Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Theor Biol ; 529: 110856, 2021 11 21.
Article in English | MEDLINE | ID: mdl-34363836

ABSTRACT

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.


Subject(s)
Magnetic Resonance Imaging , Theft , Brain , Cerebrovascular Circulation , Hematocrit , Hemodynamics , Oxygen
2.
Proc Natl Acad Sci U S A ; 117(43): 27022-27033, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33051294

ABSTRACT

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.


Subject(s)
Capillaries/physiology , Cerebrovascular Circulation , Microcirculation , Pericytes/physiology , Animals , Arterioles/physiology , Calcium Channels/metabolism , Cerebral Veins/physiology , Mice
3.
Proc Natl Acad Sci U S A ; 117(28): 16626-16637, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601236

ABSTRACT

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.


Subject(s)
Neurons/metabolism , Neurovascular Coupling , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Animals , Brain/blood supply , Brain/metabolism , Cerebrovascular Circulation , Endothelial Cells/chemistry , Endothelial Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/chemistry , Potassium/chemistry , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/genetics , Signal Transduction , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
4.
Sci Rep ; 9(1): 17072, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31745211

ABSTRACT

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.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mitochondria/metabolism , Models, Theoretical , Animals , Humans , Models, Biological
5.
Microcirculation ; 25(2)2018 02.
Article in English | MEDLINE | ID: mdl-29117630

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/physiology , Microvessels/physiology , Models, Biological , Signal Transduction/physiology , Animals , Membrane Potentials/physiology , Mice , Muscle, Skeletal/blood supply , Potassium Channels, Calcium-Activated/physiology , Regional Blood Flow , Synaptic Transmission , Vasodilation
6.
Microcirculation ; 25(1)2018 01.
Article in English | MEDLINE | ID: mdl-29247493

ABSTRACT

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.


Subject(s)
Microvessels/pathology , Potassium Channels, Voltage-Gated/analysis , Animals , CADASIL/physiopathology , Dilatation , Humans , Potassium Channels, Voltage-Gated/physiology , Subarachnoid Hemorrhage/physiopathology
7.
J Biomech ; 65: 40-48, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29054608

ABSTRACT

Fluid-induced shear stresses are involved in the development of cardiovascular tissues. In a tissue engineering framework, this stimulus has also been considered as a mechanical regulator of stem cell differentiation. We recently demonstrated that the fluid-oscillating effect in combination with a physiologically-relevant shear stress magnitude contributes to the formation of stem cell-derived de novo heart valve tissues. However, the range of oscillations necessary to induce favorable gene expression and engineered tissue formation is unknown. In this study, we took a computational approach to establish a range of oscillatory shear stresses that may optimize in vitro valvular tissue growth. Taking a biomimetic approach, three physiologically-relevant flow waveforms from the human: (i) aorta, (ii) pulmonary artery and (iii) superior vena cava were utilized to simulate pulsatile flow conditions within a bioreactor that housed 3 tissue specimens. Results were compared to non-physiological pulsatile flow (NPPF) and cyclic flexure-steady flow (Flex-Flow) conditions. The oscillatory shear index (OSI) was used to quantify the fluid-induced oscillations occurring on the specimen surfaces. The range of mean OSI under the physiological conditions investigated was found to be 0.18 ≤ OSI ≤ 0.23. On the other hand, NPPF and Flex-Flow environments yielded a mean OSI of 0.37 and 0.11 respectively, which were 46% higher and 45% lower than physiological conditions. Moreover, we subsequently conducted OSI-based human bone marrow stem cell (HBMSC) culture experiments which resulted in preferential valvular gene expression and phenotype (significant upregulation of BMP, KLF2A, CD31 and α-SMA using an OSI of 0.23 in comparison to a lower OSI of 0.10 or a higher OSI of 0.38; p < .05). These findings suggest that a distinct range or a "sweet-spot" for physiological OSI exists in the mechanical conditioning of tissue engineered heart valves grown from stem cell sources. We conclude that in vitro heart valve matrix development could be further enhanced by simultaneous exposure of the engineered tissues to physiologically-relevant magnitudes of both fluid-induced oscillations and shear stresses.


Subject(s)
Heart Valves/physiology , Stem Cells/physiology , Tissue Engineering , Aorta/physiology , Biomimetics , Bioreactors , Cell Differentiation , Gene Expression , Humans , Pulmonary Artery/physiology , Pulsatile Flow , Stress, Mechanical , Vena Cava, Superior/physiology
8.
Am J Physiol Heart Circ Physiol ; 312(4): H854-H866, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28130333

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/physiology , Hemoglobin A/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Vasodilation/physiology , Algorithms , Computer Simulation , Erythrocytes/drug effects , Feedback, Physiological , Humans , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Models, Theoretical , Nitric Oxide Synthase Type III/biosynthesis , Potassium Channels, Calcium-Activated/biosynthesis
9.
Phys Rev E ; 93: 042415, 2016 04.
Article in English | MEDLINE | ID: mdl-27176337

ABSTRACT

On the basis of experimental data and mathematical equations in the literature, we remodel the ionic dynamics of smooth muscle cells (SMCs) as an eigensystem formulation, which is valid for investigating finite variations of variables from the equilibrium such as in common experimental operations. This algorithm provides an alternate viewpoint from frequency-domain analysis and enables one to probe functionalities of SMCs' rhythm by means of a resonance-related mechanism. Numerical results show three types of calcium oscillations of SMCs in mesenteric arterioles: spontaneous calcium oscillation, agonist-dependent calcium oscillation, and agonist-dependent calcium spike. For simple single and double SMCs, we demonstrate properties of synchronization among complex signals related to calcium oscillations, and show different correlation relations between calcium and voltage signals for various synchronization and resonance conditions. For practical cell clusters, our analyses indicate that the rhythm of SMCs could (1) benefit enhancements of signal communications among remote cells, (2) respond to a significant calcium peaking against transient stimulations for triggering globally oscillating modes, and (3) characterize the globally oscillating modes via frog-leap (non-molecular-diffusion) calcium waves across inhomogeneous SMCs.


Subject(s)
Mesenteric Arteries/cytology , Models, Biological , Myocytes, Smooth Muscle/cytology , Animals , Arterioles/cytology , Arterioles/physiology , Energy Metabolism , Heart Rate , Linear Models , Mesenteric Arteries/physiology , Rats , Vasoconstriction
10.
Am J Physiol Cell Physiol ; 310(6): C479-90, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26739489

ABSTRACT

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.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Mitochondria/metabolism , Cells, Cultured , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Second Messenger Systems/physiology , Stress, Mechanical
11.
Biophys J ; 108(6): 1566-1576, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25809269

ABSTRACT

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.


Subject(s)
Models, Molecular , TRPV Cation Channels/metabolism , Calcium/metabolism , Computer Simulation , Endothelial Cells/metabolism , Kinetics , Markov Chains , Potassium Channels, Calcium-Activated/metabolism , Stochastic Processes
12.
Nitric Oxide ; 35: 65-71, 2013 Nov 30.
Article in English | MEDLINE | ID: mdl-23994446

ABSTRACT

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.


Subject(s)
Agmatine/pharmacology , Hypertension/physiopathology , Mesenteric Arteries/drug effects , Nitric Oxide/metabolism , Vasodilation/drug effects , Animals , Arginine/pharmacology , Carboxy-Lyases/antagonists & inhibitors , Carboxy-Lyases/metabolism , Male , Mesenteric Arteries/physiology , Rats , Rats, Inbred Dahl , Rats, Sprague-Dawley
13.
J Physiol ; 591(11): 2795-812, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23529128

ABSTRACT

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.


Subject(s)
Cell Surface Extensions/physiology , Endothelial Cells/physiology , Feedback, Physiological , Models, Biological , Myocytes, Smooth Muscle/physiology , Action Potentials , Animals , Calcium/metabolism , Calcium Signaling , Cell Surface Extensions/metabolism , Cell Surface Extensions/ultrastructure , Chlorides/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gap Junctions/metabolism , Gap Junctions/physiology , Gap Junctions/ultrastructure , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mesenteric Arteries/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Potassium/metabolism , Rats , Sodium/metabolism
14.
Nitric Oxide ; 28: 39-46, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23063986

ABSTRACT

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.


Subject(s)
Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Nitric Oxide/analysis , Nitric Oxide/chemistry , Calibration , Fluoresceins/analysis , Fluorescent Dyes/analysis , Kinetics , Oxidation-Reduction , Time Factors
15.
Free Radic Biol Med ; 53(10): 1968-76, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22951977

ABSTRACT

Nitrosation of thiols is thought to be mediated by dinitrogen trioxide (N(2)O(3)) or by nitrogen dioxide radical (()NO(2)). A kinetic study of glutathione (GSH) nitrosation by NO donors in aerated buffered solutions was undertaken. S-nitrosoglutathione (GSNO) formation was assessed spectrophotometrically and by chemiluminescence. The results suggest an increase in the rate of GSNO formation with an increase in GSH with a half-maximum constant EC(50) that depends on NO concentration. Our observed increase in EC(50) with NO concentration suggests a significant contribution of ()NO(2)-mediated nitrosation with the glutathiyl radical as an intermediate in the production of GSNO.


Subject(s)
Glutathione/chemistry , Glutathione/metabolism , Nitric Oxide/chemistry , Nitrogen Dioxide/chemistry , S-Nitrosoglutathione/chemistry , Catalysis , Kinetics , Nitric Oxide/analysis , Nitrosation , Sulfhydryl Compounds/chemistry
16.
J Am Soc Hypertens ; 6(5): 331-7, 2012.
Article in English | MEDLINE | ID: mdl-22995801

ABSTRACT

Bone marrow-derived endothelial progenitor cells (EPCs) constitute an important endogenous system in the maintenance of endothelial integrity and vascular homeostasis. Cardiovascular risk factors are associated with a reduced number and functional capacity of EPCs. Here we investigated the effect of transplantation of bone marrow-derived cells from Dahl salt-resistant rat into age-matched Dahl salt-sensitive (DS) rat on blood pressure, endothelial function, and circulating EPC number. The recipient DS rats were fed a normal (0.5% NaCl, NS) or high-salt (4% NaCl, HS) diet for 6 weeks after bone marrow transplantation (BMT). DS rats on a NS or a HS diet without BMT were used as controls. Hypertensive DS (HS-DS) rat (systolic blood pressure: 213 ± 4 mm Hg vs. 152 ± 4 mm Hg in NS, P < .05) manifested impaired endothelium-dependent relaxation to acetylcholine (EDR), increased gene expression of vascular oxidative stress and proinflamamtory cytokines, and decreased eNOS expression. BMT on HS-DS rat significantly improved EDR and eNOS expression, reduced oxidative stress without reduction in SBP (206 ± 6 mm Hg). Flow cytometry analysis showed that there was no difference in the number of circulating EPCs, demonstrated by expression of EPC markers CD34, cKit, and vascular endothelial growth factor, between hypertensive and normotensive rats. Surprisingly, BMT resulted in a 5- to 10-fold increase in the previously mentioned EPC markers in hypertensive, but not normotensive rat. These results suggest that DS rat has an impaired ability to increase bone marrow-derived EPCs in response to HS diet challenge, which may contribute to endothelial dysfunction.


Subject(s)
Bone Marrow Transplantation/methods , Endothelial Cells , Endothelium, Vascular , Hypertension , Sodium Chloride, Dietary/pharmacology , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Cell Count/methods , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Flow Cytometry , Hypertension/metabolism , Hypertension/physiopathology , Models, Animal , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Rats , Rats, Inbred Dahl , Stem Cells/physiology , Vascular Endothelial Growth Factor A/metabolism , Vasodilation/drug effects
17.
Biophys J ; 102(2): 211-20, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22339857

ABSTRACT

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.


Subject(s)
Calcium Signaling , Calcium/metabolism , Mesenteric Arteries/cytology , Mesenteric Arteries/physiology , Models, Biological , Animals , Electrophysiological Phenomena , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Ion Channels/metabolism , Membrane Potentials , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Rats , Time Factors
18.
Microcirculation ; 19(5): 391-402, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22340204

ABSTRACT

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.


Subject(s)
Cell Communication/physiology , Endothelium, Vascular/physiology , Hemodynamics/physiology , Models, Cardiovascular , Muscle, Smooth, Vascular/physiology , Second Messenger Systems/physiology , Animals , Humans , Ion Transport/physiology , Ions/metabolism
19.
Am J Physiol Cell Physiol ; 302(8): C1226-42, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22277756

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Arteries/drug effects , Arteries/metabolism , Cricetinae , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Feedback/drug effects , Gap Junctions/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Membrane Potentials/drug effects , Mesocricetus , Muscle, Smooth, Vascular/drug effects , Phenylephrine/pharmacology , Potassium Channels, Calcium-Activated/metabolism , Vasoconstriction/drug effects
20.
Phytother Res ; 26(4): 541-5, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21928368

ABSTRACT

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.


Subject(s)
Macrophages/drug effects , Nitric Oxide/antagonists & inhibitors , Plant Extracts/pharmacology , Tanacetum parthenium/chemistry , Animals , Blotting, Western , Carbon Dioxide/metabolism , Cell Line , Cell Survival , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/adverse effects , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...