Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 6405, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36302779

RESUMEN

Resistance artery vasodilation in response to hypoxia is essential for matching tissue oxygen and demand. In hypoxia, erythrocytic hemoglobin tetramers produce nitric oxide through nitrite reduction. We hypothesized that the alpha subunit of hemoglobin expressed in endothelium also facilitates nitrite reduction proximal to smooth muscle. Here, we create two mouse strains to test this: an endothelial-specific alpha globin knockout (EC Hba1Δ/Δ) and another with an alpha globin allele mutated to prevent alpha globin's inhibitory interaction with endothelial nitric oxide synthase (Hba1WT/Δ36-39). The EC Hba1Δ/Δ mice had significantly decreased exercise capacity and intracellular nitrite consumption in hypoxic conditions, an effect absent in Hba1WT/Δ36-39 mice. Hypoxia-induced vasodilation is significantly decreased in arteries from EC Hba1Δ/Δ, but not Hba1WT/Δ36-39 mice. Hypoxia also does not lower blood pressure in EC Hba1Δ/Δ mice. We conclude the presence of alpha globin in resistance artery endothelium acts as a nitrite reductase providing local nitric oxide in response to hypoxia.


Asunto(s)
Óxido Nítrico , Nitrito Reductasas , Ratones , Animales , Nitrito Reductasas/genética , Nitrito Reductasas/farmacología , Óxido Nítrico/farmacología , Nitritos , Globinas alfa/genética , Hipoxia , Endotelio Vascular , Hemoglobinas/genética , Vasodilatación/fisiología
2.
Kidney Int ; 98(3): 630-644, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32446934

RESUMEN

Kidney function and blood pressure homeostasis are regulated by purinergic signaling mechanisms. These autocrine/paracrine signaling pathways are initiated by the release of cellular ATP, which influences kidney hemodynamics and steady-state renin secretion from juxtaglomerular cells. However, the mechanism responsible for ATP release that supports tonic inputs to juxtaglomerular cells and regulates renin secretion remains unclear. Pannexin 1 (Panx1) channels localize to both afferent arterioles and juxtaglomerular cells and provide a transmembrane conduit for ATP release and ion permeability in the kidney and the vasculature. We hypothesized that Panx1 channels in renin-expressing cells regulate renin secretion in vivo. Using a renin cell-specific Panx1 knockout model, we found that male Panx1 deficient mice exhibiting a heightened activation of the renin-angiotensin-aldosterone system have markedly increased plasma renin and aldosterone concentrations, and elevated mean arterial pressure with altered peripheral hemodynamics. Following ovariectomy, female mice mirrored the male phenotype. Furthermore, constitutive Panx1 channel activity was observed in As4.1 renin-secreting cells, whereby Panx1 knockdown reduced extracellular ATP accumulation, lowered basal intracellular calcium concentrations and recapitulated a hyper-secretory renin phenotype. Moreover, in response to stress stimuli that lower blood pressure, Panx1-deficient mice exhibited aberrant "renin recruitment" as evidenced by reactivation of renin expression in pre-glomerular arteriolar smooth muscle cells. Thus, renin-cell Panx1 channels suppress renin secretion and influence adaptive renin responses when blood pressure homeostasis is threatened.


Asunto(s)
Conexinas , Renina , Adenosina Trifosfato , Animales , Presión Sanguínea , Conexinas/genética , Femenino , Homeostasis , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética
3.
J Immunol ; 204(11): 2995-3007, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32312847

RESUMEN

The proinflammatory cytokine IL-1ß is a significant risk factor in cardiovascular disease that can be targeted to reduce major cardiovascular events. IL-1ß expression and release are tightly controlled by changes in intracellular Ca2+ ([Ca2+]i), which has been associated with ATP release and purinergic signaling. Despite this, the mechanisms that regulate these changes have not been identified. The pannexin 1 (Panx1) channels have canonically been implicated in ATP release, especially during inflammation. We examined Panx1 in human umbilical vein endothelial cells following treatment with the proinflammatory cytokine TNF-α. Analysis by whole transcriptome sequencing and immunoblot identified a dramatic increase in Panx1 mRNA and protein expression that is regulated in an NF-κB-dependent manner. Furthermore, genetic inhibition of Panx1 reduced the expression and release of IL-1ß. We initially hypothesized that increased Panx1-mediated ATP release acted in a paracrine fashion to control cytokine expression. However, our data demonstrate that IL-1ß expression was not altered after direct ATP stimulation in human umbilical vein endothelial cells. Because Panx1 forms a large pore channel, we hypothesized it may permit Ca2+ diffusion into the cell to regulate IL-1ß. High-throughput flow cytometric analysis demonstrated that TNF-α treatments lead to elevated [Ca2+]i, corresponding with Panx1 membrane localization. Genetic or pharmacological inhibition of Panx1 reduced TNF-α-associated increases in [Ca2+]i, blocked phosphorylation of the NF-κB-p65 protein, and reduced IL-1ß transcription. Taken together, the data in our study provide the first evidence, to our knowledge, that [Ca2+]i regulation via the Panx1 channel induces a feed-forward effect on NF-κB to regulate IL-1ß synthesis and release in endothelium during inflammation.


Asunto(s)
Conexinas/metabolismo , Endotelio Vascular/metabolismo , Inflamación/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Señalización del Calcio , Conexinas/genética , Endotelio Vascular/patología , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/metabolismo , Espacio Intracelular , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Secuenciación del Exoma
4.
Circ Res ; 126(2): 232-242, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31801409

RESUMEN

RATIONALE: Increasing prevalence of obesity and its associated risk with cardiovascular diseases demands a better understanding of the contribution of different cell types within this complex disease for developing new treatment options. Previous studies could prove a fundamental role of FTO (fat mass and obesity-associated protein) within obesity; however, its functional role within different cell types is less understood. OBJECTIVES: We identify endothelial FTO as a previously unknown central regulator of both obesity-induced metabolic and vascular alterations. METHODS AND RESULTS: We generated endothelial Fto-deficient mice and analyzed the impact of obesity on those mice. While the loss of endothelial FTO did not influence the development of obesity and dyslipidemia, it protected mice from high-fat diet-induced glucose intolerance and insulin resistance by increasing AKT (protein kinase B) phosphorylation in endothelial cells and skeletal muscle. Furthermore, loss of endothelial FTO prevented the development of obesity-induced hypertension by preserving myogenic tone in resistance arteries. In Fto-deficient arteries, microarray analysis identified upregulation of L-Pgds with significant increases in prostaglandin D2 levels. Blockade of prostaglandin D2 synthesis inhibited the myogenic tone protection in resistance arteries of endothelial Fto-deficient mice on high-fat diet; conversely, direct addition of prostaglandin D2 rescued myogenic tone in high-fat diet-fed control mice. Myogenic tone was increased in obese human arteries with FTO inhibitors or prostaglandin D2 application. CONCLUSIONS: These data identify endothelial FTO as a previously unknown regulator in the development of obesity-induced metabolic and vascular changes, which is independent of its known function in regulation of obesity.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Endotelio Vascular/metabolismo , Obesidad/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Arterias/metabolismo , Arterias/patología , Endotelio Vascular/patología , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Masculino , Ratones , Tono Muscular , Músculo Esquelético/metabolismo , Obesidad/genética , Obesidad/patología , Prostaglandina D2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Circ Res ; 124(10): 1473-1481, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30900949

RESUMEN

RATIONALE: Resistance arteries and conduit arteries rely on different relative contributions of endothelial-derived hyperpolarization versus nitric oxide to achieve dilatory heterocellular signaling. Anatomically, resistance arteries use myoendothelial junctions (MEJs), endothelial cell projections that make contact with smooth muscle cells. Conduit arteries have very few to no MEJs. OBJECTIVE: Determine if the presence of MEJs in conduit arteries can alter heterocellular signaling. METHODS AND RESULTS: We previously demonstrated that PAI-1 (plasminogen activator inhibitor-1) can regulate formation of MEJs. Thus, we applied pluronic gel containing PAI-1 directly to conduit arteries (carotid arteries) to determine if this could induce formation of MEJs. We found a significant increase in endothelial cell projections resembling MEJs that correlated with increased biocytin dye transfer from endothelial cells to smooth muscle cells. Next, we used pressure myography to investigate whether these structural changes were accompanied by a functional change in vasodilatory signaling. Interestingly, PAI-1-treated carotids underwent a switch from a conduit to resistance artery vasodilatory profile via diminished nitric oxide signaling and increased endothelial-derived hyperpolarization signaling in response to the endothelium-dependent agonists acetylcholine and NS309. After PAI-1 application, we also found a significant increase in carotid expression of endothelial alpha globin, a protein predominantly expressed in resistance arteries. Carotids from mice with PAI-1, but lacking alpha globin (Hba1-/-), demonstrated that l-nitro-arginine methyl ester, an inhibitor of nitric oxide signaling, was able to prevent arterial relaxation. CONCLUSIONS: The presence or absence of MEJs is an important determinant for influencing heterocellular communication in the arterial wall. In particular, alpha globin expression, induced within newly formed endothelial cell projections, may influence the balance between endothelial-derived hyperpolarization and nitric oxide-mediated vasodilation.


Asunto(s)
Arterias Carótidas/efectos de los fármacos , Comunicación Celular/fisiología , Células Endoteliales/efectos de los fármacos , Uniones Intercelulares/fisiología , Músculo Liso Vascular/citología , Vasodilatación/fisiología , Acetilcolina/farmacología , Animales , Arterias Carótidas/fisiología , Comunicación Celular/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Indoles/farmacología , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/metabolismo , Masculino , Ratones , Miografía/métodos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Oximas/farmacología , Inhibidor 1 de Activador Plasminogénico/farmacología , Inhibidores de Serina Proteinasa/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Globinas alfa/metabolismo
6.
J Biol Chem ; 294(17): 6940-6956, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30814251

RESUMEN

Pannexin 1 (PANX1)-mediated ATP release in vascular smooth muscle coordinates α1-adrenergic receptor (α1-AR) vasoconstriction and blood pressure homeostasis. We recently identified amino acids 198-200 (YLK) on the PANX1 intracellular loop that are critical for α1-AR-mediated vasoconstriction and PANX1 channel function. We report herein that the YLK motif is contained within an SRC homology 2 domain and is directly phosphorylated by SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) at Tyr198 We demonstrate that PANX1-mediated ATP release occurs independently of intracellular calcium but is sensitive to SRC family kinase (SFK) inhibition, suggestive of channel regulation by tyrosine phosphorylation. Using a PANX1 Tyr198-specific antibody, SFK inhibitors, SRC knockdown, temperature-dependent SRC cells, and kinase assays, we found that PANX1-mediated ATP release and vasoconstriction involves constitutive phosphorylation of PANX1 Tyr198 by SRC. We specifically detected SRC-mediated Tyr198 phosphorylation at the plasma membrane and observed that it is not enhanced or induced by α1-AR activation. Last, we show that PANX1 immunostaining is enriched in the smooth muscle layer of arteries from hypertensive humans and that Tyr198 phosphorylation is detectable in these samples, indicative of a role for membrane-associated PANX1 in small arteries of hypertensive humans. Our discovery adds insight into the regulation of PANX1 by post-translational modifications and connects a significant purinergic vasoconstriction pathway with a previously identified, yet unexplored, tyrosine kinase-based α1-AR constriction mechanism. This work implicates SRC-mediated PANX1 function in normal vascular hemodynamics and suggests that Tyr198-phosphorylated PANX1 is involved in hypertensive vascular pathology.


Asunto(s)
Tirosina/metabolismo , Familia-src Quinasas/metabolismo , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Conexinas/efectos de los fármacos , Conexinas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Fenilefrina/farmacología , Fosforilación , Proto-Oncogenes Mas , Familia-src Quinasas/química
7.
Arterioscler Thromb Vasc Biol ; 38(9): 2065-2078, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026274

RESUMEN

Objective- Sympathetic nerve innervation of vascular smooth muscle cells (VSMCs) is a major regulator of arteriolar vasoconstriction, vascular resistance, and blood pressure. Importantly, α-adrenergic receptor stimulation, which uniquely couples with Panx1 (pannexin 1) channel-mediated ATP release in resistance arteries, also requires localization to membrane caveolae. Here, we test whether localization of Panx1 to Cav1 (caveolin-1) promotes channel function (stimulus-dependent ATP release and adrenergic vasoconstriction) and is important for blood pressure homeostasis. Approach and Results- We use in vitro VSMC culture models, ex vivo resistance arteries, and a novel inducible VSMC-specific Cav1 knockout mouse to probe interactions between Panx1 and Cav1. We report that Panx1 and Cav1 colocalized on the VSMC plasma membrane of resistance arteries near sympathetic nerves in an adrenergic stimulus-dependent manner. Genetic deletion of Cav1 significantly blunts adrenergic-stimulated ATP release and vasoconstriction, with no direct influence on endothelium-dependent vasodilation or cardiac function. A significant reduction in mean arterial pressure (total=4 mm Hg; night=7 mm Hg) occurred in mice deficient for VSMC Cav1. These animals were resistant to further blood pressure lowering using a Panx1 peptide inhibitor Px1IL2P, which targets an intracellular loop region necessary for channel function. Conclusions- Translocalization of Panx1 to Cav1-enriched caveolae in VSMCs augments the release of purinergic stimuli necessary for proper adrenergic-mediated vasoconstriction and blood pressure homeostasis.


Asunto(s)
Presión Sanguínea/fisiología , Caveolina 1/metabolismo , Conexinas/metabolismo , Homeostasis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenosina Trifosfato/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Masculino , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/inervación , Fenilefrina/farmacología , Sistema Nervioso Simpático/fisiología , Vasoconstricción/fisiología
8.
Hypertension ; 68(6): 1494-1503, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27802421

RESUMEN

The ability of hemoglobin to scavenge the potent vasodilator nitric oxide (NO) in the blood has been well established as a mechanism of vascular tone homeostasis. In endothelial cells, the alpha chain of hemoglobin (hereafter, alpha globin) and endothelial NO synthase form a macromolecular complex, providing a sink for NO directly adjacent to the production source. We have developed an alpha globin mimetic peptide (named HbαX) that displaces endogenous alpha globin and increases bioavailable NO for vasodilation. Here we show that, in vivo, HbαX administration increases capillary oxygenation and blood flow in arterioles acutely and produces a sustained decrease in systolic blood pressure in normal and angiotensin II-induced hypertensive states. HbαX acts with high specificity and affinity to endothelial NO synthase, without toxicity to liver and kidney and no effect on p50 of O2 binding in red blood cells. In human vasculature, HbαX blunts vasoconstrictive response to cumulative doses of phenylephrine, a potent constricting agent. By binding to endothelial NO synthase and displacing endogenous alpha globin, HbαX modulates important metrics of vascular function, increasing vasodilation and flow in the resistance vasculature.


Asunto(s)
Hipertensión/fisiopatología , Óxido Nítrico Sintasa/metabolismo , Resistencia Vascular/fisiología , Vasodilatadores/farmacología , Globinas alfa/metabolismo , Angiotensina II/farmacología , Animales , Velocidad del Flujo Sanguíneo/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Hemodinámica/efectos de los fármacos , Humanos , Ratones , Distribución Aleatoria , Resistencia Vascular/efectos de los fármacos
9.
Nat Commun ; 6: 7965, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242575

RESUMEN

Inflammatory cell recruitment to local sites of tissue injury and/or infection is controlled by a plethora of signalling processes influencing cell-to-cell interactions between the vascular endothelial cells (ECs) in post-capillary venules and circulating leukocytes. Recently, ATP-sensitive P2Y purinergic receptors have emerged as downstream regulators of EC activation in vascular inflammation. However, the mechanism(s) regulating cellular ATP release in this response remains elusive. Here we report that the ATP-release channel Pannexin1 (Panx1) opens downstream of EC activation by TNF-α. This process involves activation of type-1 TNF receptors, recruitment of Src family kinases (SFK) and SFK-dependent phosphorylation of Panx1. Using an inducible, EC-specific Panx1 knockout mouse line, we report a previously unidentified role for Panx1 channels in promoting leukocyte adhesion and emigration through the venous wall during acute systemic inflammation, placing Panx1 channels at the centre of cytokine crosstalk with purinergic signalling in the endothelium.


Asunto(s)
Conexinas/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Leucocitos/fisiología , Proteínas del Tejido Nervioso/metabolismo , Migración Transendotelial y Transepitelial , Adenosina Trifosfato/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Endotelio Vascular/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones Endogámicos C57BL , Fosforilación , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba , Vénulas/inmunología , Familia-src Quinasas/metabolismo
10.
Sci Signal ; 8(364): ra17, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25690012

RESUMEN

Both purinergic signaling through nucleotides such as ATP (adenosine 5'-triphosphate) and noradrenergic signaling through molecules such as norepinephrine regulate vascular tone and blood pressure. Pannexin1 (Panx1), which forms large-pore, ATP-releasing channels, is present in vascular smooth muscle cells in peripheral blood vessels and participates in noradrenergic responses. Using pharmacological approaches and mice conditionally lacking Panx1 in smooth muscle cells, we found that Panx1 contributed to vasoconstriction mediated by the α1 adrenoreceptor (α1AR), whereas vasoconstriction in response to serotonin or endothelin-1 was independent of Panx1. Analysis of the Panx1-deficient mice showed that Panx1 contributed to blood pressure regulation especially during the night cycle when sympathetic nervous activity is highest. Using mimetic peptides and site-directed mutagenesis, we identified a specific amino acid sequence in the Panx1 intracellular loop that is essential for activation by α1AR signaling. Collectively, these data describe a specific link between noradrenergic and purinergic signaling in blood pressure homeostasis.


Asunto(s)
Presión Sanguínea/fisiología , Conexinas/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/fisiología , Vasoconstricción/fisiología , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Análisis de Varianza , Animales , Western Blotting , Conexinas/genética , Endotelina-1/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Serotonina/metabolismo , Telemetría
11.
Arterioscler Thromb Vasc Biol ; 34(12): 2594-600, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25278292

RESUMEN

OBJECTIVE: Hemoglobin α (Hb α) and endothelial nitric oxide synthase (eNOS) form a macromolecular complex at myoendothelial junctions; the functional role of this interaction remains undefined. To test if coupling of eNOS and Hb α regulates nitric oxide signaling, vascular reactivity, and blood pressure using a mimetic peptide of Hb α to disrupt this interaction. APPROACH AND RESULTS: In silico modeling of Hb α and eNOS identified a conserved sequence of interaction. By mutating portions of Hb α, we identified a specific sequence that binds eNOS. A mimetic peptide of the Hb α sequence (Hb α X) was generated to disrupt this complex. Using in vitro binding assays with purified Hb α and eNOS and ex vivo proximity ligation assays on resistance arteries, we have demonstrated that Hb α X significantly decreased interaction between eNOS and Hb α. Fluorescein isothiocyanate labeling of Hb α X revealed localization to holes in the internal elastic lamina (ie, myoendothelial junctions). To test the functional effects of Hb α X, we measured cyclic guanosine monophosphate and vascular reactivity. Our results reveal augmented cyclic guanosine monophosphate production and altered vasoconstriction with Hb α X. To test the in vivo effects of these peptides on blood pressure, normotensive and hypertensive mice were injected with Hb α X, which caused a significant decrease in blood pressure; injection of Hb α X into eNOS(-/-) mice had no effect. CONCLUSIONS: These results identify a novel sequence on Hb α that is important for Hb α/eNOS complex formation and is critical for nitric oxide signaling at myoendothelial junctions.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Vasoconstricción/fisiología , Globinas alfa/metabolismo , Secuencia de Aminoácidos , Animales , Presión Sanguínea/fisiología , Células Cultivadas , Simulación por Computador , Secuencia Conservada , Células Endoteliales/metabolismo , Humanos , Uniones Intercelulares/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Resistencia Vascular/fisiología , Globinas alfa/química , Globinas alfa/genética
12.
Nature ; 491(7424): 473-7, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23123858

RESUMEN

Models of unregulated nitric oxide (NO) diffusion do not consistently account for the biochemistry of NO synthase (NOS)-dependent signalling in many cell systems. For example, endothelial NOS controls blood pressure, blood flow and oxygen delivery through its effect on vascular smooth muscle tone, but the regulation of these processes is not adequately explained by simple NO diffusion from endothelium to smooth muscle. Here we report a new model for the regulation of NO signalling by demonstrating that haemoglobin (Hb) α (encoded by the HBA1 and HBA2 genes in humans) is expressed in human and mouse arterial endothelial cells and enriched at the myoendothelial junction, where it regulates the effects of NO on vascular reactivity. Notably, this function is unique to Hb α and is abrogated by its genetic depletion. Mechanistically, endothelial Hb α haem iron in the Fe(3+) state permits NO signalling, and this signalling is shut off when Hb α is reduced to the Fe(2+) state by endothelial cytochrome b5 reductase 3 (CYB5R3, also known as diaphorase 1). Genetic and pharmacological inhibition of CYB5R3 increases NO bioactivity in small arteries. These data reveal a new mechanism by which the regulation of the intracellular Hb α oxidation state controls NOS signalling in non-erythroid cells. This model may be relevant to haem-containing globins in a broad range of NOS-containing somatic cells.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/metabolismo , Transducción de Señal , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Células Cultivadas , Difusión , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Hemoglobinas/genética , Humanos , Hierro/química , Ratones , Óxido Nítrico Sintasa/metabolismo , Oxidación-Reducción , Fragmentos de Péptidos/genética , Fenilefrina/farmacología
13.
J Vasc Res ; 49(5): 405-16, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22739252

RESUMEN

AIMS: Pannexins (Panx) form ATP release channels and it has been proposed that they play an important role in the regulation of vascular tone. However, distribution of Panx across the arterial vasculature is not documented. METHODS: We tested antibodies against Panx1, Panx2 and Panx3 on human embryonic kidney cells (which do not endogenously express Panx proteins) transfected with plasmids encoding each Panx isoform and Panx1(-/-) mice. Each of the Panx antibodies was found to be specific and was tested on isolated arteries using immunocytochemistry. RESULTS: We demonstrated that Panx1 is the primary isoform detected in the arterial network. In large arteries, Panx1 is primarily in endothelial cells, whereas in small arteries and arterioles it localizes primarily to the smooth muscle cells. Panx1 was the predominant isoform expressed in coronary arteries, except in arteries less than 100 µm where Panx3 became detectable. Only Panx3 was expressed in the juxtaglomerular apparatus and cortical arterioles. The pulmonary artery and alveoli had expression of all 3 Panx isoforms. No Panx isoforms were detected at the myoendothelial junctions. CONCLUSION: We conclude that the specific localized expression of Panx channels throughout the vasculature points towards an important role for these channels in regulating the release of ATP throughout the arterial network.


Asunto(s)
Conexinas/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Animales , Vasos Coronarios/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Isoformas de Proteínas/biosíntesis , Arteria Pulmonar/metabolismo , Transfección
14.
Circ Res ; 111(2): 201-11, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22652908

RESUMEN

RATIONALE: Dedifferentiation of vascular smooth muscle cells (VSMC) leading to a proliferative cell phenotype significantly contributes to the development of atherosclerosis. Mitogen-activated protein kinase (MAPK) phosphorylation of proteins including connexin 43 (Cx43) has been associated with VSMC proliferation in atherosclerosis. OBJECTIVE: To investigate whether MAPK phosphorylation of Cx43 is directly involved in VSMC proliferation. METHODS AND RESULTS: We show in vivo that MAPK-phosphorylated Cx43 forms complexes with the cell cycle control proteins cyclin E and cyclin-dependent kinase 2 (CDK2) in carotids of apolipoprotein-E receptor null (ApoE(-/-)) mice and in C57Bl/6 mice treated with platelet-derived growth factor-BB (PDGF). We tested the involvement of Cx43 MAPK phosphorylation in vitro using constructs for full-length Cx43 (Cx43) or the Cx43 C-terminus (Cx43(CT)) and produced null phosphorylation Ser>Ala (Cx43(MK4A)/Cx43(CTMK4A)) and phospho-mimetic Ser>Asp (Cx43(MK4D)/Cx43(CTMK4D)) mutations. Coimmunoprecipitation studies in primary VSMC isolated from Cx43 wild-type (Cx43(+/+)) and Cx43 null (Cx43(-/-)) mice and analytic size exclusion studies of purified proteins identify that interactions between cyclin E and Cx43 requires Cx43 MAPK phosphorylation. We further demonstrate that Cx43 MAPK phosphorylation is required for PDGF-mediated VSMC proliferation. Finally, using a novel knock-in mouse containing Cx43-MK4A mutation, we show in vivo that interactions between Cx43 and cyclin E are lost and VSMC proliferation does not occur after treatment of carotids with PDGF and that neointima formation is significantly reduced in carotids after injury. CONCLUSIONS: We identify MAPK-phosphorylated Cx43 as a novel interacting partner of cyclin E in VSMC and show that this interaction is critical for VSMC proliferation. This novel interaction may be important in the development of atherosclerotic lesions.


Asunto(s)
Proliferación Celular , Conexina 43/metabolismo , Ciclina E/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Animales , Animales Recién Nacidos , Aterosclerosis/enzimología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Conexina 43/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación/fisiología , Unión Proteica/fisiología
15.
Arterioscler Thromb Vasc Biol ; 32(5): 1271-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22383705

RESUMEN

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1) has previously been shown to be key to the formation of myoendothelial junctions (MEJs) in normal and pathological states (eg, obesity). We therefore sought to identify the mechanism whereby PAI-1 could be selectively accumulated at the MEJ. METHODS AND RESULTS: We identified PAI-1 protein enrichment at the MEJ in obese mice and in response to tumor necrosis factor (TNF-α) with a vascular cell coculture. However, PAI-1 mRNA was also found at the MEJ and transfection with a PAI-1-GFP with TNF-α did not demonstrate trafficking of the protein to the MEJ. We therefore hypothesized the PAI-1 mRNA was being locally translated and identified serpine binding protein-1, which stabilizes PAI-1 mRNA, as being enriched in obese mice and after treatment with TNF-α, whereas Staufen, which degrades PAI-1 mRNA, was absent in obese mice and after TNF-α application. We identified nicotinamide phosphoribosyl transferase as a serpine binding protein-1 binding partner with a functional τ-like microtubule binding domain. Application of peptides against the microtubule binding domain significantly decreased the number of MEJs and the amount of PAI-1 at the MEJ. CONCLUSIONS: We conclude that PAI-1 can be locally translated at the MEJ as a result of a unique mRNA binding protein complex.


Asunto(s)
Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Músculo Liso Vascular/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Endotelio Vascular/ultraestructura , Uniones Intercelulares , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Electrónica de Transmisión , Músculo Liso Vascular/ultraestructura , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transcripción Genética
16.
Circ Res ; 109(1): 80-5, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21546608

RESUMEN

RATIONALE: The coordination of vascular smooth muscle cell constriction plays an important role in vascular function, such as regulation of blood pressure; however, the mechanism responsible for vascular smooth muscle cell communication is not clear in the resistance vasculature. Pannexins (Panx) are purine-releasing channels permeable to the vasoconstrictor ATP and thus may play a role in the coordination of vascular smooth muscle cell constriction. OBJECTIVE: We investigated the role of pannexins in phenylephrine- and KCl-mediated constriction of resistance arteries. METHODS AND RESULTS: Western blot, immunohistochemistry, and immunogold labeling coupled to scanning and transmission electron microscopy revealed the presence of Panx1 but not Panx2 or Panx3 in thoracodorsal resistance arteries. Functionally, the contractile response of pressurized thoracodorsal resistance arteries to phenylephrine was decreased significantly by multiple Panx inhibitors (mefloquine, probenecid, and (10)Panx1), ectonucleotidase (apyrase), and purinergic receptor inhibitors (suramin and reactive blue-2). Electroporation of thoracodorsal resistance arteries with either Panx1-green fluorescent protein or Panx1 small interfering RNA showed enhanced and decreased constriction, respectively, in response to phenylephrine. Lastly, the Panx inhibitors did not alter constriction in response to KCl. This result is consistent with coimmunoprecipitation experiments from thoracodorsal resistance arteries, which suggested an association between Panx1 and α1D-adrenergic receptor. CONCLUSIONS: Our data demonstrate for the first time a key role for Panx1 in resistance arteries by contributing to the coordination of vascular smooth muscle cell constriction and possibly to the regulation of blood pressure.


Asunto(s)
Conexinas/fisiología , Proteínas del Tejido Nervioso/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Vasoconstricción/fisiología , Animales , Conexinas/análisis , Ratones , Músculo Liso Vascular/química , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/química , Proteínas del Tejido Nervioso/análisis , Fenilefrina/farmacología , Cloruro de Potasio/farmacología , Resistencia Vascular , Vasoconstricción/efectos de los fármacos
17.
Microcirculation ; 18(3): 163-71, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21418375

RESUMEN

OBJECTIVES: Vascular obstructive events can be partially compensated for by remodeling processes that increase vessel diameter and collateral tortuosity. However, methods for visualizing remodeling events in vivo and with temporal comparisons from the same animal remain elusive. METHODS: Using a novel infrared conjugated polyethylene glycol dye, we investigated the possibility of intravital vascular imaging of the mouse ear before and after ligation of the primary feeder artery. For comparison, we used two different mouse models known to have impaired vascular remodeling after ligation (i.e., aged and PAI-1(-/-) mice). The results obtained with the infrared dye were confirmed using immunofluorescence labeling of the ear microvasculature with confocal microscopy. RESULTS: After ligation, increases in vessel diameter (between 10% and 60%) and tortuosity (approximately 15%) were observed in C57Bl/6 mice using both the infrared dye and the immunofluorescence technique. However, aged C57Bl/6 and PAI-1(-/-) mice did not show vascular remodeling following ligation. CONCLUSIONS: Vascular remodeling can be visualized and accurately quantified using a new infrared dye in vivo. This analysis technique could be generally employed for quantitative investigations of changes in vascular remodeling.


Asunto(s)
Arterias/patología , Colorantes , Dilatación Patológica/patología , Sondas Moleculares , Animales , Dilatación Patológica/diagnóstico , Modelos Animales de Enfermedad , Ligadura , Métodos , Ratones , Sondas Moleculares/química
18.
Arterioscler Thromb Vasc Biol ; 31(2): 399-407, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21071693

RESUMEN

OBJECTIVE: To determine whether S-nitrosylation of connexins (Cxs) modulates gap junction communication between endothelium and smooth muscle. METHODS AND RESULTS: Heterocellular communication is essential for endothelium control of smooth muscle constriction; however, the exact mechanism governing this action remains unknown. Cxs and NO have been implicated in regulating heterocellular communication in the vessel wall. The myoendothelial junction serves as a conduit to facilitate gap junction communication between endothelial cells and vascular smooth muscle cells within the resistance vasculature. By using isolated vessels and a vascular cell coculture, we found that Cx43 is constitutively S-nitrosylated on cysteine 271 because of active endothelial NO synthase compartmentalized at the myoendothelial junction. Conversely, we found that stimulation of smooth muscle cells with the constrictor phenylephrine caused Cx43 to become denitrosylated because of compartmentalized S-nitrosoglutathione reductase, which attenuated channel permeability. We measured S-nitrosoglutathione breakdown and NO(x) concentrations at the myoendothelial junction and found S-nitrosoglutathione reductase activity to precede NO release. CONCLUSIONS: This study provides evidence for compartmentalized S-nitrosylation/denitrosylation in the regulation of smooth muscle cell to endothelial cell communication.


Asunto(s)
Comunicación Celular/fisiología , Conexina 43/metabolismo , Endotelio Vascular/citología , Uniones Comunicantes/metabolismo , Glutatión Reductasa/metabolismo , Músculo Liso Vascular/citología , S-Nitrosoglutatión/metabolismo , Alcohol Deshidrogenasa , Animales , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Glutatión Reductasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenilefrina/farmacología , Resistencia Vascular/fisiología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasoconstrictores/farmacología
19.
J Cell Biochem ; 110(3): 772-82, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20512937

RESUMEN

Connexins (Cxs) and gap junction (GJ)-mediated communication have been linked with the regulation of cell cycle traverse. However, it is not clear whether Cx expression or GJ channel function are the key mediators in this process or at what stage this regulation may occur. We therefore tested the hypothesis that enhanced Cx expression could alter the rate of cell cycle traverse independently of GJ channel function. Sodium butyrate (NaBu) or anti-arrhythmic peptide (AAP10) were used to enhance Cx expression in HeLa cells stably expressing Cx43 (HeLa-43) and primary cultures of human fibroblasts (HFF) that predominantly express Cx43. To reduce GJ-mediated communication, 18-alpha-glycyrrhetinic acid (GA) was used. In HeLa-43 and HFF cells, NaBu and AAP10 enhanced Cx43 expression and increased channel function, while GA reduced GJ-mediated communication but did not significantly alter Cx43 expression levels. Timelapse microscopy and flow cytometry of HeLa-WT (wild-type, Cx deficient) and HeLa-43 cells dissected cell cycle traverse and enabled measurements of intra-mitotic time and determined levels of G1 arrest. Enhanced Cx43 expression increased mitotic durations corresponding with a G1 delay in cell cycle, which was linked to an increase in expression of the cell cycle inhibitor p21(waf1/cip1) in both HeLa-43 and HFF cells. Reductions in Cx43 channel function did not abrogate these responses, indicating that GJ channel function was not a critical factor in reducing cell proliferation in either cell type. We conclude that enhanced Cx43 expression and not GJ-mediated communication, is involved in regulating cell cycle traverse.


Asunto(s)
Ciclo Celular/fisiología , Conexina 43/metabolismo , Uniones Comunicantes/fisiología , Mitosis/fisiología , Western Blotting , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunohistoquímica , Microscopía , Tiempo
20.
Circ Res ; 106(6): 1092-102, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20133900

RESUMEN

RATIONALE: Plasminogen activator inhibitor-1 (PAI-1) is a biomarker for several vascular disease states; however, its target of action within the vessel wall is undefined. OBJECTIVE: Determine the ability of PAI-1 to regulate myoendothelial junction (MEJ) formation. METHODS AND RESULTS: MEJs are found throughout the vasculature linking endothelial cells (ECs) and vascular smooth muscle cells. Using a vascular cell coculture we isolated MEJ fractions and performed two-dimensional differential gel electrophoresis. Mass spectrometry identified PAI-1 as being enriched within MEJ fractions, which we confirmed in vivo. In the vascular cell coculture, recombinant PAI-1 added to the EC monolayer significantly increased MEJs. Conversely, addition of a PAI-1 monoclonal antibody to the EC monolayer reduced the number of MEJs. This was also observed in vivo where mice fed a high fat diet had increased PAI-1 and MEJs and the number of MEJs in coronary arterioles of PAI-1(-/-) mice was significantly reduced when compared to C57Bl/6 mice. The presence of MEJs in PAI-1(-/-) coronary arterioles was restored when their hearts were transplanted into and exposed to the circulation of C57Bl/6 mice. Application of biotin-conjugated PAI-1 to the EC monolayer in vitro confirmed the ability of luminal PAI-1 to translocate to the MEJ. Functionally, phenylephrine-induced heterocellular calcium communication in the vascular cell coculture was temporally enhanced when recombinant PAI-1 was present, and prolonged when PAI-1 was absent. CONCLUSION: Our data implicate circulating PAI-1 as a key regulator of MEJ formation and a potential target for pharmacological intervention in diseases with vascular abnormalities (eg, diabetes mellitus).


Asunto(s)
Comunicación Celular , Células Endoteliales/metabolismo , Uniones Intercelulares/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Animales , Anticuerpos Monoclonales , Arteriolas/metabolismo , Señalización del Calcio , Células Cultivadas , Técnicas de Cocultivo , Vasos Coronarios/metabolismo , Electroforesis en Gel Bidimensional , Células Endoteliales/ultraestructura , Trasplante de Corazón , Uniones Intercelulares/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/inmunología , Transporte de Proteínas , Proteómica/métodos , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA