Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nitric Oxide ; 111-112: 1-13, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33813098

RESUMEN

Endothelial dysfunction, characterised by impaired nitric oxide (NO) bioavailability, arises in response to a variety of cardiovascular risk factors and precedes atherosclerosis. NO is produced by tight regulation of endothelial nitric oxide synthase (eNOS) activity in response to vasodilatory stimuli. This regulation of eNOS is mediated in part by store-operated calcium entry (SOCE). We hypothesized that both ATP- and flow-induced eNOS activation are regulated by SOCE derived from Orai1 channels and members of the transient receptor potential canonical (TRPC) channel family. Bovine aortic endothelial cells (BAECs) were pre-treated with pharmacological inhibitors of TRPC channels and Orai1 to examine their effect on calcium signaling and eNOS activation in response to flow and ATP. The peak and sustained ATP-induced calcium signal and the resulting eNOS activation were attenuated by inhibition of TRPC3, which we found to be store operated. TRPC4 blockade reduced the transient peak in calcium concentration following ATP stimulation, but did not significantly reduce eNOS activity. Simultaneous TRPC3 & 4 inhibition reduced flow-induced NO production via alterations in phosphorylation-mediated eNOS activity. Inhibition of TRPC1/6 or Orai1 failed to lower ATP-induced calcium entry or eNOS activation. Our results suggest that TRPC3 is a store-operated channel in BAECs and is the key regulator of ATP-induced eNOS activation, whereas flow stimulation also recruits TRPC4 into the pathway for the synthesis of NO.


Asunto(s)
Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Señalización del Calcio/fisiología , Bovinos , Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
2.
Microcirculation ; 25(6): e12465, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29885064

RESUMEN

OBJECTIVES: The effect of NO on smooth muscle cell contractility is crucial in regulating vascular tone, blood flow, and O2 delivery. Quantitative predictions for interactions between the NO production rate and the myogenic response for microcirculatory blood vessels are lacking. METHODS: We developed a computational model of a branching microcirculatory network with four representative classes of resistance vessels to predict the effect of endothelium-derived NO on the microvascular pressure-flow response. Our model links vessel scale biotransport simulations of NO and O2 delivery to a mechanistic model of autoregulation and myogenic tone in a simplified microcirculatory network. RESULTS: The model predicts that smooth muscle cell NO bioavailability significantly contributes to resting vascular tone of resistance vessels. Deficiencies in NO seen during hypoxia or ischemia lead to a decreased vessel diameter for all classes at a given intravascular pressure. At the network level, NO deficiencies lead to an increase in pressure drop across the vessels studied, a downward shift in the pressure-flow curve, and a decrease in the effective range of the autoregulatory response. CONCLUSIONS: Our model predicts the steady state and transient behavior of resistance vessels to perturbations in blood pressure, including effects of NO bioavailability on vascular regulation.


Asunto(s)
Velocidad del Flujo Sanguíneo , Microcirculación/fisiología , Modelos Teóricos , Músculo Liso Vascular/fisiología , Óxido Nítrico/fisiología , Animales , Presión Sanguínea , Humanos , Resistencia Vascular
3.
Microvasc Res ; 112: 79-86, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28363495

RESUMEN

Interactions between cardiac myoglobin (Mb), nitrite, and nitric oxide (NO) are vital in regulating O2 storage, transport, and NO homeostasis. Production of NO through the reduction of endogenous myocardial nitrite by deoxygenated myoglobin has been shown to significantly reduce myocardial infarction damage and ischemic injury. We developed a mathematical model for a cardiac arteriole and surrounding myocardium to examine the hypothesis that myoglobin switches functions from being a strong NO scavenger to an NO producer via the deoxymyoglobin nitrite reductase pathway. Our results predict that under ischemic conditions of flow, blood oxygen level, and tissue pH, deoxyMb nitrite reduction significantly elevates tissue and smooth muscle cell NO. The size of the effect is consistent at different flow rates, increases with decreasing blood oxygen and tissue pH and, in extreme pathophysiological conditions, NO can even be elevated above the normoxic levels. Our simulations suggest that cardiac deoxyMb nitrite reduction is a plausible mechanism for preserving or enhancing NO levels using endogenous nitrite despite the rate-limiting O2 levels for endothelial NO production. This NO could then be responsible for mitigating deleterious effects under ischemic conditions.


Asunto(s)
Arteriolas/fisiopatología , Circulación Coronaria , Modelos Cardiovasculares , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Mioglobina/metabolismo , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Animales , Velocidad del Flujo Sanguíneo , Hipoxia de la Célula , Simulación por Computador , Humanos , Concentración de Iones de Hidrógeno , Isquemia Miocárdica/sangre , Isquemia Miocárdica/fisiopatología , Análisis Numérico Asistido por Computador , Oxidación-Reducción , Oxígeno/sangre , Flujo Sanguíneo Regional
4.
Nitric Oxide ; 60: 1-9, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27565833

RESUMEN

Nitrite infusion into the bloodstream has been shown to elicit vasodilation and protect against ischemia-reperfusion injury through nitric oxide (NO) release in hypoxic conditions. However, the mechanism by which nitrite-derived NO escapes scavenging by hemoglobin in the erythrocyte has not been fully elucidated, owing in part to the difficulty in measuring the reactions and transport on NO in vivo. We developed a mathematical model for an arteriole and surrounding tissue to examine the hypothesis that dinitrogen trioxide (N2O3) acts as a stable intermediate for preserving NO. Our simulations predict that with hypoxia and moderate nitrite concentrations, the N2O3 pathway can significantly preserve the NO produced by hemoglobin nitrite reductase in the erythrocyte and elevate NO reaching the smooth muscle cells. Nitrite retains its ability to increase NO bioavailability even at varying flow conditions, but there is minimal effect under normoxia or very low nitrite concentrations. Our model demonstrates a viable pathway for reconciling experimental findings of potentially beneficial effects of nitrite infusions despite previous models showing negligible NO elevation associated with hemoglobin nitrite reductase. Our results suggest that additional mechanisms may be needed to explain the efficacy of nitrite-induced vasodilation at low infusion concentrations.


Asunto(s)
Arteriolas/metabolismo , Hipoxia/metabolismo , Óxido Nítrico/metabolismo , Nitritos/farmacología , Óxidos de Nitrógeno/metabolismo , Vasodilatación/fisiología , Animales , Arteriolas/efectos de los fármacos , Disponibilidad Biológica , Velocidad del Flujo Sanguíneo , Modelos Biológicos , Óxidos de Nitrógeno/farmacocinética , Oxígeno/metabolismo , Vasodilatación/efectos de los fármacos
5.
Nitric Oxide ; 52: 1-15, 2016 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-26529478

RESUMEN

We developed a mass transport model for a parallel-plate flow chamber apparatus to predict the concentrations of nitric oxide (NO) and adenine nucleotides (ATP, ADP) produced by cultured endothelial cells (ECs) and investigated how the net rates of production, degradation, and mass transport for these three chemical species vary with changes in wall shear stress (τw). These simulations provide an improved understanding of experimental results obtained with parallel-plate flow chambers and allows quantitative analysis of the relationship between τw, adenine nucleotide concentrations, and NO produced by ECs. Experimental data obtained after altering ATP and ADP concentrations with apyrase were analyzed to quantify changes in the rate of NO production (RNO). The effects of different isoforms of apyrase on ATP and ADP concentrations and nucleotide-dependent changes in RNO could be predicted with the model. A decrease in ATP was predicted with apyrase, but an increase in ADP was simulated due to degradation of ATP. We found that a simple proportional relationship relating a component of RNO to the sum of ATP and ADP provided a close match to the fitted curve for experimentally measured changes in RNO with apyrase. Estimates for the proportionality constant ranged from 0.0067 to 0.0321 µM/s increase in RNO per nM nucleotide concentration, depending on which isoform of apyrase was modeled, with the largest effect of nucleotides on RNO at low τw (<6 dyn/cm(2)).


Asunto(s)
Nucleótidos de Adenina/biosíntesis , Células Endoteliales/metabolismo , Modelos Biológicos , Óxido Nítrico/biosíntesis , Estrés Mecánico , Humanos
6.
J Biol Chem ; 288(7): 4810-8, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23297409

RESUMEN

This investigation was to elucidate the basis for augmentation of nitric-oxide synthesis in neutrophils exposed to hyperbaric oxygen. Hyperoxia increases synthesis of reactive species leading to S-nitrosylation of ß-actin, which causes temporary inhibition of ß(2) integrin adherence. Impaired ß(2) integrin function and actin S-nitrosylation do not occur in neutrophils from mice lacking type-2 nitric-oxide synthase (iNOS) or when incubated with 1400W, an iNOS inhibitor. Similarly, effects of hyperoxia were abrogated in cells depleted of focal adhesion kinase (FAK) by treatment with small inhibitory RNA and those exposed to a specific FAK inhibitor concurrent with hyperoxia. Nitric oxide production doubles within 10 min exposure to hyperoxia but declines to approximately half-maximum production over an additional 10 min. Elevated nitric oxide production did not occur after FAK depletion or inhibition, or when filamentous actin formation was inhibited by cytochalasin D. Intracellular content of iNOS triples over the course of a 45-min exposure to hyperoxia and iNOS dimers increase in a commensurate fashion. Confocal microscopy and immunoprecipitation demonstrated that co-localization/linkage of FAK, iNOS, and filamentous actin increased within 15 min exposure to hyperoxia but then decreased below the control level. Using isolated enzymes in ex vivo preparations an association between iNOS and filamentous actin mediated by FAK could be demonstrated and complex formation was impeded when actin was S-nitrosylated. We conclude that iNOS activity is increased by an FAK-mediated association with actin filaments but peak nitric oxide production is transient due to actin S-nitrosylation during exposure to hyperoxia.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Antígenos CD18/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Neutrófilos/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Citoesqueleto de Actina/química , Actinas/química , Animales , Citoesqueleto/metabolismo , Dimerización , Fibrinógeno/metabolismo , Radicales Libres , Glutatión Transferasa/metabolismo , Ratones , Ratones Noqueados , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/química , Oxígeno/metabolismo , Conejos , Especies de Nitrógeno Reactivo , Solubilidad
7.
Crit Rev Biomed Eng ; 39(5): 397-433, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22196161

RESUMEN

Several apparent paradoxes are evident when one compares mathematical predictions from models of nitric oxide (NO) diffusion and convection in vasculature structures with experimental measurements of NO (or related metabolites) in animal and human studies. Values for NO predicted from mathematical models are generally much lower than in vivo NO values reported in the literature for experiments, specifically with NO microelectrodes positioned at perivascular locations next to different sizes of blood vessels in the microcirculation and NO electrodes inserted into a wide range of tissues supplied by the microcirculation of each specific organ system under investigation. There continues to be uncertainty about the roles of NO scavenging by hemoglobin versus a storage function that may conserve NO, and other signaling targets for NO need to be considered. This review describes model predictions and relevant experimental data with respect to several signaling pathways in the microcirculation that involve NO.


Asunto(s)
Endotelio Vascular/metabolismo , Microcirculación/fisiología , Modelos Cardiovasculares , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología , Adenosina Trifosfato/metabolismo , Animales , Arginina/metabolismo , Transporte Biológico , Comunicación Celular/fisiología , Complejo IV de Transporte de Electrones/metabolismo , Eritrocitos/metabolismo , Guanilato Ciclasa/metabolismo , Hemodinámica/fisiología , Hemoglobinas/metabolismo , Humanos , Hiperoxia/metabolismo , Hipoxia/metabolismo , Óxido Nítrico/sangre , Ratas , Estrés Mecánico
8.
Adv Exp Med Biol ; 701: 271-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21445797

RESUMEN

Recent evidence in the literature suggests that tissues play a greater role than blood in reducing nitrite to NO under ischemic or hypoxic conditions. Our previous mathematical model for coupled NO and O(2) transport around an arteriole, modified to include superoxide generation from dysfunctional endothelium, was developed further to include nitrite reductase activity in blood and tissue. Steady-state radial and axial NO and pO(2) profiles in the arteriole and surrounding tissue were simulated for different blood flow rates and arterial blood pO(2) values. The resulting computer simulations demonstrate that nitrite reductase activity in blood is not a very effective mechanism for conserving NO due to the strong scavenging of NO by hemoglobin. In contrast, nitrite reductase activity in tissue is much more effective in increasing NO bioavailability in the vascular wall and contributes progressively more NO as tissue hypoxia becomes more severe.


Asunto(s)
Arteriolas/metabolismo , Modelos Biológicos , Modelos Teóricos , Óxido Nítrico/metabolismo , Nitrito Reductasas/metabolismo , Oxígeno/metabolismo , Transporte Biológico , Simulación por Computador , Humanos , Nitritos/metabolismo , Consumo de Oxígeno , Superóxidos/metabolismo
9.
Cell Signal ; 87: 110125, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34474112

RESUMEN

BACKGROUND: Protein Kinase C (PKC) is a promiscuous serine/threonine kinase regulating vasodilatory responses in vascular endothelial cells. Calcium-dependent PKCbeta (PKCß) and calcium-independent PKCeta (PKCη) have both been implicated in the regulation and dysfunction of endothelial responses to shear stress and agonists. OBJECTIVE: We hypothesized that PKCß and PKCη differentially modulate shear stress-induced nitric oxide (NO) production by regulating the transduced calcium signals and the resultant eNOS activation. As such, this study sought to characterize the contribution of PKCη and PKCß in regulating calcium signaling and endothelial nitric oxide synthase (eNOS) activation after exposure of endothelial cells to ATP or shear stress. METHODS: Bovine aortic endothelial cells were stimulated in vitro under pharmacological inhibition of PKCß with LY333531 or PKCη targeting with a pseudosubstrate inhibitor. The participation of PKC isozymes in calcium flux, eNOS phosphorylation and NO production was assessed following stimulation with ATP or shear stress. RESULTS: PKCη proved to be a robust regulator of agonist- and shear stress-induced eNOS activation, modulating calcium fluxes and tuning eNOS activity by multi-site phosphorylation. PKCß showed modest influence in this pathway, promoting eNOS activation basally and in response to shear stress. Both PKC isozymes contributed to the constitutive and induced phosphorylation of eNOS. The observed PKC signaling architecture is intricate, recruiting Src to mediate a portion of PKCη's control on calcium entry and eNOS phosphorylation. Elucidation of the importance of PKCη in this pathway was tempered by evidence of a single stimulus producing concurrent phosphorylation at ser1179 and thr497 which are antagonistic to eNOS activity. CONCLUSIONS: We have, for the first time, shown in a single species in vitro that shear stress- and ATP-stimulated NO production are differentially regulated by classical and novel PKCs. This study furthers our understanding of the PKC isozyme interplay that optimizes NO production. These considerations will inform the ongoing design of drugs for the treatment of PKC-sensitive cardiovascular pathologies.


Asunto(s)
Señalización del Calcio , Óxido Nítrico , Animales , Bovinos , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Estrés Mecánico
10.
J Clin Invest ; 117(5): 1249-59, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17476357

RESUMEN

Endothelial progenitor cells (EPCs) are essential in vasculogenesis and wound healing, but their circulating and wound level numbers are decreased in diabetes. This study aimed to determine mechanisms responsible for the diabetic defect in circulating and wound EPCs. Since mobilization of BM EPCs occurs via eNOS activation, we hypothesized that eNOS activation is impaired in diabetes, which results in reduced EPC mobilization. Since hyperoxia activates NOS in other tissues, we investigated whether hyperoxia restores EPC mobilization in diabetic mice through BM NOS activation. Additionally, we studied the hypothesis that impaired EPC homing in diabetes is due to decreased wound level stromal cell-derived factor-1alpha (SDF-1alpha), a chemokine that mediates EPC recruitment in ischemia. Diabetic mice showed impaired phosphorylation of BM eNOS, decreased circulating EPCs, and diminished SDF-1alpha expression in cutaneous wounds. Hyperoxia increased BM NO and circulating EPCs, effects inhibited by the NOS inhibitor N-nitro-L-arginine-methyl ester. Administration of SDF-1alpha into wounds reversed the EPC homing impairment and, with hyperoxia, synergistically enhanced EPC mobilization, homing, and wound healing. Thus, hyperoxia reversed the diabetic defect in EPC mobilization, and SDF-1alpha reversed the diabetic defect in EPC homing. The targets identified, which we believe to be novel, can significantly advance the field of diabetic wound healing.


Asunto(s)
Movimiento Celular , Quimiocinas CXC/fisiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Hiperoxia/metabolismo , Óxido Nítrico/fisiología , Células Madre/metabolismo , Cicatrización de Heridas , Animales , Movimiento Celular/genética , Células Cultivadas , Quimiocina CXCL12 , Diabetes Mellitus Experimental/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Hiperoxia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Células Madre/patología , Cicatrización de Heridas/genética
11.
Nitric Oxide ; 23(4): 335-42, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20719252

RESUMEN

Nitric oxide (NO) produced by the endothelium is involved in the regulation of vascular tone. Decreased NO production or availability has been linked to endothelial dysfunction in hypercholesterolemia and hypertension. Shear stress-induced NO release is a well-established phenomenon, yet the cellular mechanisms of this response are not completely understood. Experimental limitations have hindered direct, real-time measurements of NO under flow conditions. We have overcome these challenges with a new design for a parallel-plate flow chamber. The chamber consists of two compartments, separated by a Transwell® membrane, which isolates a NO recording electrode located in the upper compartment from flow effects. Endothelial cells are grown on the bottom of the membrane, which is inserted into the chamber flush with the upper plate. We demonstrate for the first time direct real-time NO measurements from endothelial cells with controlled variations in shear stress. Step changes in shear stress from 0.1 dyn/cm(2) to 6, 10, or 20 dyn/cm(2) elicited a transient decrease in NO followed by an increase to a new steady state. An analysis of NO transport suggests that the initial decrease is due to the increased removal rate by convection as flow increases. Furthermore, the rate at which the NO concentration approaches the new steady state is related to the time-dependent cellular response rather than transport limitations of the measurement configuration. Our design offers a method for studying the kinetics of the signaling mechanisms linking NO production with shear stress as well as pathological conditions involving changes in NO production or availability.


Asunto(s)
Células Endoteliales/metabolismo , Óxido Nítrico/biosíntesis , Resistencia al Corte , Animales , Aorta/citología , Bovinos , Células Cultivadas , Electrodos , Diseño de Equipo , Citometría de Flujo/instrumentación , Factores de Tiempo
12.
Adv Exp Med Biol ; 645: 7-12, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19227443

RESUMEN

Computer simulations were performed based on a multiple chemical species convection-diffusion model with coupled biochemical reactions for oxygen (O2), nitric oxide (NO), superoxide (O2*-), peroxynitrite (ONOO-), nitrite (NO2-) and nitrate (NO3-) in cylindrical geometry with blood flow through a 30 microm diameter arteriole. Steady state concentration gradients of all chemical species were predicted for different O2*- production rates, superoxide dismutase (SOD) concentrations, and blood flow rates. Effects of additional O2*- production from dysfunctional endothelial nitric oxide synthase (eNOS) were also simulated. The model predicts that convection is essential for characterizing O2 partial pressure gradients (PO2) in the bloodstream and surrounding tissue, but has little direct effect on NO gradients in blood and tissue.


Asunto(s)
Modelos Biológicos , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Superóxidos/metabolismo , Simulación por Computador
13.
Int J Cancer ; 123(2): 464-475, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18449880

RESUMEN

This study investigates the role of tumor nitric oxide (NO) and vascular regulation in tumor ulceration following high-dose tumor necrosis factor-alpha (TNF) treatment. Using TNF-responsive (MethA) and nonresponsive (LL2) mouse tumors, tumor NO concentration was measured with an electrochemical sensor and tumor blood flow by Doppler ultrasound. Mice were also pretreated with a selective inducible nitric oxide synthase (iNOS) inhibitor, 1400 W. Tumors harvested from TNF-treated mice were cryosectioned and immunostained for murine macrophages, or/and iNOS. MethA tumor-bearing mice were depleted of macrophages. Pre- and post-TNF tumor NO levels were measured continuously, and mice were followed for gross tumor response. In MethA tumors, TNF caused a 96% response rate, and tumor NO concentration doubled. Tumor blood flow decreased to 3% of baseline by 4 hr and was sustained at 24 hr and 10 days post-TNF. Selective NO inhibition with 1400 W blocked NO rise and decreased response rate to 38%. MethA tumors showed tumor infiltration by macrophages post-TNF and the pattern of macrophage immunostaining overlapped with iNOS immunostaining. Depletion of macrophages inhibited tumor NO increase and response to TNF. LL2 tumors had a 0% response rate to TNF and exhibited no change in NO concentration. Blood flow decreased to 2% of baseline by 4 hr, recovered to 56% by 24 hr and increased to 232% by 10 days. LL2 tumors showed no infiltration by macrophages post-TNF. We conclude that TNF causes tumor infiltrating, macrophage-derived iNOS-mediated tumor NO rise and sustained tumor blood flow shutdown, resulting in tumor ulceration in the responsive tumor.


Asunto(s)
Antineoplásicos/farmacología , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Ácido Clodrónico/administración & dosificación , Ácido Clodrónico/farmacología , Selectina E/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrosarcoma/enzimología , Fibrosarcoma/metabolismo , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Flujo Sanguíneo Regional , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
J Clin Invest ; 115(7): 1816-27, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15951843

RESUMEN

NO has been shown to mediate angiogenesis; however, its role in vessel morphogenesis and maturation is not known. Using intravital microscopy, histological analysis, alpha-smooth muscle actin and chondroitin sulfate proteoglycan 4 staining, microsensor NO measurements, and an NO synthase (NOS) inhibitor, we found that NO mediates mural cell coverage as well as vessel branching and longitudinal extension but not the circumferential growth of blood vessels in B16 murine melanomas. NO-sensitive fluorescent probe 4,5-diaminofluorescein imaging, NOS immunostaining, and the use of NOS-deficient mice revealed that eNOS in vascular endothelial cells is the predominant source of NO and induces these effects. To further dissect the role of NO in mural cell recruitment and vascular morphogenesis, we performed a series of independent analyses. Transwell and under-agarose migration assays demonstrated that endothelial cell-derived NO induces directional migration of mural cell precursors toward endothelial cells. An in vivo tissue-engineered blood vessel model revealed that NO mediates endothelial-mural cell interaction prior to vessel perfusion and also induces recruitment of mural cells to angiogenic vessels, vessel branching, and longitudinal extension and subsequent stabilization of the vessels. These data indicate that endothelial cell-derived NO induces mural cell recruitment as well as subsequent morphogenesis and stabilization of angiogenic vessels.


Asunto(s)
Melanoma Experimental/irrigación sanguínea , Neovascularización Patológica , Óxido Nítrico/fisiología , Animales , Células Cultivadas , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Fisiológica , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/deficiencia , Óxido Nítrico Sintasa/genética , Ingeniería de Tejidos , omega-N-Metilarginina/farmacología
15.
Cell Mol Bioeng ; 11(2): 143-155, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30288177

RESUMEN

INTRODUCTION: Colocalization of endothelial nitric oxide synthase (eNOS) and capacitative Ca2+ entry (CCE) channels in microdomains such as cavaeolae in endothelial cells (ECs) has been shown to significantly affect intracellular Ca2+ dynamics and NO production, but the effect has not been well quantified. METHODS: We developed a two-dimensional continuum model of an EC integrating shear stress-mediated ATP production, intracellular Ca2+ mobilization, and eNOS activation to investigate the effects of spatial colocalization of plasma membrane eNOS and CCE channels on Ca2+ dynamics and NO production in response to flow-induced shear stress. Our model examines the hypothesis that subcellular colocalization of cellular components can be critical for optimal coupling of NO production to blood flow. RESULTS: Our simulations predict that heterogeneity of CCE can result in formation of microdomains with significantly higher Ca2+ compared to the average cytosolic Ca2+. Ca2+ buffers with lower or no mobility further enhanced Ca2+ gradients relative to mobile buffers. Colocalization of eNOS to CCE channels significantly increased NO production. CONCLUSIONS: Our results provide quantitative understanding for the role of spatial heterogeneity and the compartmentalization of signals in regulation of shear stress-induced NO production.

16.
Antioxid Redox Signal ; 9(7): 829-43, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17508909

RESUMEN

The role of nitric oxide (NO) as a highly diffusible free radical gaseous vasodilator is intrinsically linked to the control of blood flow and oxygen (O(2)) delivery to tissue. NO also is involved in regulating mitochondrial O(2) metabolism, growth of new blood vessels, and blood oxygenation through control of respiratory ventilation. Hemoglobin and myoglobin may help to conserve NO for subsequent release of a NO-related vasoactive species under hypoxic conditions. NO has a major role in regulating microvascular O(2), and dysfunctional NO signaling is associated with the pathogenesis of metabolic and cardiovascular diseases.


Asunto(s)
Óxido Nítrico/fisiología , Oxígeno/metabolismo , Transducción de Señal/fisiología , Vasodilatación/fisiología , Animales , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Humanos , Modelos Biológicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Oxígeno/sangre
17.
Nat Biotechnol ; 21(4): 392-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12652312

RESUMEN

Vascular immunotargeting may facilitate the rapid and specific delivery of therapeutic agents to endothelial cells. We investigated whether targeting of an antioxidant enzyme, catalase, to the pulmonary endothelium alleviates oxidative stress in an in vivo model of lung transplantation. Intravenously injected enzymes, conjugated with an antibody to platelet-endothelial cell adhesion molecule-1, accumulate in the pulmonary vasculature and retain their activity during prolonged cold storage and transplantation. Immunotargeting of catalase to donor rats augments the antioxidant capacity of the pulmonary endothelium, reduces oxidative stress, ameliorates ischemia-reperfusion injury, prolongs the acceptable cold ischemia period of lung grafts, and improves the function of transplanted lung grafts. These findings validate the therapeutic potential of vascular immunotargeting as a drug delivery strategy to reduce endothelial injury. Potential applications of this strategy include improving the outcome of clinical lung transplantation and treating a wide variety of endothelial disorders.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Catalasa/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antioxidantes/administración & dosificación , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Inmunoterapia/métodos , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Trasplante de Pulmón/efectos adversos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo
18.
Adv Exp Med Biol ; 599: 117-23, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17727255

RESUMEN

Increasing efforts have been directed towards investigating the involvement of nitric oxide (NO) for normal kidney function. Recently, a crucial role of NO in the development of progressive renal dysfunction has been reported during diabetes and hypertension. Indirect estimation of renal NO production include urinary nitrite/nitrate measurements, but there are several disadvantages of indirect methods since production and bioavailability of NO rarely coincide. Thus, direct measurement of in vivo NO bioavailability is preferred, although these methods are more time consuming and require highly specialized equipment and knowledge. This review focuses on two techniques for in vivo measurement of bioavailable NO in the kidney. We have applied Whalen-type recessed NO microsensors for measurement of NO in the kidney cortex, whereas the hemoglobin-trapping technique seems to be more suitable for NO measurement in the renal medulla. Both methods are robust and reliable, and we discuss advantages and shortcomings of each method.


Asunto(s)
Riñón/química , Óxido Nítrico/análisis , Animales , Técnicas Biosensibles/instrumentación , Hemoglobinas/metabolismo , Isoenzimas/metabolismo , Riñón/anatomía & histología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo
19.
Cell Mol Bioeng ; 10(1): 30-40, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28138348

RESUMEN

Endothelial dysfunction, characterized by decreased production or availability of nitric oxide (NO), is widely believed to be the hallmark of early-stage atherosclerosis. In addition, hypercholesterolemia is considered a major risk factor for development of atherosclerosis and is associated with impaired flow-induced dilation. However, the mechanism by which elevated cholesterol levels leads to decreased production of NO is unclear. NO is released in response to shear stress and agonist-evoked changes in intracellular calcium. Although calcium signaling is complex, we have previously shown that NO production by endothelial nitric oxide synthase (eNOS) is preferentially activated by calcium influx via store-operated channels. We hypothesized that cholesterol enrichment altered this signaling pathway (known as capacitive calcium entry; CCE) ultimately leading to decreased NO. Our results show that cholesterol enrichment abolished ATP-induced eNOS phosphorylation and attenuated the calcium response by the preferential inhibition of CCE. Furthermore, cholesterol enrichment also inhibited shear stress-induced NO production and eNOS phosporylation, consistent with our previous results showing a significant role for ATP autocrine stimulation and subsequent activation of CCE in the endothelial flow response.

20.
Front Physiol ; 8: 1053, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29321744

RESUMEN

Nitric oxide (NO) generated from nitrite through nitrite reductase activity in red blood cells has been proposed to play a major role in hypoxic vasodilation. However, we have previously predicted from mathematical modeling that much more NO can be derived from tissue nitrite reductase activity than from red blood cell nitrite reductase activity. Evidence in the literature suggests that tissue nitrite reductase activity is associated with xanthine oxidoreductase (XOR) and/or aldehyde oxidoreductase (AOR). We investigated the role of XOR and AOR in nitrite-mediated vasodilation from computer simulations and from in vivo exteriorized rat mesentery experiments. Vasodilation responses to nitrite in the superfusion medium bathing the mesentery equilibrated with 5% O2 (normoxia) or zero O2 (hypoxia) at either normal or acidic pH were quantified. Experiments were also conducted following intraperitoneal (IP) injection of nitrite before and after inhibiting XOR with allopurinol or inhibiting AOR with raloxifene. Computer simulations for NO and O2 transport using reaction parameters reported in the literature were also conducted to predict nitrite-dependent NO production from XOR and AOR activity as a function of nitrite concentration, PO2 and pH. Experimentally, the largest arteriolar responses were found with nitrite >10 mM in the superfusate, but no statistically significant differences were found with hypoxic and acidic conditions in the superfusate. Nitrite-mediated vasodilation with IP nitrite injections was reduced or abolished after inhibiting XOR with allopurinol (p < 0.001). Responses to IP nitrite before and after inhibiting AOR with raloxifene were not as consistent. Our mathematical model predicts that under certain conditions, XOR and AOR nitrite reductase activity in tissue can significantly elevate smooth muscle cell NO and can serve as a compensatory pathway when endothelial NO production is limited by hypoxic conditions. Our theoretical and experimental results provide further evidence for a role of tissue nitrite reductases to contribute additional NO to compensate for reduced NO production by endothelial nitric oxide synthase during hypoxia. Our mathematical model demonstrates that under extreme hypoxic conditions with acidic pH, endogenous nitrite levels alone can be sufficient for a functionally significant increase in NO bioavailability. However, these conditions are difficult to achieve experimentally.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA