Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Biol Chem ; 276(38): 35652-9, 2001 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-11470781

RESUMEN

Recycling endosomes in astrocytes show hormone-regulated, actin fiber-dependent delivery to the endosomal sorting pool. Recycling vesicle trafficking was followed in real time using a fusion protein composed of green fluorescent protein coupled to the 29-kDa subunit of the short-lived, membrane-bound enzyme type 2 deiodinase. Primary endosomes budded from the plasma membrane and oscillated near the cell periphery for 1-4 min. The addition of thyroid hormone triggered the processive, centripetal movement of the recycling vesicle in linear bursts at velocities of up to 200 nm/s. Vesicle migration was hormone-specific and blocked by inhibitors of actin polymerization and myosin ATPase. Domain mapping confirmed that the hormone-dependent vesicle-binding domain was located at the C terminus of the motor. In addition, the interruption of normal dimerization of native myosin 5a monomers inactivated vesicle transport, indicating that single-headed myosin 5a motors do not transport cargo in situ. This is the first demonstration of processive hormone-dependent myosin 5a movement in living cells.


Asunto(s)
Astrocitos/metabolismo , Movimiento Celular/fisiología , Miosinas/metabolismo , Actinas/fisiología , Animales , Animales Recién Nacidos , Astrocitos/fisiología , Secuencia de Bases , Cartilla de ADN , Miosinas/fisiología , Ratas
2.
J Physiol ; 531(Pt 2): 315-27, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11230506

RESUMEN

We recorded Ca2+ sparks and spontaneous transient outward currents (STOCs) simultaneously in smooth muscle cells using whole-cell patch recording and a unique, high-speed widefield digital imaging system to monitor fluo-3 fluorescence in both two and three dimensions (2D and 3D). In 2D imaging, the correlation between the amplitude of a spark and its corresponding STOC was a weak one, and 27 % of the sparks failed to cause STOCs. The STOCless sparks were not significantly different in amplitude from those that caused STOCs. Three-dimensional imaging disclosed that STOCless sparks were located close to the cell surface, and on average their apparent distance from the cell surface was not significantly different from the sparks that cause STOCs. Statistical evaluation of spark clusters disclosed that there were regions of the cell where the probability of spark occurrence was high and others where it was quite low.


Asunto(s)
Calcio/fisiología , Esófago/fisiología , Músculo Liso/fisiología , Compuestos de Anilina , Animales , Gatos , Membrana Celular/fisiología , Conductividad Eléctrica , Esófago/citología , Colorantes Fluorescentes , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Músculo Liso/citología , Técnicas de Placa-Clamp , Xantenos
3.
Br J Haematol ; 112(3): 644-54, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11260067

RESUMEN

Haematopoietic stem cells (HSCs) have been extensively characterized regarding in vivo engraftment, surface epitopes and genetic regulation. However, little is known about the homing of these rare cells, and their intrinsic motility and membrane deformation capacity. We used high-speed optical-sectioning microscopy and inverted fluorescent videomicroscopy to study highly purified murine lineage-negative, rhodamine-low, Hoechst-low HSCs over time under various in vitro conditions. We discovered extremely rapid motility, directed migration to stromal cells and marked membrane modulation. High resolution images with three-dimensional reconstruction showed the general presence of microspikes. Further, pseudopodia (proteopodia) were observed that were induced by stromal-derived factor-1 and steel factor. Proteopodia were directed towards and were quenched by stromal cells, at times bridged HSCs, and could rapidly retract or detach from cells. Proteopodia were also observed in vivo with homed HSCs in frozen sections of murine spleen, lung and heart. This is the first demonstration that HSCs are both fast and highly malleable in phenotype.


Asunto(s)
Extensiones de la Superficie Celular/ultraestructura , Células Madre Hematopoyéticas/fisiología , Animales , Células de la Médula Ósea , Adhesión Celular , Comunicación Celular , Movimiento Celular , Separación Celular , Extensiones de la Superficie Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Técnicas de Cocultivo , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/ultraestructura , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Microscopía Fluorescente , Microscopía por Video , Factor de Células Madre/farmacología
4.
Mol Biol Cell ; 12(1): 129-41, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11160828

RESUMEN

A novel imaging technology, high-speed microscopy, has been used to visualize the process of GLUT4 translocation in response to insulin in single 3T3-L1 adipocytes. A key advantage of this technology is that it requires extremely low light exposure times, allowing the quasi-continuous capture of information over 20-30 min without photobleaching or photodamage. The half-time for the accumulation of GLUT4-eGFP (enhanced green fluorescent protein) at the plasma membrane in a single cell was found to be of 5-7 min at 37 degrees C. This half-time is substantially longer than that of exocytic vesicle fusion in neuroendocrine cells, suggesting that additional regulatory mechanisms are involved in the stimulation of GLUT4 translocation by insulin. Analysis of four-dimensional images (3-D over time) revealed that, in response to insulin, GLUT4-eGFP-enriched vesicles rapidly travel from the juxtanuclear region to the plasma membrane. In nontransfected adipocytes, impairment of microtubule and actin filament function inhibited insulin-stimulated glucose transport by 70 and 50%, respectively. When both filament systems were impaired insulin-stimulated glucose transport was completely inhibited. Taken together, the data suggest that the regulation of long-range motility of GLUT4-containing vesicles through the interaction with microtubule- and actin-based cytoskeletal networks plays an important role in the overall effect of insulin on GLUT4 translocation.


Asunto(s)
Células 3T3/citología , Adipocitos/citología , Insulina/farmacología , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Citoesqueleto , Diagnóstico por Imagen/instrumentación , Diagnóstico por Imagen/métodos , Exocitosis/efectos de los fármacos , Transportador de Glucosa de Tipo 4 , Proteínas Fluorescentes Verdes , Semivida , Proteínas Luminiscentes/genética , Ratones , Microscopía Confocal/instrumentación , Microscopía Confocal/métodos , Proteínas de Transporte de Monosacáridos/genética , Transporte de Proteínas/efectos de los fármacos , Ratas , Proteínas Recombinantes de Fusión/genética , Vesículas Transportadoras
5.
J Gen Physiol ; 116(6): 845-64, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11099351

RESUMEN

Ca(2+) sparks are highly localized cytosolic Ca(2+) transients caused by a release of Ca(2+) from the sarcoplasmic reticulum via ryanodine receptors (RyRs); they are the elementary events underlying global changes in Ca(2+) in skeletal and cardiac muscle. In smooth muscle and some neurons, Ca(2+) sparks activate large conductance Ca(2+)-activated K(+) channels (BK channels) in the spark microdomain, causing spontaneous transient outward currents (STOCs) that regulate membrane potential and, hence, voltage-gated channels. Using the fluorescent Ca(2+) indicator fluo-3 and a high speed widefield digital imaging system, it was possible to capture the total increase in fluorescence (i.e., the signal mass) during a spark in smooth muscle cells, which is the first time such a direct approach has been used in any system. The signal mass is proportional to the total quantity of Ca(2+) released into the cytosol, and its rate of rise is proportional to the Ca(2+) current flowing through the RyRs during a spark (I(Ca(spark))). Thus, Ca(2+) currents through RyRs can be monitored inside the cell under physiological conditions. Since the magnitude of I(Ca(spark)) in different sparks varies more than fivefold, Ca(2+) sparks appear to be caused by the concerted opening of a number of RyRs. Sparks with the same underlying Ca(2+) current cause STOCs, whose amplitudes vary more than threefold, a finding that is best explained by variability in coupling ratio (i.e., the ratio of RyRs to BK channels in the spark microdomain). The time course of STOC decay is approximated by a single exponential that is independent of the magnitude of signal mass and has a time constant close to the value of the mean open time of the BK channels, suggesting that STOC decay reflects BK channel kinetics, rather than the time course of [Ca(2+)] decline at the membrane. Computer simulations were carried out to determine the spatiotemporal distribution of the Ca(2+) concentration resulting from the measured range of I(Ca(spark)). At the onset of a spark, the Ca(2+) concentration within 200 nm of the release site reaches a plateau or exceeds the [Ca(2+)](EC50) for the BK channels rapidly in comparison to the rate of rise of STOCs. These findings suggest a model in which the BK channels lie close to the release site and are exposed to a saturating [Ca(2+)] with the rise and fall of the STOCs determined by BK channel kinetics. The mechanism of signaling between RyRs and BK channels may provide a model for Ca(2+) action on a variety of molecular targets within cellular microdomains.


Asunto(s)
Señalización del Calcio , Calcio/fisiología , Membranas Intracelulares/fisiología , Canales de Potasio/fisiología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Bradiquinina/metabolismo , Bufo marinus , Citosol/metabolismo , Conductividad Eléctrica , Fluorescencia , Procesamiento de Imagen Asistido por Computador , Canales Iónicos/metabolismo , Cinética , Microscopía , Modelos Biológicos , Técnicas de Placa-Clamp
7.
J Biol Chem ; 275(44): 34512-20, 2000 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-10944522

RESUMEN

Myosin plays an important role in mitosis, especially during cytokinesis. Although it has been assumed that phosphorylation of regulatory light chain of myosin (RLC) controls motility of mammalian non-muscle cells, the functional significance of RLC phosphorylation remains uninvestigated. To address this problem, we have produced unphosphorylatable RLC (T18A/S19A RLC) and overexpressed it in COS-7 cells and normal rat kidney cells. Overexpression of T18A/S19A RLC but not wild type RLC almost completely abolished concanavalin A-induced receptor cap formation. The results indicate that myosin phosphorylation is critical for concanavalin A-induced gathering of surface receptors. T18A/S19A RLC overexpression resulted in the production of multinucleated cells, suggesting the failure of proper cell division in these cells. Video microscopic observation revealed that cells expressing T18A/S19A RLC showed abnormalities during mitosis in two respects. One is that the cells produced abnormal cleavage furrows, resulting in incomplete cytokinesis, which suggests that myosin phosphorylation is important for the normal recruitment of myosin molecules into the contractile ring structure. The other is that separation of chromosomes from the metaphase plate is disrupted in T18A/S19A RLC expressing cells, thus preventing proper transition from metaphase to anaphase. These results suggest that, in addition to cytokinesis, myosin and myosin phosphorylation play a role in the karyokinetic process.


Asunto(s)
Ciclo Celular , Miosinas/metabolismo , Animales , División Celular , Línea Celular , Células Cultivadas , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Miosinas/genética , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo
8.
Mol Biol Cell ; 11(6): 2047-56, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10848628

RESUMEN

Centrosome assembly is important for mitotic spindle formation and if defective may contribute to genomic instability in cancer. Here we show that in somatic cells centrosome assembly of two proteins involved in microtubule nucleation, pericentrin and gamma tubulin, is inhibited in the absence of microtubules. A more potent inhibitory effect on centrosome assembly of these proteins is observed after specific disruption of the microtubule motor cytoplasmic dynein by microinjection of dynein antibodies or by overexpression of the dynamitin subunit of the dynein binding complex dynactin. Consistent with these observations is the ability of pericentrin to cosediment with taxol-stabilized microtubules in a dynein- and dynactin-dependent manner. Centrosomes in cells with reduced levels of pericentrin and gamma tubulin have a diminished capacity to nucleate microtubules. In living cells expressing a green fluorescent protein-pericentrin fusion protein, green fluorescent protein particles containing endogenous pericentrin and gamma tubulin move along microtubules at speeds of dynein and dock at centrosomes. In Xenopus extracts where gamma tubulin assembly onto centrioles can occur without microtubules, we find that assembly is enhanced in the presence of microtubules and inhibited by dynein antibodies. From these studies we conclude that pericentrin and gamma tubulin are novel dynein cargoes that can be transported to centrosomes on microtubules and whose assembly contributes to microtubule nucleation.


Asunto(s)
Antígenos/metabolismo , Centrosoma/metabolismo , Dineínas/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Antígenos/genética , Células CHO , Células COS , Cricetinae , Citoplasma/metabolismo , Complejo Dinactina , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Xenopus
9.
Mol Biol Cell ; 11(5): 1815-27, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10793154

RESUMEN

This study was undertaken to obtain direct evidence for the involvement of gap junctions in the propagation of intercellular Ca(2+) waves. Gap junction-deficient HeLa cells were transfected with plasmids encoding for green fluorescent protein (GFP) fused to the cytoplasmic carboxyl termini of connexin 43 (Cx43), 32 (Cx32), or 26 (Cx26). The subsequently expressed GFP-labeled gap junctions rendered the cells dye- and electrically coupled and were detected at the plasma membranes at points of contact between adjacent cells. To correlate the distribution of gap junctions with the changes in [Ca(2+)](i) associated with Ca(2+) waves and the distribution of the endoplasmic reticulum (ER), cells were loaded with fluorescent Ca(2+)-sensitive (fluo-3 and fura-2) and ER membrane (ER-Tracker) dyes. Digital high-speed microscopy was used to collect a series of image slices from which the three-dimensional distribution of the gap junctions and ER were reconstructed. Subsequently, intercellular Ca(2+) waves were induced in these cells by mechanical stimulation with or without extracellular apyrase, an ATP-degrading enzyme. In untransfected HeLa cells and in the absence of apyrase, cell-to-cell propagating [Ca(2+)](i) changes were characterized by initiating Ca(2+) puffs associated with the perinuclear ER. By contrast, in Cx-GFP-transfected cells and in the presence of apyrase, [Ca(2+)](i) changes were propagated without initiating perinuclear Ca(2+) puffs and were communicated between cells at the sites of the Cx-GFP gap junctions. The efficiency of Cx expression determined the extent of Ca(2+) wave propagation. These results demonstrate that intercellular Ca(2+) waves may be propagated simultaneously via an extracellular pathway and an intracellular pathway through gap junctions and that one form of communication may mask the other.


Asunto(s)
Señalización del Calcio , Conexina 43/metabolismo , Conexinas/metabolismo , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Calcio/metabolismo , Conexina 26 , Conexina 43/genética , Conexinas/genética , Retículo Endoplásmico/metabolismo , Matriz Extracelular/metabolismo , Uniones Comunicantes/metabolismo , Proteínas Fluorescentes Verdes , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Proteína beta1 de Unión Comunicante
10.
Biophys J ; 78(5): 2298-306, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10777728

RESUMEN

InsP(3)-evoked elementary Ca(2+) release events have been postulated to play a role in providing the building blocks of larger Ca(2+) signals. In pancreatic acinar cells, low concentrations of acetylcholine or the injection of low concentrations of InsP(3) elicit a train of spatially localized Ca(2+) spikes. In this study we have quantified these responses and compared the Ca(2+) signals to the elementary events shown in Xenopus oocytes. The results demonstrate, at the same concentrations of InsP(3), Ca(2+) signals consisting of one population of small transient Ca(2+) release events and a second distinct population of larger Ca(2+) spikes. The signal mass amplitudes of both types of events are within the range of amplitudes for the elementary events in Xenopus oocytes. However, the bimodal Ca(2+) distribution of Ca(2+) responses we observe is not consistent with the continuum of event sizes seen in Xenopus. We conclude that the two types of InsP(3)-dependent events in acinar cells are both elementary Ca(2+) signals, which are independent of one another. Our data indicate a complexity to the organization of the Ca(2+) release apparatus in acinar cells, which might result from the presence of multiple InsP(3) receptor isoforms, and is likely to be important in the physiology of these cells.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Inositol 1,4,5-Trifosfato/farmacología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Animales , Fenómenos Biofísicos , Biofisica , Canales de Calcio/metabolismo , Femenino , Técnicas In Vitro , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato , Masculino , Ratones , Microscopía Fluorescente , Oocitos/metabolismo , Páncreas/citología , Técnicas de Placa-Clamp , Isoformas de Proteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Xenopus
11.
J Physiol ; 524 Pt 1: 3-17, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10747180

RESUMEN

1. A digital imaging microscope with fura-2 as the Ca2+ indicator was used to determine the sources for the rise in intracellular calcium concentration ([Ca2+]i) that occurs when the membrane in a cell-attached patch is stretched. Unitary ionic currents from stretch-activated channels and [Ca2+]i images were recorded simultaneously. 2. When suction was applied to the patch pipette to stretch a patch of membrane, Ca2+-permeable cation channels (stretch-activated channels) opened and a global increase in [Ca2+]i occurred, as well as a greater focal increase in the vicinity of the patch pipette. The global changes in [Ca2+]i occurred only when stretch-activated currents were sufficient to cause membrane depolarization, as indicated by the reduction in amplitude of the unitary currents. 3. When Ca2+ was present only in the pipette solution, just the focal change in [Ca2+]i was obtained. This focal change was not seen when the contribution from Ca2+ stores was eliminated using caffeine and ryanodine. 4. These results suggest that the opening of stretch-activated channels allows ions, including Ca2+, to enter the cell. The entry of positive charge triggers the influx of Ca2+ into the cell by causing membrane depolarization, which presumably activates voltage-gated Ca2+ channels. The entry of Ca2+ through stretch-activated channels is also amplified by Ca2+ release from internal stores. This amplification appears to be greater than that obtained by activation of whole-cell Ca2+ currents. These multiple pathways whereby membrane stretch causes a rise in [Ca2+]i may play a role in stretch-induced contraction, which is a characteristic of many smooth muscle tissues.


Asunto(s)
Canales de Calcio/fisiología , Calcio/metabolismo , Contracción Muscular/fisiología , Músculo Liso/fisiología , Estómago/fisiología , Animales , Bufo marinus , Técnicas In Vitro , Potenciales de la Membrana , Técnicas de Placa-Clamp
12.
J Physiol ; 522 Pt 3: 375-90, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10713963

RESUMEN

1. The Ca(2+)-sensitive fluorescent indicator rhod-2 was used to monitor mitochondrial Ca2+ concentration ([Ca2+]m) in gastric smooth muscle cells from Bufo marinus. In some studies, fura-2 was used in combination with rhod-2, allowing simultaneous measurement of cytoplasmic Ca2+ concentration ([Ca2+]i) and [Ca2+]m, respectively. 2. During a short train of depolarizations, which causes Ca2+ influx from the extracellular medium, there was an increase in both [Ca2+]i and [Ca2+]m. The half-time (t1/2) to peak for the increase in [Ca2+]m was considerably longer than the t1/2 to peak for the increase in [Ca2+]i. [Ca2+]m remained elevated for tens of seconds after [Ca2+]i had returned to its resting value. 3. Stimulation with caffeine, which causes release of Ca2+ from the sarcoplasmic reticulum (SR), also produced increases in both [Ca2+]i and [Ca2+]m. The values of t1/2 to peak for the increase in [Ca2+] in both cytoplasm and mitochondria were similar; however, [Ca2+]i returned to baseline values much faster than [Ca2+]m. 4. Using a wide-field digital imaging microscope, changes in [Ca2+]m were monitored within individual mitochondria in situ, during stimulation of Ca2+ influx or Ca2+ release from the SR. 5. Mitochondrial Ca2+ uptake during depolarizing stimulation caused depolarization of the mitochondrial membrane potential. The mitochondrial membrane potential recovered considerably faster than the recovery of [Ca2+]m. 6. This study shows that Ca2+ influx from the extracellular medium and Ca2+ release from the SR are capable of increasing [Ca2+]m in smooth muscle cells. The efflux of Ca2+ from the mitochondria is a slow process and appears to be dependent upon the amount of Ca2+ in the SR.


Asunto(s)
Calcio/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Músculo Liso/metabolismo , Animales , Bufo marinus , Cafeína/farmacología , Células Cultivadas , Citoplasma/metabolismo , Estimulación Eléctrica , Colorantes Fluorescentes/farmacocinética , Compuestos Heterocíclicos con 3 Anillos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Microscopía por Video , Músculo Liso/citología , Técnicas de Placa-Clamp , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/metabolismo , Estómago
13.
J Physiol ; 520 Pt 1: 187-201, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10517811

RESUMEN

1. Cytosolic Ca2+ has been proposed to act as both a positive and a negative feedback signal on the inositol trisphosphate (InsP3) receptor. However, it is unclear how this might affect the Ca2+ response in vivo. 2. Mouse pancreatic acinar cells were whole-cell patch clamped to record the Ca2+-dependent chloride (Cl(Ca)) current spikes and imaged to record the cytosolic Ca2+ spikes elicited by the injection of Ins(2,4,5)P3. Increasing concentrations of Ca2+ buffer (up to 200 microM EGTA or BAPTA) were associated with the appearance of steps in the current activation phase and a prevalence of smaller-amplitude Cl(Ca) spikes. Imaging experiments showed that with increased buffer the secretory pole cytosolic Ca2+ signal became fragmented and spatially discrete Ca2+ release events were observed. 3. At higher buffer concentrations (200-500 microM), increasing concentrations of EGTA increased spike frequency and reduced spike amplitude. In contrast, BAPTA decreased spike frequency and maintained large spike amplitudes. 4. We conclude that, during InsP3-evoked spiking, long-range Ca2+ feedback ( approximately 2-4 microm) shapes the rising phase of the Ca2+ signal by acting to co-ordinate discrete Ca2+ release events and short-range ( approximately 40 nm) Ca2+ feedback acts to inhibit further Ca2+ release.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/fisiología , Inositol 1,4,5-Trifosfato/fisiología , Páncreas/fisiología , Acetatos/farmacología , Animales , Tampones (Química) , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Estimulación Eléctrica , Electrofisiología , Etilenodiaminas/farmacología , Retroalimentación/fisiología , Colorantes Fluorescentes , Masculino , Potenciales de la Membrana/fisiología , Ratones , Páncreas/citología , Técnicas de Placa-Clamp , Fotólisis
14.
J Gen Physiol ; 114(4): 575-88, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10498675

RESUMEN

Discrete localized fluorescence transients due to openings of a single plasma membrane Ca(2+) permeable cation channel were recorded using wide-field digital imaging microscopy with fluo-3 as the Ca(2+) indicator. These transients were obtained while simultaneously recording the unitary channel currents using the whole-cell current-recording configuration of the patch-clamp technique. This cation channel in smooth muscle cells is opened by caffeine (Guerrero, A., F.S. Fay, and J.J. Singer. 1994. J. Gen. Physiol. 104:375-394). The localized fluorescence transients appeared to occur at random locations on the cell membrane, with the duration of the rising phase matching the duration of the channel opening. Moreover, these transients were only observed in the presence of sufficient extracellular Ca(2+), suggesting that they are due to Ca(2+) influx from the bathing solution. The fluorescence transient is characterized by an initial fast rising phase when the channel opens, followed by a slower rising phase during prolonged openings. When the channel closes there is an immediate fast falling phase followed by a slower falling phase. Computer simulations of the underlying events were used to interpret the time course of the transients. The rapid phases are mainly due to the establishment or removal of Ca(2+) and Ca(2+)-bound fluo-3 gradients near the channel when the channel opens or closes, while the slow phases are due to the diffusion of Ca(2+) and Ca(2+)-bound fluo-3 into the cytoplasm. Transients due to short channel openings have a "Ca(2+) spark-like" appearance, suggesting that the rising and early falling components of sparks (due to openings of ryanodine receptors) reflect the fast phases of the fluorescence change. The results presented here suggest methods to determine the relationship between the fluorescence transient and the underlying Ca(2+) current, to study intracellular localized Ca(2+) handling as might occur from single Ca(2+) channel openings, and to localize Ca(2+) permeable ion channels on the plasma membrane.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Citoplasma/metabolismo , Animales , Bufo marinus , Cafeína/farmacología , Agonistas de los Canales de Calcio/farmacología , Canales de Calcio/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Simulación por Computador , Citoplasma/efectos de los fármacos , Colorantes Fluorescentes , Procesamiento de Imagen Asistido por Computador , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo
16.
Curr Biol ; 9(6): 285-91, 1999 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-10209094

RESUMEN

BACKGROUND: Messenger RNA (mRNA) is transcribed and processed in the nucleus of eucaryotic cells and then exported to the cytoplasm through nuclear pores. It is not known whether the movement of mRNA from its site of synthesis to the nuclear pore is directed or random. Directed movement would suggest that there is an energy-requiring step in addition to the step required for active transport through the pore, whereas random movement would indicate that mRNAs can make their way to the nuclear envelope by diffusion. RESULTS: We devised a method to visualize movement of endogenous polymerase II transcripts in the nuclei of living cells. Oligo(dT) labeled with chemically masked (caged) fluorescein was allowed to penetrate cells and hybridize to nuclear poly(A) RNA. Laser spot photolysis then uncaged the oligo(dT) at a given intranuclear site and the resultant fluorescent, hybridized oligo(dT) was tracked using high-speed imaging microscopy. Poly(A) RNA moved away from the uncaging spot in all directions with a mean square displacement that varied linearly with time, and the same apparent diffusion coefficient was measured for the movement at both 37 degrees C and 23 degrees C. These properties are characteristic of a random diffusive process. High resolution three-dimensional imaging of live cells containing both Hoechst-labeled chromosomes and uncaged oligo(dT) showed that, excluding nucleoli, the poly(A) RNA could access most, if not all, of the non-chromosomal space in the nucleus. CONCLUSIONS: Poly(A) RNA can move freely throughout the interchromatin space of the nucleus with properties characteristic of diffusion.


Asunto(s)
Núcleo Celular/metabolismo , Líquido Intracelular/metabolismo , ARN Mensajero/metabolismo , Animales , Bencimidazoles , Transporte Biológico , Células Cultivadas , Difusión , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Membrana Nuclear/metabolismo , Poli A/metabolismo , Poli T/metabolismo , Ratas , Temperatura
17.
J Physiol ; 516 ( Pt 1): 139-47, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10066929

RESUMEN

1. The Ca2+-sensitive fluorescent indicator rhod-2 was used to measure mitochondrial [Ca2+] ([Ca2+]m) in single smooth muscle cells from the rat pulmonary artery, while simultaneously monitoring cytosolic [Ca2+] ([Ca2+]i) with fura-2. 2. Application of caffeine produced an increase in [Ca2+]i and also increased [Ca2+]m. The increase in [Ca2+]m occurred after the increase in [Ca2+]i, and remained elevated for a considerable time after [Ca2+]i had returned to resting values. 3. The protonophore carbonyl cyanide p-(trifluoromethoxy)phenylhydrazone (FCCP), which causes the mitochondrial membrane potential to collapse, markedly attenuated the increase in [Ca2+]m following caffeine application and also increased the half-time for recovery of [Ca2+]i to resting values. 4. Activation of purinoceptors with ATP also produced increases in both [Ca2+]i and [Ca2+]m in these smooth muscle cells. In some cells, oscillations in [Ca2+]i were observed during ATP application, which produced corresponding oscillations in [Ca2+]m and membrane currents. 5. This study provides direct evidence that Ca2+ release from the sarcoplasmic reticulum, either through ryanodine or inositol 1,4, 5-trisphosphate (InsP3) receptors, increases both cytosolic and mitochondrial [Ca2+] in smooth muscle cells. These results have potential implications both for the role of mitochondria in Ca2+ regulation in smooth muscle, and for understanding how cellular metabolism is regulated.


Asunto(s)
Calcio/metabolismo , Mitocondrias Musculares/metabolismo , Arteria Pulmonar/metabolismo , Retículo Sarcoplasmático/metabolismo , Adenosina Trifosfato/farmacología , Animales , Cafeína/farmacología , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Citosol/efectos de los fármacos , Citosol/metabolismo , Colorantes Fluorescentes , Técnicas In Vitro , Ionóforos/farmacología , Masculino , Potenciales de la Membrana/fisiología , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/ultraestructura , Técnicas de Placa-Clamp , Inhibidores de Fosfodiesterasa/farmacología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/ultraestructura , Agonistas Purinérgicos , Ratas , Ratas Sprague-Dawley , Rianodina/farmacología , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/ultraestructura
18.
J Gen Physiol ; 113(2): 215-28, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9925820

RESUMEN

Localized, transient elevations in cytosolic Ca2+, known as Ca2+ sparks, caused by Ca2+ release from sarcoplasmic reticulum, are thought to trigger the opening of large conductance Ca2+-activated potassium channels in the plasma membrane resulting in spontaneous transient outward currents (STOCs) in smooth muscle cells. But the precise relationships between Ca2+ concentration within the sarcoplasmic reticulum and a Ca2+ spark and that between a Ca2+ spark and a STOC are not well defined or fully understood. To address these problems, we have employed two approaches using single patch-clamped smooth muscle cells freshly dissociated from toad stomach: a high speed, wide-field imaging system to simultaneously record Ca2+ sparks and STOCs, and a method to simultaneously measure free global Ca2+ concentration in the sarcoplasmic reticulum ([Ca2+]SR) and in the cytosol ([Ca2+]CYTO) along with STOCs. At a holding potential of 0 mV, cells displayed Ca2+ sparks and STOCs. Ca2+ sparks were associated with STOCs; the onset of the sparks coincided with the upstroke of STOCs, and both had approximately the same decay time. The mean increase in [Ca2+]CYTO at the time and location of the spark peak was approximately 100 nM above a resting concentration of approximately 100 nM. The frequency and amplitude of spontaneous Ca2+ sparks recorded at -80 mV were unchanged for a period of 10 min after removal of extracellular Ca2+ (nominally Ca2+-free solution with 50 microM EGTA), indicating that Ca2+ influx is not necessary for Ca2+sparks. A brief pulse of caffeine (20 mM) elicited a rapid decrease in [Ca2+]SR in association with a surge in [Ca2+]CYTO and a fusion of STOCs, followed by a fast restoration of [Ca2+]CYTO and a gradual recovery of [Ca2+]SR and STOCs. The return of global [Ca2+]CYTO to rest was an order of magnitude faster than the refilling of the sarcoplasmic reticulum with Ca2+. After the global [Ca2+]CYTO was fully restored, recovery of STOC frequency and amplitude were correlated with the level of [Ca2+]SR, even though the time for refilling varied greatly. STOC frequency did not recover substantially until the [Ca2+]SR was restored to 60% or more of resting levels. At [Ca2+]SR levels above 80% of rest, there was a steep relationship between [Ca2+]SR and STOC frequency. In contrast, the relationship between [Ca2+]SR and STOC amplitude was linear. The relationship between [Ca2+]SR and the frequency and amplitude was the same for Ca2+ sparks as it was for STOCs. The results of this study suggest that the regulation of [Ca2+]SR might provide one mechanism whereby agents could govern Ca2+ sparks and STOCs. The relationship between Ca2+ sparks and STOCs also implies a close association between a sarcoplasmic reticulum Ca2+ release site and the Ca2+-activated potassium channels responsible for a STOC.


Asunto(s)
Canales de Calcio/fisiología , Señalización del Calcio/fisiología , Músculo Liso/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Bufo marinus , Citosol/metabolismo , Estimulación Eléctrica , Electrofisiología , Espacio Extracelular/metabolismo , Fura-2 , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Indicadores y Reactivos , Potenciales de la Membrana/fisiología , Microscopía Confocal , Músculo Liso/citología , Músculo Liso/ultraestructura , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Retículo Sarcoplasmático/ultraestructura
20.
J Physiol ; 513 ( Pt 3): 711-8, 1998 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9824712

RESUMEN

1. Local changes in cytosolic [Ca2+] were imaged with a wide-field, high-speed, digital imaging system while membrane currents were simultaneously recorded using whole-cell, perforated patch recording in freshly dissociated guinea-pig tracheal myocytes. 2. Depending on membrane potential, Ca2+ sparks triggered 'spontaneous' transient inward currents (STICs), 'spontaneous' transient outward currents (STOCs) and biphasic currents in which the outward phase always preceded the inward (STOICs). The outward currents resulted from the opening of large-conductance Ca2+-activated K+ (BK) channels and the inward currents from Ca2+-activated Cl- (ClCa) channels. 3. A single Ca2+ spark elicited both phases of a STOIC, and sparks originating from the same site triggered STOCs, STICs and STOICs, depending on membrane potential. 4. STOCs had a shorter time to peak (TTP) than Ca2+ sparks and a much shorter half-time of decay. In contrast, STICs had a somewhat longer TTP than sparks but the same half-time of decay. Thus, the STIC, not the STOC, more closely reflected the time course of cytosolic Ca2+ elevation during a Ca2+ spark. 5. These findings suggest that ClCa channels and BK channels may be organized spatially in quite different ways in relation to points of Ca2+ release from intracellular Ca2+ stores. The results also suggest that Ca2+ sparks may have functions in smooth muscle not previously suggested, such as a stabilizing effect on membrane potential and hence on the contractile state of the cell, or as activators of voltage-gated Ca2+ channels due to depolarization mediated by STICs.


Asunto(s)
Señalización del Calcio/fisiología , Canales de Cloruro/metabolismo , Canales de Potasio/metabolismo , Tráquea/metabolismo , Algoritmos , Animales , Bradiquinina/metabolismo , Estimulación Eléctrica , Electrofisiología , Cobayas , Procesamiento de Imagen Asistido por Computador , Técnicas In Vitro , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Factores de Tiempo , Tráquea/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA