Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 16: 1125932, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36937050

RESUMEN

Introduction: The disintegrin and metalloproteinase 17 (ADAM17) exhibits α-secretase activity, whereby it can prevent the production of neurotoxic amyloid precursor protein-α (APP). ADAM17 is abundantly expressed in vascular endothelial cells and may act to regulate vascular homeostatic responses, including vasomotor function, vascular wall morphology, and formation of new blood vessels. The role of vascular ADAM17 in neurodegenerative diseases remains poorly understood. Here, we hypothesized that cerebrovascular ADAM17 plays a role in the pathogenesis of Alzheimer's disease (AD). Methods and results: We found that 9-10 months old APP/PS1 mice with b-amyloid accumulation and short-term memory and cognitive deficits display a markedly reduced expression of ADAM17 in cerebral microvessels. Systemic delivery and adeno-associated virus (AAV)-mediated re-expression of ADAM17 in APP/PS1 mice improved cognitive functioning, without affecting b-amyloid plaque density. In isolated and pressurized cerebral arteries of APP/PS1 mice the endothelium-dependent dilation to acetylcholine was significantly reduced, whereas the vascular smooth muscle-dependent dilation to the nitric oxide donor, sodium nitroprusside was maintained when compared to WT mice. The impaired endothelium-dependent vasodilation of cerebral arteries in APP/PS1 mice was restored to normal level by ADAM17 re-expression. The cerebral artery biomechanical properties (wall stress and elasticity) and microvascular network density was not affected by ADAM17 re-expression in the APP/PS1 mice. Additionally, proteomic analysis identified several differentially expressed molecules involved in AD neurodegeneration and neuronal repair mechanisms that were reversed by ADAM17 re-expression. Discussion: Thus, we propose that a reduced ADAM17 expression in cerebral microvessels impairs vasodilator function, which may contribute to the development of cognitive dysfunction in APP/PS1 mice, and that ADAM17 can potentially be targeted for therapeutic intervention in AD.

2.
Brain Res ; 1789: 147947, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35597325

RESUMEN

In this study a manganese-enhanced magnetic resonance imaging (MEMRI) method was developed for mice for measuring axonal transport (AXT) rates in real time in olfactory receptor neurons, which project from the olfactory epithelium to the olfactory neuronal layer of the olfactory bulb. Using this MEMRI method, two major experiments were conducted: 1) an evaluation of the effects of age on AXT rates and 2) an evaluation of the brain-penetrant, microtubule-stabilizing agent, Epothilone D for effect on AXT rates in aged mice. In these studies, we improved upon previous MEMRI approaches to develop a method where real-time measurements (32 time points) of AXT rates in mice can be determined over a single (approximately 100 min) scanning session. In the age comparisons, AXT rates were significantly higher in young (mean age ∼4.0 months old) versus aged (mean age ∼24.5 months old) mice. Moreover, in aged mice, eight weeks of treatment with Epothilone D, (0.3 and 1.0 mg/kg) was associated with statistically significant increases in AXT rates compared to vehicle-treated subjects. These experiments conducted in a living mammalian model (i.e., wild type, C57BL/6 mice), using a new modified MEMRI method, thus provide further evidence that the process of aging leads to decreases in AXT rates in the brain and they further support the argument that microtubule-based therapeutic strategies designed to improve AXT rates have potential for age-related neurological disorders.


Asunto(s)
Transporte Axonal , Manganeso , Animales , Humanos , Imagen por Resonancia Magnética/métodos , Mamíferos , Manganeso/farmacología , Ratones , Ratones Endogámicos C57BL , Microtúbulos
3.
Geroscience ; 44(1): 349-369, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34718985

RESUMEN

Physiological and pathological vascular remodeling is uniquely driven by mechanical forces from blood flow in which wall shear stress (WSS) mechanosensing by the vascular endothelium plays a pivotal role. This study aimed to determine the novel role for a disintegrin and metalloproteinase 17 (ADAM17) in impaired WSS mechanosensing, which was hypothesized to contribute to aging-associated abnormal vascular remodeling. Without changes in arterial blood pressure and blood flow rate, skeletal muscle resistance arteries of aged mice (30-month-old vs. 12-week-old) exhibited impaired WSS mechanosensing and displayed inward hypertrophic arterial remodeling. These vascular changes were recapitulated by in vivo confined, AAV9-mediated overexpression of ADAM17 in the resistance arteries of young mice. An aging-related increase in ADAM17 expression reduced the endothelial junction level of its cleavage substrate, junctional adhesion molecule-A/F11 receptor (JAM-A/F11R). In cultured endothelial cells subjected to steady WSS ADAM17 activation or JAM-A/F11R knockdown inhibited WSS mechanosensing. The ADAM17-activation induced, impaired WSS mechanosensing was normalized by overexpression of ADAM17 cleavage resistant, mutated JAM-AV232Y both in cultured endothelial cells and in resistance arteries of aged mice, in vivo. These data demonstrate a novel role for ADAM17 in JAM-A/F11R cleavage-mediated impaired endothelial WSS mechanosensing and subsequently developed abnormal arterial remodeling in aging. ADAM17 could prove to be a key regulator of WSS mechanosensing, whereby it can also play a role in pathological vascular remodeling in diseases.


Asunto(s)
Proteína ADAM17 , Moléculas de Adhesión Celular , Molécula A de Adhesión de Unión , Receptores de Superficie Celular , Proteína ADAM17/metabolismo , Envejecimiento , Animales , Arterias , Fenómenos Biomecánicos , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales , Endotelio Vascular/metabolismo , Molécula A de Adhesión de Unión/metabolismo , Ratones , Receptores de Superficie Celular/metabolismo , Resistencia al Corte
5.
Chronobiol Int ; 38(1): 1-26, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33342316

RESUMEN

Current hypertension guidelines fail to provide a recommendation on when-to-treat, thus disregarding relevant circadian rhythms that regulate blood pressure (BP) level and 24 h patterning and medication pharmacokinetics and pharmacodynamics. The ideal purpose of ingestion-time (chronopharmacology, i.e. biological rhythm-dependent effects on the kinetics and dynamics of medications, and chronotherapy, i.e. the timing of pharmaceutical and other treatments to optimize efficacy and safety) trials should be to explore the potential impact of endogenous circadian rhythms on the effects of medications. Such investigations and outcome trials mandate adherence to the basic standards of human chronobiology research. In-depth review of the more than 150 human hypertension pharmacology and therapeutic trials published since 1974 that address the differential impact of upon-waking/morning versus at-bedtime/evening schedule of treatment reveals diverse protocols of sometimes suboptimal or defective design and conduct. Many have been "time-of-day," i.e. morning versus evening, rather than circadian-time-based, and some relied on wake-time office BP rather than around-the-clock ambulatory BP measurements (ABPM). Additionally, most past studies have been of too small sample size and thus statistically underpowered. As of yet, there has been no consensual agreement on the proper design, methods and conduct of such trials. This Position Statement recommends ingestion-time hypertension trials to follow minimum guidelines: (i) Recruitment of participants should be restricted to hypertensive individuals diagnosed according to ABPM diagnostic thresholds and of a comparable activity/sleep routine. (ii) Tested treatment-times should be selected according to internal biological time, expressed by the awakening and bed times of the sleep/wake cycle. (iii) ABPM should be the primary or sole method of BP assessment. (iv) The minimum-required features for analysis of the ABPM-determined 24 h BP pattern ought to be the asleep (not "nighttime") BP mean and sleep-time relative BP decline, calculated in reference to the activity/rest cycle per individual. (v) ABPM-obtained BP means should be derived by the so-called adjusted calculation procedure, not by inaccurate arithmetic averages. (vi) ABPM should be performed with validated and calibrated devices at least hourly throughout two or more consecutive 24 h periods (48 h in total) to achieve the highest reproducibility of mean wake-time, sleep-time and 48 h BP values plus the reliable classification of dipping status. (vii) Calculation of minimum required sample size in adherence with proper statistical methods must be provided. (viii) Hypertension chronopharmacology and chronotherapy trials should preferably be randomized double-blind, randomized open-label with blinded-endpoint, or crossover in design, the latter with sufficient washout period between tested treatment-time regimens.


Asunto(s)
Monitoreo Ambulatorio de la Presión Arterial , Hipertensión , Antihipertensivos/uso terapéutico , Presión Sanguínea , Cronoterapia , Ritmo Circadiano , Ingestión de Alimentos , Humanos , Hipertensión/tratamiento farmacológico , Reproducibilidad de los Resultados , Factores de Riesgo , Factores de Tiempo
6.
Antioxid Redox Signal ; 31(14): 1053-1069, 2019 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-30767565

RESUMEN

Significance: Pulmonary arterial hypertension (PAH) is a progressive disease arising from the narrowing of pulmonary arteries (PAs) resulting in high pulmonary arterial blood pressure and ultimately right ventricle (RV) failure. A defining characteristic of PAH is the excessive and unrelenting inward remodeling of PAs that includes increased proliferation, inflammation, and fibrosis. Critical Issues: There is no cure for PAH nor interventions that effectively arrest or reverse PA remodeling, and intensive research over the past several decades has sought to identify novel molecular mechanisms of therapeutic value. Recent Advances: Galectin-3 (Gal-3) is a carbohydrate-binding lectin remarkable for its chimeric structure, composed of an N-terminal oligomerization domain and a C-terminal carbohydrate-recognition domain. Gal-3 has been identified as a regulator of numerous changes in cell behavior that contributes to aberrant PA remodeling, including cell proliferation, inflammation, and fibrosis, but its role in PAH has remained poorly understood until recently. In contrast, pathological roles for Gal-3 have been proposed in cancer and inflammatory and fibroproliferative disorders, such as pulmonary vascular and cardiac fibrosis. Herein, we summarize the recent literature on the role of Gal-3 in the development of PAH. We provide experimental evidence supporting the ability of Gal-3 to influence reactive oxygen species production, NADPH oxidase enzyme expression, and redox signaling, which have been shown to contribute to both vascular remodeling and increased pulmonary arterial pressure. Future Directions: While several preclinical studies suggest that Gal-3 promotes hypertensive pulmonary vascular remodeling, the clinical significance of Gal-3 in human PAH remains to be established. Antioxid. Redox Signal. 00, 000-000.


Asunto(s)
Fibrosis/metabolismo , Galectina 3/metabolismo , Inflamación/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos
7.
Arterioscler Thromb Vasc Biol ; 37(6): 1180-1193, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28473444

RESUMEN

OBJECTIVE: A disintegrin and metalloproteinase ADAM17 (tumor necrosis factor-α [TNF]-converting enzyme) regulates soluble TNF levels. We tested the hypothesis that aging-induced activation in adipose tissue (AT)-expressed ADAM17 contributes to the development of remote coronary microvascular dysfunction in obesity. APPROACH AND RESULTS: Coronary arterioles (CAs, ≈90 µm) from right atrial appendages and mediastinal AT were examined in patients (aged: 69±11 years, BMI: 30.2±5.6 kg/m2) who underwent open heart surgery. CA and AT were also studied in 6-month and 24-month lean and obese mice fed a normal or high-fat diet. We found that obesity elicited impaired endothelium-dependent CA dilations only in older patients and in aged high-fat diet mice. Transplantation of AT from aged obese, but not from young or aged, mice increased serum cytokine levels, including TNF, and impaired CA dilation in the young recipient mice. In patients and mice, obesity was accompanied by age-related activation of ADAM17, which was attributed to vascular endothelium-expressed ADAM17. Excess, ADAM17-shed TNF from AT arteries in older obese patients was sufficient to impair CA dilation in a bioassay in which the AT artery was serially connected to a CA. Moreover, we found that the increased activity of endothelial ADAM17 is mediated by a diminished inhibitory interaction with caveolin-1, owing to age-related decline in caveolin-1 expression in obese patients and mice or to genetic deletion of caveolin-1. CONCLUSIONS: The present study indicates that aging and obesity cooperatively reduce caveolin-1 expression and increase vascular endothelial ADAM17 activity and soluble TNF release in AT, which may contribute to the development of remote coronary microvascular dysfunction in older obese patients.


Asunto(s)
Proteína ADAM17/metabolismo , Tejido Adiposo/enzimología , Envejecimiento/metabolismo , Arteriolas/enzimología , Enfermedad de la Arteria Coronaria/enzimología , Vasos Coronarios/enzimología , Vasodilatación , Proteína ADAM17/genética , Tejido Adiposo/trasplante , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/genética , Animales , Arteriolas/fisiopatología , Caveolina 1/deficiencia , Caveolina 1/genética , Caveolina 1/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Vasos Coronarios/fisiopatología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Obesidad/enzimología , Obesidad/genética , Obesidad/fisiopatología , Interferencia de ARN , Factores de Riesgo , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Vasc Res ; 53(5-6): 269-278, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27923220

RESUMEN

The circadian clock is rhythmically expressed in blood vessels, but the interaction between the circadian clock and disturbed blood flow remains unclear. We examined the relationships between BMAL1 and CLOCK and 2 regulators of endothelial function, AKT1 and endothelial nitric oxide synthase (eNOS), in vascular regions of altered blood flow. We found that the aortic arch from WT mice exhibited reduced sensitivity to acetylcholine (Ach)-mediated relaxation relative to the thoracic aorta. In Clock-mutant (mut) mice the aorta exhibited a reduced sensitivity to Ach. In WT mice, the phosphorylated forms of eNOS and AKT were decreased in the aortic arch, while BMAL1 and CLOCK expression followed a similar pattern of reduction in the arch. In conditions of surgically induced flow reduction, phosphorylated-eNOS (serine 1177) increased, as did p-AKT in the ipsilateral left common carotid artery (LC) of WT mice. Similarly, BMAL1 and CLOCK exhibited increased expression after 5 days in the remodeled LC. eNOS expression was increased at 8 p.m. versus 8 a.m. in WT mice, and this pattern was abolished in mut and Bmal1-KO mice. These data suggest that the circadian clock may be a biomechanical and temporal sensor that acts to coordinate timing, flow dynamics, and endothelial function.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Aorta Torácica/metabolismo , Proteínas CLOCK/metabolismo , Enfermedades de las Arterias Carótidas/metabolismo , Arteria Carótida Externa/metabolismo , Ritmo Circadiano , Mecanotransducción Celular , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Animales , Aorta Torácica/efectos de los fármacos , Proteínas CLOCK/genética , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/fisiopatología , Arteria Carótida Externa/fisiopatología , Arteria Carótida Externa/cirugía , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Genotipo , Ligadura , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Vasodilatación , Vasodilatadores/farmacología
9.
PLoS One ; 11(5): e0155075, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27168152

RESUMEN

The circadian clock is a transcriptional network that functions to regulate the expression of genes important in the anticipation of changes in cellular and organ function. Recent studies have revealed that the recognition of pathogens and subsequent initiation of inflammatory responses are strongly regulated by a macrophage-intrinsic circadian clock. We hypothesized that the circadian pattern of gene expression might be influenced by inflammatory stimuli and that loss of circadian function in immune cells can promote pro-inflammatory behavior. To investigate circadian rhythms in inflammatory cells, peritoneal macrophages were isolated from mPer2luciferase transgenic mice and circadian oscillations were studied in response to stimuli. Using Cosinor analysis, we found that LPS significantly altered the circadian period in peritoneal macrophages from mPer2luciferase mice while qPCR data suggested that the pattern of expression of the core circadian gene (Bmal1) was disrupted. Inhibition of TLR4 offered protection from the LPS-induced impairment in rhythm, suggesting a role for toll-like receptor signaling. To explore the mechanisms involved, we inhibited LPS-stimulated NO and superoxide. Inhibition of NO synthesis with L-NAME had no effect on circadian rhythms. In contrast, inhibition of superoxide with Tempol or PEG-SOD ameliorated the LPS-induced changes in circadian periodicity. In gain of function experiments, we found that overexpression of NOX5, a source of ROS, could significantly disrupt circadian function in a circadian reporter cell line (U2OS) whereas iNOS overexpression, a source of NO, was ineffective. To assess whether alteration of circadian rhythms influences macrophage function, peritoneal macrophages were isolated from Bmal1-KO and Per-TKO mice. Compared to WT macrophages, macrophages from circadian knockout mice exhibited altered balance between NO and ROS release, increased uptake of oxLDL and increased adhesion and migration. These results suggest that pro-inflammatory stimuli can disrupt circadian rhythms in macrophages and that impaired circadian rhythms may contribute to cardiovascular diseases by altering macrophage behavior.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Endotoxinas/toxicidad , Macrófagos Peritoneales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ritmo Circadiano/genética , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/farmacología , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones Noqueados , Óxido Nítrico/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/metabolismo , Factores de Transcripción/metabolismo
10.
Hypertension ; 67(3): 661-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26781276

RESUMEN

Blood pressure exhibits a robust circadian rhythm in health. In hypertension, sleep apnea, and even shift work, this balanced rhythm is perturbed via elevations in night-time blood pressure, inflicting silent damage to the vasculature and body organs. Herein, we examined the influence of circadian dysfunction during experimental hypertension in mice. Using radiotelemetry to measure ambulatory blood pressure and activity, the effects of angiotensin II administration were studied in wild-type (WT) and period isoform knockout (KO) mice (Per2-KO, Per2, 3-KO, and Per1, 2, 3-KO/Per triple KO [TKO] mice). On a normal diet, administration of angiotensin II caused nondipping blood pressure and exacerbated vascular hypertrophy in the Period isoform KO mice relative to WT mice. To study the endogenous effects of angiotensin II stimulation, we then administered a low-salt diet to the mice, which does stimulate endogenous angiotensin II in addition to lowering blood pressure. A low-salt diet decreased blood pressure in wild-type mice. In contrast, Period isoform KO mice lost their circadian rhythm in blood pressure on a low-salt diet, because of an increase in resting blood pressure, which was restorable to rhythmicity by the angiotensin receptor blocker losartan. Chronic administration of low salt caused vascular hypertrophy in Period isoform KO mice, which also exhibited increased renin levels and altered angiotensin 1 receptor expression. These data suggest that circadian clock genes may act to inhibit or control renin/angiotensin signaling. Moreover, circadian disorders such as sleep apnea and shift work may alter the homeostatic responses to sodium restriction to potentially influence nocturnal hypertension.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/fisiología , Ritmo Circadiano/fisiología , Dieta Hiposódica , Hipertensión/fisiopatología , Cloruro de Sodio Dietético/farmacología , Animales , Modelos Animales de Enfermedad , Hipertensión/dietoterapia , Masculino , Ratones , Ratones Noqueados
11.
PLoS One ; 8(10): e78626, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24205282

RESUMEN

Recent studies have shown that circadian clock disruption is associated with pathological remodeling in the arterial structure and vascular stiffness. Moreover, chronic circadian disruption is associated with dysfunction in endothelial responses and signaling. Reactive oxygen species have emerged as key regulators in vascular pathology. Previously, we have demonstrated that circadian clock dysfunction exacerbates superoxide production through eNOS uncoupling. To date, the impact of circadian clock mutation on vascular NADPH oxidase expression and function is not known. The goal in the current study was to determine if the circadian clock controls vascular Nox4 expression and hydrogen peroxide formation in arteries, particularly in endothelial and vascular smooth muscle cells. In aorta, there was an increase in hydrogen peroxide and Nox4 expression in mice with a dysfunctional circadian rhythm (Bmal1-KO mice). In addition, the Nox4 gene promoter is activated by the core circadian transcription factors. Lastly, in synchronized cultured human endothelial cells, Nox4 gene expression exhibited rhythmic oscillations. These data reveal that the circadian clock plays an important role in the control of Nox4 and disruption of the clock leads to subsequent production of reaction oxygen species.


Asunto(s)
Aorta/metabolismo , Relojes Circadianos , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Animales , Aorta/fisiología , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Regiones Promotoras Genéticas/genética
12.
Circ Res ; 111(9): 1157-65, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22912383

RESUMEN

RATIONALE: Disruption of the circadian clock in mice produces vascular dysfunction as evidenced by impairments in endothelium-dependent signaling, vasomotion, and blood vessel remodeling. Although the altered function of endothelial NO synthase and the overproduction of reactive oxygen species are central to dysfunction of the endothelium, to date, the impact of the circadian clock on endothelial NO synthase coupling and vascular reactive oxygen species production is not known. OBJECTIVE: The goals of the present study were to determine whether deletion of a critical component of the circadian clock, Bmal1, can influence endothelial NO synthase coupling and reactive oxygen species levels in arteries from Bmal1-knockout (KO) mice. METHODS AND RESULTS: Endothelial function was reduced in aortae from Bmal1-KO mice and improved by scavenging reactive oxygen species with polyethylene glycol-superoxide dismutase and nonselectively inhibiting cyclooxygenase isoforms with indomethacin. Aortae from Bmal1-KO mice exhibited enhanced superoxide levels as determined by electron paramagnetic resonance spectroscopy and dihydroethidium fluorescence, an elevation that was abrogated by administration of nitro-l-arginine methyl ester. High-performance liquid chromatography analysis revealed a reduction in tetrahydrobiopterin and an increase in dihydrobiopterin levels in the lung and aorta of Bmal1-KO mice, whereas supplementation with tetrahydrobiopterin improved endothelial function in the circadian clock KO mice. Furthermore, levels of tetrahydrobiopterin, dihydrobiopterin, and the key enzymes that regulate biopterin bioavailability, GTP cyclohydrolase and dihydrofolate reductase exhibited a circadian expression pattern. CONCLUSIONS: Having an established influence in the metabolic control of glucose and lipids, herein, we describe a novel role for the circadian clock in metabolism of biopterins, with a significant impact in the vasculature, to regulate coupling of endothelial NO synthase, production of superoxide, and maintenance of endothelial function.


Asunto(s)
Factores de Transcripción ARNTL/deficiencia , Aorta/metabolismo , Arterias/metabolismo , Relojes Circadianos/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Superóxidos/metabolismo , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Aorta/citología , Arterias/citología , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , GTP Ciclohidrolasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Especies Reactivas de Oxígeno/metabolismo , Tetrahidrofolato Deshidrogenasa/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 302(10): H1919-28, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22427510

RESUMEN

The expression and functional significance of NADPH oxidase 5 (Nox5) and its five isoforms in vascular cells is poorly understood. The goal of this study was to determine whether Nox5-α, -ß, -δ, -γ, and -ε (short) are expressed in human blood vessels and evaluate their respective functions. Nox5 mRNA and protein were detected in human blood vessels, cultured human vascular smooth muscle (HVSMC) and endothelium, but not fibroblasts. The most abundant isoforms were α and ß, whereas δ and γ were not detected. Nox5-α and -ß produced reactive oxygen species (ROS), but -δ, -γ, and -ε were not catalytically active. Coexpression of the active Nox5 isoforms with inactive Nox5 variants suppressed ROS production, and coimmunoprecipitation revealed that Nox5-ß binds the inactive ε variant, which may account for reduced ROS production. In HVSMC, angiotensin II, endothelin-1 and TNF-α increased endogenous Nox5 mRNA levels, while adenovirus-mediated overexpression of Nox5 promoted p38 MAPK, JAK2, JNK, and ERK1/2 phosphorylation in endothelial cells (EC), but only increased ERK1/2 phosphorylation in HVSMC. At higher levels of Nox5, there was evidence of increased apoptosis in EC, but not in HVSMC, as detected by the presence of cleaved caspase-3 and cleaved poly(ADP-ribose)polymerase. Although catalytically inactive, Nox5-ε potently activated ERK in HVSMC, and increased expression of Nox5-ε promoted HVSMC proliferation. Nox5 is expressed in human blood vessels. The Nox5-α and -ß splice variants are the major isoforms that are expressed and the only variants capable of ROS production. Nox5-ε can inhibit Nox5 activity and activate ERK and HVSMC proliferation.


Asunto(s)
Vasos Sanguíneos/metabolismo , Proteínas de la Membrana/metabolismo , NADPH Oxidasas/metabolismo , Transducción de Señal/fisiología , Vasos Sanguíneos/citología , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Isoenzimas/metabolismo , Proteínas de la Membrana/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , NADPH Oxidasa 5 , NADPH Oxidasas/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo
14.
PLoS One ; 7(2): e31495, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355372

RESUMEN

Chronic alterations in blood flow initiate structural changes in vessel lumen caliber to normalize shear stress. The loss of endothelial derived nitric oxide synthase (eNOS) in mice promotes abnormal flow dependent vascular remodeling, thus uncoupling mechanotransduction from adaptive vascular remodeling. However, the mechanisms of how the loss of eNOS promotes abnormal remodeling are not known. Here we show that abnormal flow-dependent remodeling in eNOS knockout mice (eNOS (-/-)) is associated with activation of the platelet derived growth factor (PDGF) signaling pathway leading to the induction of the inhibitor of apoptosis, survivin. Interfering with PDGF signaling or survivin function corrects the abnormal remodeling seen in eNOS (-/-) mice. Moreover, nitric oxide (NO) negatively regulates PDGF driven survivin expression and cellular proliferation in cultured vascular smooth muscle cells. Collectively, our data suggests that eNOS negatively regulates the PDGF-survivin axis to maintain proportional flow-dependent luminal remodeling and vascular quiescence.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Endotelio Vascular/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Músculo Liso Vascular/metabolismo , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Represoras/metabolismo , Animales , Apoptosis , Western Blotting , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Técnicas para Inmunoenzimas , Proteínas Inhibidoras de la Apoptosis/genética , Masculino , Mecanotransducción Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Óxido Nítrico/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Unión Proteica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Transducción de Señal , Survivin
15.
Proc Natl Acad Sci U S A ; 108(41): 17147-52, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21969583

RESUMEN

The suprachiasmatic nucleus of the brain is the circadian center, relaying rhythmic environmental and behavioral information to peripheral tissues to control circadian physiology. As such, central clock dysfunction can alter systemic homeostasis to consequently impair peripheral physiology in a manner that is secondary to circadian malfunction. To determine the impact of circadian clock function in organ transplantation and dissect the influence of intrinsic tissue clocks versus extrinsic clocks, we implemented a blood vessel grafting approach to surgically assemble a chimeric mouse that was part wild-type (WT) and part circadian clock mutant. Arterial isografts from donor WT mice that had been anastamosed to common carotid arteries of recipient WT mice (WT:WT) exhibited no pathology in this syngeneic transplant strategy. Similarly, when WT grafts were anastamosed to mice with disrupted circadian clocks, the structural features of the WT grafts immersed in the milieu of circadian malfunction were normal and absent of lesions, comparable to WT:WT grafts. In contrast, aortic grafts from Bmal1 knockout (KO) or Period-2,3 double-KO mice transplanted into littermate control WT mice developed robust arteriosclerotic disease. These lesions observed in donor grafts of Bmal1-KO were associated with up-regulation in T-cell receptors, macrophages, and infiltrating cells in the vascular grafts, but were independent of hemodynamics and B and T cell-mediated immunity. These data demonstrate the significance of intrinsic tissue clocks as an autonomous influence in experimental models of arteriosclerotic disease, which may have implications with regard to the influence of circadian clock function in organ transplantation.


Asunto(s)
Aorta/trasplante , Arteriosclerosis/etiología , Relojes Circadianos/fisiología , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/fisiología , Animales , Aorta/patología , Aorta/fisiopatología , Arteriosclerosis/genética , Arteriosclerosis/patología , Arteriosclerosis/fisiopatología , Arteria Carótida Común/patología , Arteria Carótida Común/fisiopatología , Arteria Carótida Común/cirugía , Relojes Circadianos/genética , Macrófagos/fisiología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Especificidad de Órganos , Proteínas Circadianas Period/deficiencia , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Núcleo Supraquiasmático/fisiopatología , Trasplante Isogénico , Regulación hacia Arriba
16.
Arterioscler Thromb Vasc Biol ; 31(7): 1634-42, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21527745

RESUMEN

OBJECTIVE: Increased protein SUMOylation (small ubiquitin-related modifier [SUMO]) provides protection from cellular stress, including oxidative stress, but the mechanisms involved are incompletely understood. The NADPH oxidases (Nox) are a primary source of reactive oxygen species (ROS) and oxidative stress, and thus our goal was to determine whether SUMO regulates NADPH oxidase activity. METHODS AND RESULTS: Increased expression of SUMO1 potently inhibited the activity of Nox1 to Nox5. In contrast, inhibition of endogenous SUMOylation with small interfering RNA to SUMO1 or ubiquitin conjugating enzyme 9 or with the inhibitor anacardic acid increased ROS production from human embryonic kidney-Nox5 cells, human vascular smooth muscle cells, and neutrophils. The suppression of ROS production was unique to SUMO1, and it required a C-terminal diglycine and the SUMO-specific conjugating enzyme ubiquitin conjugating enzyme 9. SUMO1 did not modify intracellular calcium or Nox5 phosphorylation but reduced ROS output in an isolated enzyme assay, suggesting direct effects of SUMOylation on enzyme activity. However, we could not detect the presence of SUMO1 conjugation on Nox5 using a variety of approaches. Moreover, the mutation of more than 17 predicted and conserved lysine residues on Nox5 did not alter the inhibitory actions of SUMO1. CONCLUSIONS: Together, these results suggest that SUMO is an important regulatory mechanism that indirectly represses the production of ROS to ameliorate cellular stress.


Asunto(s)
NADPH Oxidasas/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Proteína SUMO-1/metabolismo , Ácidos Anacárdicos/farmacología , Análisis de Varianza , Animales , Células COS , Calcio/metabolismo , Chlorocebus aethiops , Regulación hacia Abajo , Células HEK293 , Humanos , Isoenzimas , Lisina , Músculo Liso Vascular/enzimología , Mutación , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/genética , Neutrófilos/enzimología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica , Interferencia de ARN , Proteína SUMO-1/genética , Sumoilación , Factores de Tiempo , Transfección , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 30(12): 2535-43, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20829506

RESUMEN

OBJECTIVE: To determine if elasticity in blood vessels is compromised in circadian clock-mutant mice (Bmal1-knockout [KO] and Per-triple KO) and if matrix metalloproteinases (MMPs) might confer these changes in compliance. METHODS AND RESULTS: High-resolution ultrasonography in vivo revealed impaired remodeling and increased pulse-wave velocity in the arteries of Bmal1-KO and Per-triple KO mice. In addition, compliance of remodeled arteries and naïve pressurized arterioles ex vivo from Bmal1-KO and Per-triple KO mice was reduced, consistent with stiffening of the vascular bed. The observed vascular stiffness was coincident with dysregulation of MMP-2 and MMP-9 in Bmal1-KO mice. Furthermore, inhibition of MMPs improved indexes of pathological remodeling in wild-type mice, but the effect was abolished in Bmal1-KO mice. CONCLUSIONS: Circadian clock dysfunction contributes to hardening of arteries, which may involve impaired control of the extracellular matrix composition.


Asunto(s)
Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/enzimología , Relojes Circadianos/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Animales , Presión Sanguínea , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiopatología , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/fisiopatología , Células Cultivadas , Elasticidad , Células Endoteliales/enzimología , Matriz Extracelular/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Miocitos del Músculo Liso/enzimología , Proteínas Circadianas Period/deficiencia , Proteínas Circadianas Period/genética , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología , Factores de Tiempo , Ultrasonografía Doppler de Pulso
19.
J Appl Physiol (1985) ; 107(4): 1328-38, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19679741

RESUMEN

Hypertension is a major risk factor for cardiovascular disease and death. The "silent" rise of blood pressure that occurs over time is largely asymptomatic. However, its impact is deafening-causing and exacerbating cardiovascular disease, end-organ damage, and death. The present article addresses recent observations from human and animal studies that provide new insights into how the circadian clock regulates blood pressure, contributes to hypertension, and ultimately evolves vascular disease. Further, the molecular components of the circadian clock and their relationship with locomotor activity, metabolic control, fluid balance, and vascular resistance are discussed with an emphasis on how these novel, circadian clock-controlled mechanisms contribute to hypertension.


Asunto(s)
Relojes Biológicos , Presión Sanguínea , Enfermedades Cardiovasculares/fisiopatología , Ritmo Circadiano , Hipertensión/fisiopatología , Animales , Antihipertensivos/administración & dosificación , Relojes Biológicos/genética , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Ritmo Circadiano/genética , Modelos Animales de Enfermedad , Cronoterapia de Medicamentos , Metabolismo Energético , Regulación de la Expresión Génica , Homeostasis , Humanos , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Hipertensión/metabolismo , Actividad Motora , Resultado del Tratamiento , Resistencia Vascular , Equilibrio Hidroelectrolítico
20.
Am J Pathol ; 174(6): 2086-95, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19435785

RESUMEN

Inhibition of soluble epoxide hydrolase (SEH), the enzyme responsible for degradation of vasoactive epoxides, protects against cerebral ischemia in rats. However, the molecular and biological mechanisms that confer protection in normotension and hypertension remain unclear. Here we show that 6 weeks of SEH inhibition via 2 mg/day of 12-(3-adamantan-1-yl-ureido) dodecanoic acid (AUDA) in spontaneously hypertensive stroke-prone (SHRSP) rats protects against cerebral ischemia induced by middle cerebral artery occlusion, reducing percent hemispheric infarct and neurodeficit score without decreasing blood pressure. This level of cerebral protection was similar to that of the angiotensin-converting enzyme inhibitor, enalapril, which significantly lowered blood pressure. SEH inhibition is also protective in normotensive Wistar-Kyoto (WKY) rats, reducing both hemispheric infarct and neurodeficit score. In SHRSP rats, SEH inhibition reduced wall-to-lumen ratio and collagen deposition and increased cerebral microvessel density, although AUDA did not alter middle cerebral artery structure or microvessel density in WKY rats. An apoptosis mRNA expression microarray of brain tissues from AUDA-treated rats revealed that AUDA modulates gene expression of mediators involved in the regulation of apoptosis in neural tissues of both WKY and SHRSP rats. Hence, we conclude that chronic SEH inhibition protects against cerebral ischemia via vascular protection in SHRSP rats and neural protection in both the SHRSP and WKY rats, indicating that SEH inhibition has broad pharmacological potential for treating ischemic stroke.


Asunto(s)
Adamantano/análogos & derivados , Isquemia Encefálica/enzimología , Epóxido Hidrolasas/metabolismo , Expresión Génica/efectos de los fármacos , Ácidos Láuricos/farmacología , Fármacos Neuroprotectores/farmacología , Adamantano/farmacología , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/patología , Circulación Cerebrovascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Masculino , Microvasos/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...