMLCK were obtained in isolated arteries. For in vivo experiments, mice were anesthetized (1.5% isoflurane), and arterioles within a depilated ear were visualized through the intact skin (i.e., noninvasively), by 2-p excitation of exMLCK (at 820 nm) or GCaMP2 (at 920 nm). Spontaneous or agonist-evoked [Ca(2+)] transients in arteriolar smooth muscle cells were imaged (at 2 Hz) with both exMLCK and GCaMP2. To examine changes in arteriolar [Ca(2+)] that might accompany hypertension, five exMLCK mice were implanted with telemetric blood pressure transducers and osmotic minipumps containing ANG II (350 ng·kg(-1)·min(-1)) and fed a high (6%)-salt diet for 9 days. [Ca(2+)] was measured every other day in five smooth muscle cells of two to three arterioles in each animal. Prior to ANG II/salt, [Ca(2+)] was 246 ± 42 nM. [Ca(2+)] increased transiently to 599 nM on day 2 after beginning ANG II/salt, then remained elevated at 331 ± 42 nM for 4 more days, before returning to 265 ± 47 nM 6 days after removal of ANG II/salt. In summary, two-photon excitation of exMLCK and GCaMP2 provides a method for noninvasive, longitudinal quantification of [Ca(2+)] dynamics and vascular structure in individual arterioles of a particular animal over an extended period of time, a capability that should enhance future studies of hypertension and vascular function.


Asunto(s)
Técnicas Biosensibles , Señalización del Calcio , Calcio/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Hipertensión/metabolismo , Microscopía de Fluorescencia por Excitación Multifotónica , Músculo Liso Vascular/metabolismo , Piel/irrigación sanguínea , Angiotensina II , Animales , Presión Arterial , Arteriolas/metabolismo , Arteriolas/fisiopatología , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Ratones , Ratones Transgénicos , Músculo Liso Vascular/fisiopatología , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Cloruro de Sodio Dietético , Factores de Tiempo
7.
PLoS One ; 8(6): e65969, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23776582

RESUMEN

BACKGROUND AND PURPOSE: Determining the role of vascular receptors in vivo is difficult and not readily accomplished by systemic application of antagonists or genetic manipulations. Here we used intravital microscopy to measure the contributions of sympathetic receptors, particularly α1-adrenoceptor subtypes, to contractile activation of femoral artery in vivo. EXPERIMENTAL APPROACH: Diameter and intracellular calcium ([Ca(2+)]i) in femoral arteries were determined by intravital fluorescence microscopy in mice expressing a Myosin Light Chain Kinase (MLCK) based calcium-calmodulin biosensor. Pharmacological agents were applied locally to the femoral artery to determine the contributions of vascular receptors to tonic contraction and [Ca(2+)]i,. KEY RESULTS: In the anesthetized animal, femoral arteries were constricted to a diameter equal to 54% of their passive diameter (i.e. tone = 46%). Of this total basal tone, 16% was blocked by RS79948 (0.1 µM) and thus attributable to α2-adrenoceptors. A further 46% was blocked by prazosin (0.1 µM) and thus attributable to α1-adrenoceptors. Blockade of P2X and NPY1 receptors with suramin (0.5 mM) and BIBP3226 (1.0 µM) respectively, reduced tone by a further 22%, leaving 16% of basal tone unaffected at these concentrations of antagonists. Application of RS100329 (α1A-selective antagonist) and BMY7378 (α1D-selective) decreased tone by 29% and 26%, respectively, and reduced [Ca(2+)]i. Chloroethylclonidine (1 µM preferential for α1B-) had no effect. Abolition of sympathetic nerve activity (hexamethonium, i.p.) reduced basal tone by 90%. CONCLUSION AND IMPLICATIONS: Tone of mouse femoral arteries in vivo is almost entirely sympathetic in origin. Activation of α1A- and α1D-adrenoceptors elevates [Ca(2+)]i and accounts for at least 55% of the tone.


Asunto(s)
Fibras Adrenérgicas/fisiología , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Arteria Femoral/inervación , Arteria Femoral/fisiología , Tono Muscular/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Arginina/análogos & derivados , Calcio/metabolismo , Isoquinolinas , Ratones , Microscopía Fluorescente , Tono Muscular/fisiología , Músculo Liso Vascular/fisiología , Miografía , Quinasa de Cadena Ligera de Miosina , Naftiridinas , Piperazinas , Prazosina , Suramina , Timina
8.
Microcirculation ; 20(3): 269-77, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23140521

RESUMEN

OBJECTIVES: We sought to determine some of the molecular requirements for basal state "tone" of skeletal muscle arterioles in vivo, and whether asynchronous Ca(2+) waves are involved or not. METHODS: Cremaster muscles of anesthetized exMLCK and smGCaMP2 biosensor mice were exteriorized, and the fluorescent arterioles were visualized with wide-field, confocal or multiphoton microscopy to observe Ca(2+) signaling and arteriolar diameter. RESULTS: Basal state tone of the arterioles was ~50%. Local block of Ang-II receptors (AT1 ) or α1 -adrenoceptors (α1 -AR) had no effect on diameter, nor did complete block of sympathetic nerve activity (SNA). Inhibition of phospholipase C caused dilation nearly to the Ca(2+) -free (passive) diameter, as did exposure to nifedipine or 2-APB. Arterioles were also dilated when treated with SKF96365. High-resolution imaging of exMLCK fluorescence (ratio) or GCaMP2 fluorescence in smooth muscle cells failed to reveal Ca(2+) waves (although Ca(2+) waves/transients were readily detected by both biosensors in small arteries, ex vivo). CONCLUSIONS: Arterioles of cremaster muscle have vascular tone of ~ 50%, which is not due to α1 -AR, AT1 R, or SNA. PLC activity, L-type Ca(2+) channels, 2-APB- and SKF96365-sensitive channels are required. Propagating Ca(2+) waves are not present. A key role for PLC and InsP3 R in vascular tone in vivo, other than producing Ca(2+) waves, is suggested.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Músculo Esquelético/irrigación sanguínea , Animales , Arteriolas/citología , Arteriolas/metabolismo , Canales de Calcio/metabolismo , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Músculo Esquelético/citología
9.
PLoS One ; 7(10): e46316, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071553

RESUMEN

A-kinase anchoring proteins (AKAPs) bind the regulatory subunits of protein kinase A (PKA) and localize the holoenzyme to discrete signaling microdomains in multiple subcellular compartments. Despite emerging evidence for a nuclear pool of PKA that rapidly responds to activation of the PKA signaling cascade, only a few AKAPs have been identified that localize to the nucleus. Here we show a PKA-binding domain in the amino terminus of Chd8, and demonstrate subcellular colocalization of Chd8 with RII. RII overlay and immunoprecipitation assays demonstrate binding between Chd8-S and RIIα. Binding is abrogated upon dephosphorylation of RIIα. By immunofluorescence, we identified nuclear and perinuclear pools of Chd8 in HeLa cells and rat neonatal cardiomyocytes. We also show high levels of Chd8 mRNA in RNA extracted from post-natal rat hearts. These data add Chd8 to the short list of known nuclear AKAPs, and implicate a function for Chd8 in post-natal rat cardiac development.


Asunto(s)
Proteínas Portadoras/metabolismo , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Femenino , Humanos , Inmunoprecipitación , Microscopía Confocal , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas Nucleares/química , Fosforilación , Embarazo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Fracciones Subcelulares/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 297(3): H1140-50, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19617413

RESUMEN

Prolonged ouabain administration to normal rats causes sustained blood pressure (BP) elevation. This ouabain-induced hypertension (OH) has been attributed, in part, to the narrowing of third-order resistance arteries (approximately 320 microm internal diameter) as a result of collagen deposition in the artery media. Here we describe the structural and functional properties of fourth-order mesenteric small arteries from control and OH rats, including the effect of low-dose ouabain on myogenic tone in these arteries. Systolic BP in OH rats was 138 +/- 3 versus 124 +/- 4 mmHg in controls (P < 0.01). Pressurized (70 mmHg) control and OH arteries, with only a single layer of myocytes, both had approximately 165-microm internal diameters and approximately 20-microm wall thicknesses. Even after fixation, despite vasoconstriction, the diameters and wall thicknesses did not differ between control and OH fourth-order arteries, whereas in third-order arteries, both parameters were significantly smaller in OH than in controls. Myogenic reactivity was significantly augmented in OH fourth-order arteries. Nevertheless, phenylephrine- (1 microM) and high K(+)-induced vasoconstrictions and acetylcholine-induced vasodilation were comparable in control and OH arteries. Vasoconstrictions induced by 5 microM phenylephrine and by 10 mM caffeine in Ca(2+)-free media indicated that releasable sarcoplasmic reticulum Ca(2+) stores were normal in OH arteries. Importantly, 100 nM ouabain constricted both control and OH arteries by approximately 26 microm, indicating that this response was not downregulated in OH rats. This maximal ouabain-induced constriction corresponds to a approximately 90% increase in resistance to flow in these small arteries; thus ouabain at EC(50) of approximately 0.66 nM should raise resistance by approximately 35%. We conclude that dynamic constriction in response to circulating nanomolar ouabain in small arteries likely makes a major contribution to the increased vascular tone and BP in OH rats.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Ouabaína/farmacología , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología , Acetilcolina/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cafeína/farmacología , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Fenilefrina/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Retículo Sarcoplasmático/metabolismo , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
11.
Am J Physiol Cell Physiol ; 290(2): C325-36, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16162658

RESUMEN

Agonist stimulation of human pulmonary artery smooth muscle cells (PASMC) and endothelial cells (PAEC) with histamine showed similar spatiotemporal patterns of Ca(2+) release. Both sustained elevation and oscillatory patterns of changes in cytosolic Ca(2+) concentration ([Ca(2+)](cyt)) were observed in the absence of extracellular Ca(2+). Capacitative Ca(2+) entry (CCE) was induced in PASMC and PAEC by passive depletion of intracellular Ca(2+) stores with 10 microM cyclopiazonic acid (CPA; 15-30 min). The pyrazole derivative BTP2 inhibited CPA-activated Ca(2+) influx, suggesting that depletion of CPA-sensitive internal stores is sufficient to induce CCE in both PASMC and PAEC. The recourse of histamine-mediated Ca(2+) release was examined after exposure of cells to CPA, thapsigargin, caffeine, ryanodine, FCCP, or bafilomycin. In PASMC bathed in Ca(2+)-free solution, treatment with CPA almost abolished histamine-induced rises in [Ca(2+)](cyt). In PAEC bathed in Ca(2+)-free solution, however, treatment with CPA eliminated histamine-induced sustained and oscillatory rises in [Ca(2+)](cyt) but did not affect initial transient increase in [Ca(2+)](cyt). Furthermore, treatment of PAEC with a combination of CPA (or thapsigargin) and caffeine (and ryanodine), FCCP, or bafilomycin did not abolish histamine-induced transient [Ca(2+)](cyt) increases. These observations indicate that 1) depletion of CPA-sensitive stores is sufficient to cause CCE in both PASMC and PAEC; 2) induction of CCE in PAEC does not require depletion of all internal Ca(2+) stores; 3) the histamine-releasable internal stores in PASMC are mainly CPA-sensitive stores; 4) PAEC, in addition to a CPA-sensitive functional pool, contain other stores insensitive to CPA, thapsigargin, caffeine, ryanodine, FCCP, and bafilomycin; and 5) although the CPA-insensitive stores in PAEC may not contribute to CCE, they contribute to histamine-mediated Ca(2+) release.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Células Endoteliales/metabolismo , Histamina/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Pulmonar/citología , Adolescente , Adulto , Animales , Cafeína/farmacología , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Indoles/farmacología , Ionóforos/farmacología , Macrólidos/farmacología , Masculino , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Arteria Pulmonar/metabolismo , Tapsigargina/farmacología
12.
J Appl Physiol (1985) ; 98(1): 415-20, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15591311

RESUMEN

Acute hypoxia induces pulmonary vasoconstriction and chronic hypoxia causes structural changes of the pulmonary vasculature including arterial medial hypertrophy. Electro- and pharmacomechanical mechanisms are involved in regulating pulmonary vasomotor tone, whereas intracellular Ca(2+) serves as an important signal in regulating contraction and proliferation of pulmonary artery smooth muscle cells. Herein, we provide a basic overview of the cellular mechanisms involved in the development of hypoxic pulmonary vasoconstriction. Our discussion focuses on the roles of ion channels permeable to K(+) and Ca(2+), membrane potential, and cytoplasmic Ca(2+) in the development of acute hypoxic pulmonary vasoconstriction and chronic hypoxia-mediated pulmonary vascular remodeling.


Asunto(s)
Calcio/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Activación del Canal Iónico , Canales Iónicos/metabolismo , Arteria Pulmonar/fisiopatología , Vasoconstricción , Animales , Hemostasis , Humanos , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Pulmón/irrigación sanguínea , Pulmón/fisiopatología , Modelos Biológicos , Oxígeno/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 287(2): H608-16, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15059779

RESUMEN

The possible roles of endothelial intracellular Ca(2+) concentration ([Ca(2+)](i)), nitric oxide (NO), arachidonic acid (AA) metabolites, and Ca(2+)-activated K(+) (K(Ca)) channels in adrenergically induced vasomotion were examined in pressurized rat mesenteric arteries. Removal of the endothelium or buffering [Ca(2+)](i) selectively in endothelial cells with BAPTA eliminated vasomotion in response to phenylephrine (PE; 10.0 microM). In arteries with intact endothelium, inhibition of NO synthase with N(omega)-nitro-l-arginine methyl ester (l-NAME; 300.0 microM) or N(omega)-nitro-l-arginine (l-NNA; 300.0 microM) did not eliminate vasomotion. Neither inhibition of cGMP formation with 10.0 microM 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) nor inhibition of prostanoid formation (10.0 microM indomethacin) eliminated vasomotion. Similarly, inhibition of AA cytochrome P-450 metabolism with an intraluminal application of 17-octadecynoic acid (17-ODYA) or 6-(2-propargyloxyphenyl)hexanoic acid (PPOH) failed to eliminate vasomotion. In contrast, intraluminal application of the K(Ca) channel blockers apamin (250.0 nM) and charybdotoxin (100.0 nM), together, abolished vasomotion and changed synchronous Ca(2+) oscillations in smooth muscle cells to asynchronous propagating Ca(2+) waves. Apamin, charybdotoxin, or iberiotoxin (100.0 nM) alone did not eliminate vasomotion, nor did the combination of apamin and iberiotoxin. The results show that adrenergic vasomotion in rat mesenteric arteries is critically dependent on Ca(2+)-activated K(+) channels in endothelial cells. Because these channels (small- and intermediate-conductance K(Ca) channels) are a recognized component of EDHF, we conclude therefore that EDHF is essential for the development of adrenergically induced vasomotion.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Factores Biológicos/fisiología , Ácido Egtácico/análogos & derivados , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Fenilefrina/farmacología , Sistema Vasomotor/fisiología , Animales , Apamina/farmacología , Ácido Araquidónico/metabolismo , Tampones (Química) , Calcio/metabolismo , Caribdotoxina/farmacología , Quelantes/farmacología , GMP Cíclico/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Técnicas Histológicas , Técnicas In Vitro , Masculino , Óxido Nítrico/fisiología , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA