Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(28): 16626-16637, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32601236

RESUMEN

Neuronal activity leads to an increase in local cerebral blood flow (CBF) to allow adequate supply of oxygen and nutrients to active neurons, a process termed neurovascular coupling (NVC). We have previously shown that capillary endothelial cell (cEC) inwardly rectifying K+ (Kir) channels can sense neuronally evoked increases in interstitial K+ and induce rapid and robust dilations of upstream parenchymal arterioles, suggesting a key role of cECs in NVC. The requirements of this signal conduction remain elusive. Here, we utilize mathematical modeling to investigate how small outward currents in stimulated cECs can elicit physiologically relevant spread of vasodilatory signals within the highly interconnected brain microvascular network to increase local CBF. Our model shows that the Kir channel can act as an "on-off" switch in cECs to hyperpolarize the cell membrane as extracellular K+ increases. A local hyperpolarization can be amplified by the voltage-dependent activation of Kir in neighboring cECs. Sufficient Kir density enables robust amplification of the hyperpolarizing stimulus and produces responses that resemble action potentials in excitable cells. This Kir-mediated excitability can remain localized in the stimulated region or regeneratively propagate over significant distances in the microvascular network, thus dramatically increasing the efficacy of K+ for eliciting local hyperemia. Modeling results show how changes in cEC transmembrane current densities and gap junctional resistances can affect K+-mediated NVC and suggest a key role for Kir as a sensor of neuronal activity and an amplifier of retrograde electrical signaling in the cerebral vasculature.


Asunto(s)
Neuronas/metabolismo , Acoplamiento Neurovascular , Canales de Potasio de Rectificación Interna/metabolismo , Potasio/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Circulación Cerebrovascular , Células Endoteliales/química , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Neuronas/química , Potasio/química , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Transducción de Señal , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(43): 27022-27033, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33051294

RESUMEN

The essential function of the circulatory system is to continuously and efficiently supply the O2 and nutrients necessary to meet the metabolic demands of every cell in the body, a function in which vast capillary networks play a key role. Capillary networks serve an additional important function in the central nervous system: acting as a sensory network, they detect neuronal activity in the form of elevated extracellular K+ and initiate a retrograde, propagating, hyperpolarizing signal that dilates upstream arterioles to rapidly increase local blood flow. Yet, little is known about how blood entering this network is distributed on a branch-to-branch basis to reach specific neurons in need. Here, we demonstrate that capillary-enwrapping projections of junctional, contractile pericytes within a postarteriole transitional region differentially constrict to structurally and dynamically determine the morphology of capillary junctions and thereby regulate branch-specific blood flow. We further found that these contractile pericytes are capable of receiving propagating K+-induced hyperpolarizing signals propagating through the capillary network and dynamically channeling red blood cells toward the initiating signal. By controlling blood flow at junctions, contractile pericytes within a functionally distinct postarteriole transitional region maintain the efficiency and effectiveness of the capillary network, enabling optimal perfusion of the brain.


Asunto(s)
Capilares/fisiología , Circulación Cerebrovascular , Microcirculación , Pericitos/fisiología , Animales , Arteriolas/fisiología , Canales de Calcio/metabolismo , Venas Cerebrales/fisiología , Ratones
3.
J Theor Biol ; 529: 110856, 2021 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-34363836

RESUMEN

Blood Oxygen Level Dependent (BOLD) signal indirectly characterizes neuronal activity by measuring hemodynamic and metabolic changes in the nearby microvasculature. A deeper understanding of how localized changes in electrical, metabolic and hemodynamic factors translate into a BOLD signal is crucial for the interpretation of functional brain imaging techniques. While positive BOLD responses (PBR) are widely considered to be linked with neuronal activation, the origins of negative BOLD responses (NBR) have remained largely unknown. As NBRs are sometimes observed in close proximity of regions with PBR, a blood "stealing" effect, i.e., redirection of blood from a passive periphery to the area with high neuronal activity, has been postulated. In this study, we used the Hagen-Poiseuille equation to model hemodynamics in an idealized microvascular network that account for the particulate nature of blood and nonlinearities arising from the red blood cell (RBC) distribution (i.e., the Fåhraeus, Fåhraeus-Lindqvist and the phase separation effects). Using this detailed model, we evaluate determinants driving this "stealing" effect in a microvascular network with geometric parameters within physiological ranges. Model simulations predict that during localized cerebral blood flow (CBF) increases due to neuronal activation-hyperemic response, blood from surrounding vessels is reallocated towards the activated region. This stealing effect depended on the resistance of the microvasculature and the uneven distribution of RBCs at vessel bifurcations. A parsimonious model consisting of two-connected windkessel regions sharing a supplying artery was proposed to simulate the stealing effect with a minimum number of parameters. Comparison with the detailed model showed that the parsimonious model can reproduce the observed response for hematocrit values within the physiological range for different species. Our novel parsimonious model promise to be of use for statistical inference (top-down analysis) from direct blood flow measurements (e.g., arterial spin labeling and laser Doppler/Speckle flowmetry), and when combined with theoretical models for oxygen extraction/diffusion will help account for some types of NBRs.


Asunto(s)
Imagen por Resonancia Magnética , Robo , Encéfalo , Circulación Cerebrovascular , Hematócrito , Hemodinámica , Oxígeno
4.
Microcirculation ; 25(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29117630

RESUMEN

OBJECTIVE: Electrical signaling along the endothelium underlies spreading vasodilation and blood flow control. We use mathematical modeling to determine the electrical properties of the endothelium and gain insight into the biophysical determinants of electrical conduction. METHODS: Electrical conduction data along endothelial tubes (40 µm wide, 2.5 mm long) isolated from mouse skeletal muscle resistance arteries were analyzed using cable equations and a multicellular computational model. RESULTS: Responses to intracellular current injection attenuate with an axial length constant (λ) of 1.2-1.4 mm. Data were fitted to estimate the axial (ra ; 10.7 MΩ/mm) and membrane (rm ; 14.5 MΩ∙mm) resistivities, EC membrane resistance (Rm ; 12 GΩ), and EC-EC coupling resistance (Rgj ; 4.5 MΩ) and predict that stimulation of ≥30 neighboring ECs is required to elicit 1 mV of hyperpolarization at distance = 2.5 mm. Opening Ca2+ -activated K+ channels (KCa ) along the endothelium reduced λ by up to 55%. CONCLUSIONS: High Rm makes the endothelium sensitive to electrical stimuli and able to conduct these signals effectively. Whereas the activation of a group of ECs is required to initiate physiologically relevant hyperpolarization, this requirement is increased by myoendothelial coupling and KCa activation along the endothelium inhibits conduction by dissipating electrical signals.


Asunto(s)
Endotelio Vascular/fisiología , Microvasos/fisiología , Modelos Biológicos , Transducción de Señal/fisiología , Animales , Potenciales de la Membrana/fisiología , Ratones , Músculo Esquelético/irrigación sanguínea , Canales de Potasio Calcio-Activados/fisiología , Flujo Sanguíneo Regional , Transmisión Sináptica , Vasodilatación
5.
Microcirculation ; 25(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29247493

RESUMEN

Cerebral SVDs encompass a group of genetic and sporadic pathological processes leading to brain lesions, cognitive decline, and stroke. There is no specific treatment for SVDs, which progress silently for years before becoming clinically symptomatic. Here, we examine parallels in the functional defects of PAs in CADASIL, a monogenic form of SVD, and in response to SAH, a common type of hemorrhagic stroke that also targets the brain microvasculature. Both animal models exhibit dysregulation of the voltage-gated potassium channel, KV 1, in arteriolar myocytes, an impairment that compromises responses to vasoactive stimuli and impacts CBF autoregulation and local dilatory responses to neuronal activity (NVC). However, the extent to which this channelopathy-like defect ultimately contributes to these pathologies is unknown. Combining experimental data with computational modeling, we describe the role of KV 1 channels in the regulation of myocyte membrane potential at rest and during the modest increase in extracellular potassium associated with NVC. We conclude that PA resting membrane potential and myogenic tone depend strongly on KV 1.2/1.5 channel density, and that reciprocal changes in KV channel density in CADASIL and SAH produce opposite effects on extracellular potassium-mediated dilation during NVC.


Asunto(s)
Microvasos/patología , Canales de Potasio con Entrada de Voltaje/análisis , Animales , CADASIL/fisiopatología , Dilatación , Humanos , Canales de Potasio con Entrada de Voltaje/fisiología , Hemorragia Subaracnoidea/fisiopatología
6.
Am J Physiol Heart Circ Physiol ; 312(4): H854-H866, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130333

RESUMEN

We used mathematical modeling to investigate nitric oxide (NO)-dependent vasodilatory signaling in the arteriolar wall. Detailed continuum cellular models of calcium (Ca2+) dynamics and membrane electrophysiology in smooth muscle and endothelial cells (EC) were coupled with models of NO signaling and biotransport in an arteriole. We used this theoretical approach to examine the role of endothelial hemoglobin-α (Hbα) as a modulator of NO-mediated myoendothelial feedback, as previously suggested in Straub et al. (Nature 491: 473-477, 2012). The model considers enriched expression of inositol 1,4,5-triphosphate receptors (IP3Rs), endothelial nitric oxide synthase (eNOS) enzyme, Ca2+-activated potassium (KCa) channels and Hbα in myoendothelial projections (MPs) between the two cell layers. The model suggests that NO-mediated myoendothelial feedback is plausible if a significant percentage of eNOS is localized within or near the myoendothelial projection. Model results show that the ability of Hbα to regulate the myoendothelial feedback is conditional to its colocalization with eNOS near MPs at concentrations in the high nanomolar range (>0.2 µM or 24,000 molecules). Simulations also show that the effect of Hbα observed in in vitro experimental studies may overestimate its contribution in vivo, in the presence of blood perfusion. Thus, additional experimentation is required to quantify the presence and spatial distribution of Hbα in the EC, as well as to test that the strong effect of Hbα on NO signaling seen in vitro, translates also into a physiologically relevant response in vivo.NEW & NOTEWORTHY Mathematical modeling shows that although regulation of nitric oxide signaling by hemoglobin-α (Hbα) is plausible, it is conditional to its presence in significant concentrations colocalized with endothelial nitric oxide synthase in myoendothelial projections. Additional experimentation is required to test that the strong effect of Hbα seen in vitro translates into a physiologically relevant response in vivo.


Asunto(s)
Endotelio Vascular/fisiología , Hemoglobina A/fisiología , Óxido Nítrico/fisiología , Transducción de Señal/fisiología , Vasodilatación/fisiología , Algoritmos , Simulación por Computador , Eritrocitos/efectos de los fármacos , Retroalimentación Fisiológica , Humanos , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Modelos Teóricos , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Canales de Potasio Calcio-Activados/biosíntesis
7.
Am J Physiol Cell Physiol ; 310(6): C479-90, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26739489

RESUMEN

Shear stress is known to stimulate an intracellular free calcium concentration ([Ca(2+)]i) response in vascular endothelial cells (ECs). [Ca(2+)]i is a key second messenger for signaling that leads to vasodilation and EC survival. Although it is accepted that the shear-induced [Ca(2+)]i response is, in part, due to Ca(2+) release from the endoplasmic reticulum (ER), the role of mitochondria (second largest Ca(2+) store) is unknown. We hypothesized that the mitochondria play a role in regulating [Ca(2+)]i in sheared ECs. Cultured ECs, loaded with a Ca(2+)-sensitive fluorophore, were exposed to physiological levels of shear stress. Shear stress elicited [Ca(2+)]i transients in a percentage of cells with a fraction of them displaying oscillations. Peak magnitudes, percentage of oscillating ECs, and oscillation frequencies depended on the shear level. [Ca(2+)]i transients/oscillations were present when experiments were conducted in Ca(2+)-free solution (plus lanthanum) but absent when ECs were treated with a phospholipase C inhibitor, suggesting that the ER inositol 1,4,5-trisphosphate receptor is responsible for the [Ca(2+)]i response. Either a mitochondrial uncoupler or an electron transport chain inhibitor, but not a mitochondrial ATP synthase inhibitor, prevented the occurrence of transients and especially inhibited the oscillations. Knockdown of the mitochondrial Ca(2+) uniporter also inhibited the shear-induced [Ca(2+)]i transients/oscillations compared with controls. Hence, EC mitochondria, through Ca(2+) uptake/release, regulate the temporal profile of shear-induced ER Ca(2+) release. [Ca(2+)]i oscillation frequencies detected were within the range for activation of mechanoresponsive kinases and transcription factors, suggesting that dysfunctional EC mitochondria may contribute to cardiovascular disease by deregulating the shear-induced [Ca(2+)]i response.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Mitocondrias/metabolismo , Células Cultivadas , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Sistemas de Mensajero Secundario/fisiología , Estrés Mecánico
8.
Biophys J ; 108(6): 1566-1576, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25809269

RESUMEN

We examined the endothelial transient receptor vanilloid 4 (TRPV4) channel's vasodilatory signaling using mathematical modeling. The model analyzes experimental data by Sonkusare and coworkers on TRPV4-induced endothelial Ca(2+) events (sparklets). A previously developed continuum model of an endothelial and a smooth muscle cell coupled through microprojections was extended to account for the activity of a TRPV4 channel cluster. Different stochastic descriptions for the TRPV4 channel flux were examined using finite-state Markov chains. The model also took into consideration recent evidence for the colocalization of intermediate-conductance calcium-activated potassium channels (IKCa) and TRPV4 channels near the microprojections. A single TRPV4 channel opening resulted in a stochastic localized Ca(2+) increase in a small region (i.e., few µm(2) area) close to the channel. We predict micromolar Ca(2+) increases lasting for the open duration of the channel sufficient for the activation of low-affinity endothelial KCa channels. Simulations of a cluster of four TRPV4 channels incorporating burst and cooperative gating kinetics provided quantal Ca(2+) increases (i.e., steps of fixed amplitude), similar to the experimentally observed Ca(2+) sparklets. These localized Ca(2+) events result in endothelium-derived hyperpolarization (and SMC relaxation), with magnitude that depends on event frequency. The gating characteristics (bursting, cooperativity) of the TRPV4 cluster enhance Ca(2+) spread and the distance of KCa channel activation. This may amplify the EDH response by the additional recruitment of distant KCa channels.


Asunto(s)
Modelos Moleculares , Canales Catiónicos TRPV/metabolismo , Calcio/metabolismo , Simulación por Computador , Células Endoteliales/metabolismo , Cinética , Cadenas de Markov , Canales de Potasio Calcio-Activados/metabolismo , Procesos Estocásticos
9.
J Physiol ; 591(11): 2795-812, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23529128

RESUMEN

We investigated the role of myoendothelial projections (MPs) in endothelial cell (EC) feedback response to smooth muscle cell (SMC) stimulation using mathematical modelling. A previously developed compartmental EC-SMC model is modified to include MPs as subcellular compartments in the EC. The model is further extended into a 2D continuum model using a finite element method (FEM) approach and electron microscopy images to account for MP geometry. The EC and SMC are coupled via non-selective myoendothelial gap junctions (MEGJs) which are located on MPs and allow exchange of Ca(2+), K(+), Na(+) and Cl(-) ions and inositol 1,4,5-triphosphate (IP3). Models take into consideration recent evidence for co-localization of intermediate-conductance calcium-activated potassium channels (IKCa) and IP3 receptors (IP3Rs) in the MPs. SMC stimulation causes an IP3-mediated Ca(2+) transient in the MPs with limited global spread in the bulk EC. A hyperpolarizing feedback generated by the localized IKCa channels is transmitted to the SMC via MEGJs. MEGJ resistance (Rgj) and the density of IKCa and IP3R in the projection influence the extent of EC response to SMC stimulation. The predicted Ca(2+) transients depend also on the volume and geometry of the MP. We conclude that in the myoendothelial feedback response to SMC stimulation, MPs are required to amplify the SMC initiated signal. Simulations suggest that the signal is mediated by IP3 rather than Ca(2+) diffusion and that a localized rather than a global EC Ca(2+) mobilization is more likely following SMC stimulation.


Asunto(s)
Extensiones de la Superficie Celular/fisiología , Células Endoteliales/fisiología , Retroalimentación Fisiológica , Modelos Biológicos , Miocitos del Músculo Liso/fisiología , Potenciales de Acción , Animales , Calcio/metabolismo , Señalización del Calcio , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Cloruros/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Uniones Comunicantes/metabolismo , Uniones Comunicantes/fisiología , Uniones Comunicantes/ultraestructura , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Arterias Mesentéricas/citología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Potasio/metabolismo , Ratas , Sodio/metabolismo
10.
Nitric Oxide ; 28: 39-46, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23063986

RESUMEN

Nitric oxide (NO) research in biomedicine has been hampered by the absence of a method that will allow quantitative measurement of NO in biological tissues with high sensitivity and selectivity, and with adequate spatial and temporal resolution. 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM) is a NO sensitive fluorescence probe that has been used widely for qualitative assessment of cellular NO production. However, calibration of the fluorescent signal and quantification of NO concentration in cells and tissues using fluorescent probes, have provided significant challenge. In this study we utilize a combination of mathematical modeling and experimentation to elucidate the kinetics of NO/DAF-FM reaction in solution. Modeling and experiments suggest that the slope of fluorescent intensity (FI) can be related to NO concentration according to the equation: ddtFI=2αk(1)NO(2)O(2)DAF-FMkNO+DAF-FM where α is a proportionality coefficient that relates FI to unit concentration of activated DAF-FM, k(1) is the NO oxidation rate constant, and k was estimated to be 4.3±0.6. The FI slope exhibits saturation kinetics with DAF-FM concentration. Interestingly, the effective half-maximum constant (EC(50)) increases proportionally to NO concentration. This result is not in agreement with the proposition that N(2)O(3) is the NO oxidation byproduct that activates DAF-FM. Kinetic analysis suggests that the reactive intermediate should exhibit NO-dependent consumption and thus NO(2)() is a more likely candidate. The derived rate law can be used for the calibration of DAF-FM fluorescence and the quantification of NO concentration in biological tissues.


Asunto(s)
Fluoresceínas/química , Colorantes Fluorescentes/química , Óxido Nítrico/análisis , Óxido Nítrico/química , Calibración , Fluoresceínas/análisis , Colorantes Fluorescentes/análisis , Cinética , Oxidación-Reducción , Factores de Tiempo
11.
Nitric Oxide ; 35: 65-71, 2013 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23994446

RESUMEN

l-Arginine and its decarboxylated product, agmatine are important mediators of NO production and vascular relaxation. However, the underlying mechanisms of their action are not understood. We have investigated the role of arginine and agmatine in resistance vessel relaxation of Sprague-Dawley (SD) and Dahl salt-sensitive hypertensive rats. Second or 3rd-order mesenteric arterioles were cannulated in an organ chamber, pressurized and equilibrated before perfusing intraluminally with agonists. The vessel diameters were measured after mounting on the stage of a microscope fitted with a video camera. The gene expression in Dahl rat vessel homogenates was ascertained by real-time PCR. l-Arginine initiated relaxations (EC50, 5.8±0.7mM; n=9) were inhibited by arginine decarboxylase (ADC) inhibitor, difluoromethylarginine (DFMA) (EC50, 18.3±1.3mM; n=5) suggesting that arginine-induced vessel relaxation was mediated by agmatine formation. Agmatine relaxed the SD rat vessels at significantly lower concentrations (EC50, 138.7±12.1µM; n=22), which was compromised by l-NAME (l-N(G)-nitroarginine methyl ester, an eNOS inhibitor), RX821002 (α-2 AR antagonist) and pertussis toxin (G-protein inhibitor). The agmatine-mediated vessel relaxation from high salt Dahl rats was abolished as compared to that from normal salt rats (EC50, 143.9±23.4µM; n=5). The α-2A AR, α-2B AR and eNOS mRNA expression was downregulated in mesenteric arterioles of high-salt treated Dahl hypertensive rats. These findings demonstrate that agmatine facilitated the relaxation via activation of α-2 adrenergic G-protein coupled receptor and NO synthesis, and this pathway is compromised in salt-sensitive hypertension.


Asunto(s)
Agmatina/farmacología , Hipertensión/fisiopatología , Arterias Mesentéricas/efectos de los fármacos , Óxido Nítrico/metabolismo , Vasodilatación/efectos de los fármacos , Animales , Arginina/farmacología , Carboxiliasas/antagonistas & inhibidores , Carboxiliasas/metabolismo , Masculino , Arterias Mesentéricas/fisiología , Ratas , Ratas Endogámicas Dahl , Ratas Sprague-Dawley
12.
Biophys J ; 102(2): 211-20, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22339857

RESUMEN

The intercellular synchronization of spontaneous calcium (Ca(2+)) oscillations in individual smooth muscle cells is a prerequisite for vasomotion. A detailed mathematical model of Ca(2+) dynamics in rat mesenteric arteries shows that a number of synchronizing and desynchronizing pathways may be involved. In particular, Ca(2+)-dependent phospholipase C, the intercellular diffusion of inositol trisphosphate (IP(3), and to a lesser extent Ca(2+)), IP(3) receptors, diacylglycerol-activated nonselective cation channels, and Ca(2+)-activated chloride channels can contribute to synchronization, whereas large-conductance Ca(2+)-activated potassium channels have a desynchronizing effect. Depending on the contractile state and agonist concentrations, different pathways become predominant, and can be revealed by carefully inhibiting the oscillatory component of their total activity. The phase shift between the Ca(2+) and membrane potential oscillations can change, and thus electrical coupling through gap junctions can mediate either synchronization or desynchronization. The effect of the endothelium is highly variable because it can simultaneously enhance the intercellular coupling and affect multiple smooth muscle cell components. Here, we outline a system of increased complexity and propose potential synchronization mechanisms that need to be experimentally tested.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Arterias Mesentéricas/citología , Arterias Mesentéricas/fisiología , Modelos Biológicos , Animales , Fenómenos Electrofisiológicos , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Canales Iónicos/metabolismo , Potenciales de la Membrana , Arterias Mesentéricas/metabolismo , Óxido Nítrico/metabolismo , Ratas , Factores de Tiempo
13.
Am J Physiol Cell Physiol ; 302(8): C1226-42, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22277756

RESUMEN

When arteries constrict to agonists, the endothelium inversely responds, attenuating the initial vasomotor response. The basis of this feedback mechanism remains uncertain, although past studies suggest a key role for myoendothelial communication in the signaling process. The present study examined whether second messenger flux through myoendothelial gap junctions initiates a negative-feedback response in hamster retractor muscle feed arteries. We specifically hypothesized that when agonists elicit depolarization and a rise in second messenger concentration, inositol trisphosphate (IP(3)) flux activates a discrete pool of IP(3) receptors (IP(3)Rs), elicits localized endothelial Ca(2+) transients, and activates downstream effectors to moderate constriction. With use of integrated experimental techniques, this study provided three sets of supporting observations. Beginning at the functional level, we showed that blocking intermediate-conductance Ca(2+)-activated K(+) channels (IK) and Ca(2+) mobilization from the endoplasmic reticulum (ER) enhanced the contractile/electrical responsiveness of feed arteries to phenylephrine. Next, structural analysis confirmed that endothelial projections make contact with the overlying smooth muscle. These projections retained membranous ER networks, and IP(3)Rs and IK channels localized in or near this structure. Finally, Ca(2+) imaging revealed that phenylephrine induced discrete endothelial Ca(2+) events through IP(3)R activation. These events were termed recruitable Ca(2+) wavelets on the basis of their spatiotemporal characteristics. From these findings, we conclude that IP(3) flux across myoendothelial gap junctions is sufficient to induce focal Ca(2+) release from IP(3)Rs and activate a discrete pool of IK channels within or near endothelial projections. The resulting hyperpolarization feeds back on smooth muscle to moderate agonist-induced depolarization and constriction.


Asunto(s)
Calcio/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Cricetinae , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Retroalimentación/efectos de los fármacos , Uniones Comunicantes/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Mesocricetus , Músculo Liso Vascular/efectos de los fármacos , Fenilefrina/farmacología , Canales de Potasio Calcio-Activados/metabolismo , Vasoconstricción/efectos de los fármacos
14.
Microcirculation ; 19(5): 391-402, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22340204

RESUMEN

Movement of ions (Ca(2+) , K(+) , Na(+) , and Cl(-) ) and second messenger molecules like inositol 1, 4, 5-trisphosphate inside and in between different cells is the basis of many signaling mechanisms in the microcirculation. In spite of the vast experimental efforts directed toward evaluation of these fluxes, it has been a challenge to establish their roles in many essential microcirculatory phenomena. Recently, detailed theoretical models of calcium dynamics and plasma membrane electrophysiology have emerged to assist in the quantification of these intra and intercellular fluxes and enhance understanding of their physiological importance. This perspective reviews selected models relevant to estimation of such intra and intercellular ionic and second messenger fluxes and prediction of their relative significance to a variety of vascular phenomena, such as myoendothelial feedback, conducted responses, and vasomotion.


Asunto(s)
Comunicación Celular/fisiología , Endotelio Vascular/fisiología , Hemodinámica/fisiología , Modelos Cardiovasculares , Músculo Liso Vascular/fisiología , Sistemas de Mensajero Secundario/fisiología , Animales , Humanos , Transporte Iónico/fisiología , Iones/metabolismo
15.
Phytother Res ; 26(4): 541-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21928368

RESUMEN

Feverfew is the most commonly used medicinal herb against migraine headache. The antimigraine mechanism of feverfew supercritical extract was investigated in vitro using the mouse macrophage cell line (RAW 264.7). Mouse macrophage cells were treated with lipopolysaccharide in the presence and absence of feverfew extracts. Inhibition of lipopolysaccharide-induced nitric oxide and TNF-α synthesis were quantified by ELISA. The mRNA and protein expression of iNOS and eNOS genes were analysed by RT-PCR and western blot analysis, respectively. The feverfew extract inhibited both nitric oxide (NO) and TNF-α production in a dose-dependent manner with complete inhibition of NO occurring at 5 µg/mL of feverfew extract. Both eNOS and iNOS mRNA levels were unchanged with the feverfew treatment. However, eNOS and iNOS proteins were significantly down-regulated by the feverfew extract. Feverfew inhibition of NO is due to the down-regulation of both eNOS and iNOS enzymes at the translational and/or post-translational level.


Asunto(s)
Macrófagos/efectos de los fármacos , Óxido Nítrico/antagonistas & inhibidores , Extractos Vegetales/farmacología , Tanacetum parthenium/química , Animales , Western Blotting , Dióxido de Carbono/metabolismo , Línea Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Lipopolisacáridos/efectos adversos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
16.
Crit Rev Biomed Eng ; 39(5): 435-60, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22196162

RESUMEN

A network of intracellular signaling pathways and complex intercellular interactions regulate calcium mobilization in vascular cells, arteriolar tone, and blood flow. Different endothelium-derived vasoreactive factors have been identified and the importance of myoendothelial communication in vasoreactivity is now well appreciated. The ability of many vascular networks to conduct signals upstream also is established. This phenomenon is critical for both short-term changes in blood perfusion as well as long-term adaptations of a vascular network. In addition, in a phenomenon termed vasomotion, arterioles often exhibit spontaneous oscillations in diameter. This is thought to improve tissue oxygenation and enhance blood flow. Experimentation has begun to reveal important aspects of the regulatory machinery and the significance of these phenomena for the regulation of local perfusion and oxygenation. Mathematical modeling can assist in elucidating the complex signaling mechanisms that participate in these phenomena. This review highlights some of the important experimental studies and relevant mathematical models that provide the current understanding of these mechanisms in vasoreactivity.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Endotelio Vascular/fisiología , Microcirculación/fisiología , Modelos Cardiovasculares , Sistema Vasomotor/fisiología , Adaptación Fisiológica , Animales , Arteriolas/metabolismo , Fenómenos Fisiológicos Cardiovasculares , Comunicación Celular/fisiología , Hemodinámica , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología
17.
Am J Physiol Heart Circ Physiol ; 298(1): H52-65, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19855062

RESUMEN

This study presents a multicellular computational model of a rat mesenteric arteriole to investigate the signal transduction mechanisms involved in the generation of conducted vasoreactivity. The model comprises detailed descriptions of endothelial (ECs) and smooth muscle (SM) cells (SMCs), coupled by nonselective gap junctions. With strong myoendothelial coupling, local agonist stimulation of the EC or SM layer causes local changes in membrane potential (V(m)) that are conducted electrotonically, primarily through the endothelium. When myoendothelial coupling is weak, signals initiated in the SM conduct poorly, but the sensitivity of the SMCs to current injection and agonist stimulation increases. Thus physiological transmembrane currents can induce different levels of local V(m) change, depending on cell's gap junction connectivity. The physiological relevance of current and voltage clamp stimulations in intact vessels is discussed. Focal agonist stimulation of the endothelium reduces cytosolic calcium (intracellular Ca(2+) concentration) in the prestimulated SM layer. This SMC Ca(2+) reduction is attributed to a spread of EC hyperpolarization via gap junctions. Inositol (1,4,5)-trisphosphate, but not Ca(2+), diffusion through homocellular gap junctions can increase intracellular Ca(2+) concentration in neighboring ECs. The small endothelial Ca(2+) spread can amplify the total current generated at the local site by the ECs and through the nitric oxide pathway, by the SMCs, and thus reduces the number of stimulated cells required to induce distant responses. The distance of the electrotonic and Ca(2+) spread depends on the magnitude of SM prestimulation and the number of SM layers. Model results are consistent with experimental data for vasoreactivity in rat mesenteric resistance arteries.


Asunto(s)
Arterias Mesentéricas/fisiología , Acetilcolina/farmacología , Algoritmos , Animales , Arteriolas/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Simulación por Computador , Estimulación Eléctrica , Uniones Comunicantes/efectos de los fármacos , Cinética , Arterias Mesentéricas/efectos de los fármacos , Modelos Estadísticos , Contracción Muscular/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
18.
Microcirculation ; 16(8): 694-713, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19905969

RESUMEN

To study the effect of myoendothelial communication on vascular reactivity, we integrated detailed mathematical models of Ca(2+) dynamics and membrane electrophysiology in arteriolar smooth muscle (SMC) and endothelial (EC) cells. Cells are coupled through the exchange of Ca(2+), Cl(-), K(+), and Na(+) ions, inositol 1,4,5-triphosphate (IP(3)), and the paracrine diffusion of nitric oxide (NO). EC stimulation reduces intracellular Ca(2+) ([Ca(2+)](i)) in the SMC by transmitting a hyperpolarizing current carried primarily by K(+). The NO-independent endothelium-derived hyperpolarization was abolished in a synergistic-like manner by inhibition of EC SK(Ca) and IK(Ca) channels. During NE stimulation, IP(3) diffusing from the SMC induces EC Ca(2+) release, which, in turn, moderates SMC depolarization and [Ca(2+)](i) elevation. On the contrary, SMC [Ca(2+)](i) was not affected by EC-derived IP(3). Myoendothelial Ca(2+) fluxes had no effect in either cell. The EC exerts a stabilizing effect on calcium-induced calcium release-dependent SMC Ca(2+) oscillations by increasing the norepinephrine concentration window for oscillations. We conclude that a model based on independent data for subcellular components can capture major features of the integrated vessel behavior. This study provides a tissue-specific approach for analyzing complex signaling mechanisms in the vasculature.


Asunto(s)
Arteriolas/fisiología , Potenciales de la Membrana , Modelos Teóricos , Miocitos del Músculo Liso/metabolismo , Comunicación Paracrina/fisiología , Circulación Esplácnica/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio , Electrofisiología , Células Endoteliales , Inositol 1,4,5-Trifosfato/metabolismo , Músculo Liso Vascular , Miocitos del Músculo Liso/fisiología , Óxido Nítrico/metabolismo , Norepinefrina , Ratas
19.
Sci Rep ; 9(1): 17072, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31745211

RESUMEN

Upon inositol trisphosphate (IP3) stimulation of non-excitable cells, including vascular endothelial cells, calcium (Ca2+) shuttling between the endoplasmic reticulum (ER) and mitochondria, facilitated by complexes called Mitochondria-Associated ER Membranes (MAMs), is known to play an important role in the occurrence of cytosolic Ca2+ concentration ([Ca2+]Cyt) oscillations. A mathematical compartmental closed-cell model of Ca2+ dynamics was developed that accounts for ER-mitochondria Ca2+ microdomains as the µd compartment (besides the cytosol, ER and mitochondria), Ca2+ influx to/efflux from each compartment and Ca2+ buffering. Varying the distribution of functional receptors in MAMs vs. the rest of ER/mitochondrial membranes, a parameter called the channel connectivity coefficient (to the µd), allowed for generation of [Ca2+]Cytoscillations driven by distinct mechanisms at various levels of IP3 stimulation. Oscillations could be initiated by the transient opening of IP3 receptors facing either the cytosol or the µd, and subsequent refilling of the respective compartment by Ca2+ efflux from the ER and/or the mitochondria. Only under conditions where the µd became the oscillation-driving compartment, silencing the Mitochondrial Ca2+ Uniporter led to oscillation inhibition. Thus, the model predicts that alternative mechanisms can yield [Ca2+]Cyt oscillations in non-excitable cells, and, under certain conditions, the ER-mitochondria µd can play a regulatory role.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mitocondrias/metabolismo , Modelos Teóricos , Animales , Humanos , Modelos Biológicos
20.
Microcirculation ; 15(8): 813-34, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18608992

RESUMEN

Over the last 30 years nitric oxide (NO) has emerged as a key signaling molecule involved in a number of physiological functions, including in the regulation of microcirculatory tone. Despite significant scientific contributions, fundamental questions about NO's role in the microcirculation remain unanswered. Mathematical modeling can assist in investigations of microcirculatory NO physiology and address experimental limitations in quantifying vascular NO concentrations. The number of mathematical models investigating the fate of NO in the vasculature has increased over the last few years, and new models are continuously emerging, incorporating an increasing level of complexity and detail. Models investigate mechanisms that affect NO availability in health and disease. They examine the significance of NO release from nonendothelial sources, the effect of transient release, and the complex interaction of NO with other substances, such as heme-containing proteins and reactive oxygen species. Models are utilized to test and generate hypotheses for the mechanisms that regulate NO-dependent signaling in the microcirculation.


Asunto(s)
Microcirculación , Modelos Cardiovasculares , Óxido Nítrico/sangre , Animales , GMP Cíclico/sangre , Cisteína/metabolismo , Difusión , Endotelio Vascular/metabolismo , Eritrocitos/metabolismo , Guanilato Ciclasa/metabolismo , Hemo/metabolismo , Hemoglobinas/metabolismo , Humanos , Microvasos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Guanilil Ciclasa Soluble , Compuestos de Sulfhidrilo/metabolismo , Vasodilatación/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA