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

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Exerc Sci Fit ; 19(2): 75-80, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33335553

RESUMEN

BACKGROUND/OBJECTIVE: Measures of oxygen uptake efficiency (OUE) have been used to evaluate cardiorespiratory fitness (CRF) in adolescents unable to perform maximal exercise. The oxygen uptake efficiency slope (OUES) and oxygen uptake efficiency plateau (OUEP) have been proposed as surrogates for maximal oxygen consumption (V̇O2max). We assessed the validity of the OUES and OUEP as predictors of V̇O2max in healthy male adolescents. METHODS: Sixty-three healthy male adolescents aged 15.40 ± 0.34 years underwent an incremental treadmill test to determine V̇O2max, OUES and OUEP. OUE throughout the test was assessed by dividing each V̇O2 value by the corresponding minute ventilation (V̇E) value. OUEP was determined as the 90 s average highest consecutive values for OUE. OUES was determined using data up to the ventilatory threshold (VT) by calculating the slope of the linear relation between V̇O2 and the logarithm of V̇E. RESULTS: Limits of agreement for V̇O2max predicted by OUES (±13.3 mL kg-1.min-1) and OUEP (±16.7 mL kg-1.min-1) relative to V̇O2max were wide and a magnitude bias was found for OUES and OUEP as predictors of V̇O2max (p < 0.001). CONCLUSION: The OUES and OUEP do not accurately predict V̇O2max in male adolescents and should not replace V̇O2max when assessing CRF in this population.

2.
Eur J Appl Physiol ; 119(3): 743-752, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30673849

RESUMEN

INTRODUCTION: Although commonly understood as immune cells, certain T lymphocyte and monocyte subsets have angiogenic potential, contributing to blood vessel growth and repair. These cells are highly exercise responsive and may contribute to the cardiovascular benefits seen with exercise. PURPOSE: To compare the effects of a single bout of continuous (CONTEX) and sprint interval exercise (SPRINT) on circulating angiogenic cells (CAC) in healthy recreationally active adults. METHODS: Twelve participants (aged 29 ± 2 years, BMI 25.5 ± 0.9 kg m- 2, [Formula: see text]peak 44.3 ± 1.8 ml kg- 1 min- 1; mean ± SEM) participated in the study. Participants completed a 45-min bout of CONTEX at 70% peak oxygen uptake and 6 × 20 s sprints on a cycle ergometer, in a counterbalanced design. Blood was sampled pre-, post-, 2 h and 24 h post-exercise for quantification of CAC subsets by whole blood flow cytometric analysis. Angiogenic T lymphocytes (TANG) and angiogenic Tie2-expressing monocytes (TEM) were identified by the expression of CD31 and Tie2, respectively. RESULTS: Circulating (cells µL- 1) CD3+CD31+ TANG increased immediately post-exercise in both trials (p < 0.05), with a significantly greater increase (p < 0.05) following SPRINT (+ 57%) compared to CONTEX (+ 14%). Exercise increased (p < 0.05) the expression of the chemokine receptor CXCR4 on TANG at 24 h. Tie2-expressing classical (CD14++CD16-), intermediate (CD14++CD16+) and non-classical (CD14+CD16++) monocytes and circulating CD34+CD45dim progenitor cells were higher post-exercise in SPRINT, but unchanged in CONTEX. All post-exercise increases in SPRINT were back to pre-exercise levels at 2 h and 24 h. CONCLUSION: Acute exercise transiently increases circulating TANG, TEM and progenitor cells with greater increases evident following very high intensity sprint exercise than following prolonged continuous paced endurance exercise.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares , Ejercicio Físico/fisiología , Monocitos/citología , Consumo de Oxígeno/fisiología , Adulto , Terapia por Ejercicio/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/fisiología , Receptores CXCR4/metabolismo
4.
BMC Genomics ; 16: 1036, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26645212

RESUMEN

BACKGROUND: 3'untranslated regions (3'UTRs) are poorly understood portions of eukaryotic mRNAs essential for post-transcriptional gene regulation. Sequence elements in 3'UTRs can be target sites for regulatory molecules such as RNA binding proteins and microRNAs (miRNAs), and these interactions can exert significant control on gene networks. However, many such interactions remain uncharacterized due to a lack of high-throughput (HT) tools to study 3'UTR biology. HT cloning efforts such as the human ORFeome exemplify the potential benefits of genomic repositories for studying human disease, especially in relation to the discovery of biomarkers and targets for therapeutic agents. Currently there are no publicly available human 3'UTR libraries. To address this we have prepared the first version of the human 3'UTRome (h3'UTRome v1) library. The h3'UTRome is produced to a single high quality standard using the same recombinational cloning technology used for the human ORFeome, enabling universal operating methods and high throughput experimentation. The library is thoroughly sequenced and annotated with simple online access to information, and made publically available through gene repositories at low cost to all scientists with minimal restriction. RESULTS: The first release of the h3'UTRome library comprises 1,461 human 3'UTRs cloned into Gateway® entry vectors, ready for downstream analyses. It contains 3'UTRs for 985 transcription factors, 156 kinases, 171 RNA binding proteins, and 186 other genes involved in gene regulation and in disease. We demonstrate the feasibility of the h3'UTRome library by screening a panel of 87 3'UTRs for targeting by two miRNAs: let-7c, which is implicated in tumorigenesis, and miR-221, which is implicated in atherosclerosis and heart disease. The panel is enriched with genes involved in the RAS signaling pathway, putative novel targets for the two miRNAs, as well as genes implicated in tumorigenesis and heart disease. CONCLUSIONS: The h3'UTRome v1 library is a modular resource that can be utilized for high-throughput screens to identify regulatory interactions between trans-acting factors and 3'UTRs, Importantly, the library can be customized based on the specifications of the researcher, allowing the systematic study of human 3'UTR biology.


Asunto(s)
Regiones no Traducidas 3' , Procesamiento Postranscripcional del ARN , Perfilación de la Expresión Génica , Biblioteca de Genes , Humanos , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Transcriptoma
5.
Blood ; 121(1): 188-96, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23160460

RESUMEN

Wnt signaling is involved in numerous aspects of vertebrate development and homeostasis, including the formation and function of blood cells. Here, we show that canonical and noncanonical Wnt signaling pathways are present and functional in megakaryocytes (MKs), with several Wnt effectors displaying MK-restricted expression. Using the CHRF288-11 cell line as a model for human MKs, the canonical Wnt3a signal was found to induce a time and dose-dependent increase in ß-catenin expression. ß-catenin accumulation was inhibited by the canonical antagonist dickkopf-1 (DKK1) and by the noncanonical agonist Wnt5a. Whole genome expression analysis demonstrated that Wnt3a and Wnt5a regulated distinct patterns of gene expression in MKs, and revealed a further interplay between canonical and noncanonical Wnt pathways. Fetal liver cells derived from low-density-lipoprotein receptor-related protein 6-deficient mice (LRP6(-/-)), generated dramatically reduced numbers of MKs in culture of lower ploidy (2N and 4N) than wild-type controls, implicating LRP6-dependent Wnt signaling in MK proliferation and maturation. Finally, in wild-type mature murine fetal liver-derived MKs, Wnt3a potently induced proplatelet formation, an effect that could be completely abrogated by DKK1. These data identify novel extrinsic regulators of proplatelet formation, and reveal a profound role for Wnt signaling in platelet production.


Asunto(s)
Megacariocitos/citología , Trombopoyesis/fisiología , Vía de Señalización Wnt/fisiología , Animales , Plaquetas/citología , Línea Celular , Células Cultivadas/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Hígado/embriología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Proteínas Recombinantes/farmacología , Trombopoyesis/genética , Proteínas Wnt/farmacología , Proteína Wnt3A/farmacología , beta Catenina/biosíntesis , beta Catenina/genética
7.
Am J Physiol Heart Circ Physiol ; 304(12): H1585-97, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23604713

RESUMEN

Thrombomodulin (TM) is a 557-amino acid protein with a broad cell and tissue distribution consistent with its wide-ranging physiological roles. When expressed on the lumenal surface of vascular endothelial cells in both large vessels and capillaries, its primary function is to mediate endothelial thromboresistance. The complete integral membrane-bound protein form displays five distinct functional domains, although shorter soluble (functional) variants comprising the extracellular domains have also been reported in fluids such as serum and urine. TM-mediated binding of thrombin is known to enhance the specificity of the latter serine protease toward both protein C and thrombin activatable fibrinolysis inhibitor (TAFI), increasing their proteolytic activation rate by almost three orders of magnitude with concomitant anticoagulant, antifibrinolytic, and anti-inflammatory benefits to the vascular wall. Recent years have seen an abundance of research into the cellular mechanisms governing endothelial TM production, processing, and regulation (including flow-mediated mechanoregulation)--from transcriptional and posttranscriptional (miRNA) regulation of TM gene expression, to posttranslational processing and release of the expressed protein--facilitating greater exploitation of its therapeutic potential. The goal of the present paper is to comprehensively review the endothelial/TM system from these regulatory perspectives and draw some fresh conclusions. This paper will conclude with a timely examination of the current status of TM's growing therapeutic appeal, from novel strategies to improve the clinical efficacy of recombinant TM analogs for resolution of vascular disorders such as disseminated intravascular coagulation (DIC), to an examination of the complex pleiotropic relationship between statin treatment and TM expression.


Asunto(s)
Endotelio Vascular/metabolismo , Trombomodulina/metabolismo , Animales , Expresión Génica , Regulación de la Expresión Génica , Humanos , Trombomodulina/química , Trombomodulina/genética , Enfermedades Vasculares/terapia
8.
J Cell Physiol ; 226(11): 3053-63, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21302304

RESUMEN

Blood-brain barrier (BBB) regulation involves the coordinated interaction of intercellular adherens and tight junctions in response to stimuli. One such stimulus, shear stress, has been shown to upregulate brain microvascular endothelial cell (BMvEC) barrier function, although our knowledge of the signaling mechanisms involved is limited. In this article, we examined the hypothesis that VE-cadherin can transmit shear signals to tight junction occludin with consequences for pTyr-occludin and barrier function. In initial studies, chronic shear enhanced membrane localization of ZO-1 and claudin-5, decreased pTyr-occludin (in part via a dephostatin-sensitive mechanism), and reduced BMvEC permeability, with flow reduction in pre-sheared BMvECs having converse effects. In further studies, VE-cadherin inhibition (VE-cad ΔEXD) blocked shear-induced Rac1 activation, pTyr-occludin reduction, and barrier upregulation, consistent with an upstream role for VE-cadherin in transmitting shear signals to tight junctions through Rac1. As VE-cadherin is known to mediate Rac1 activation via Tiam1 recruitment, we subsequently confirmed that Tiam1 inhibition (Tiam1-C580) could elicit effects similar to VE-cad ΔEXD. Finally, the observed attenuation of shear-induced changes in pTyr-occludin level and barrier phenotype following Rac1 inhibition (NSC23766, T17N) establishes a downstream role for Rac1 in this pathway. In summary, we describe for the first time in BMvECs a role for VE-cadherin in the transmission of physiological shear signals to tight junction occludin through engagement of Tiam1/Rac1 leading to barrier stabilization. A downstream role is also strongly indicated for a protein tyrosine phosphatase in pTyr-occludin modulation. Importantly, these findings suggest an important route of inter-junctional signaling cross-talk during BBB response to flow.


Asunto(s)
Antígenos CD/metabolismo , Barrera Hematoencefálica/fisiología , Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Proteínas de la Membrana/metabolismo , Resistencia al Corte , Estrés Mecánico , Animales , Permeabilidad Capilar/fisiología , Bovinos , Claudina-5 , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Microvasos/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Uniones Estrechas/fisiología , Proteína de la Zonula Occludens-1 , Proteína de Unión al GTP rac1/metabolismo
9.
Sci Rep ; 11(1): 21906, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753989

RESUMEN

Ground based research modalities of microgravity have been proposed as innovative methods to investigate the aetiology of chronic age-related conditions such as cardiovascular disease. Dry Immersion (DI), has been effectively used to interrogate the sequelae of physical inactivity (PI) and microgravity on multiple physiological systems. Herein we look at the causa et effectus of 3-day DI on platelet phenotype, and correlate with both miRomic and circulating biomarker expression. The miRomic profile of platelets is reflective of phenotype, which itself is sensitive and malleable to the exposome, undergoing responsive transitions in order to fulfil platelets role in thrombosis and haemostasis. Heterogeneous platelet subpopulations circulate at any given time, with varying degrees of sensitivity to activation. Employing a DI model, we investigate the effect of acute PI on platelet function in 12 healthy males. 3-day DI resulted in a significant increase in platelet count, plateletcrit, platelet adhesion, aggregation, and a modest elevation of platelet reactivity index (PRI). We identified 15 protein biomarkers and 22 miRNA whose expression levels were altered after DI. A 3-day DI model of microgravity/physical inactivity induced a prothrombotic platelet phenotype with an unique platelet miRNA signature, increased platelet count and plateletcrit. This correlated with a unique circulating protein biomarker signature. Taken together, these findings highlight platelets as sensitive adaptive sentinels and functional biomarkers of epigenetic drift within the cardiovascular compartment.


Asunto(s)
Plaquetas/citología , Proteínas Sanguíneas/metabolismo , MicroARNs/genética , Modelos Biológicos , Ingravidez , Adulto , Biomarcadores/sangre , Hemostasis , Humanos , Masculino , Trombosis/metabolismo
10.
Burns ; 46(7): 1585-1602, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32475797

RESUMEN

BACKGROUND: Burn injuries are a major cause of morbidity and mortality worldwide. Despite advances in therapeutic strategies for the management of patients with severe burns, the sequelae are pathophysiologically profound, up to the systemic and metabolic levels. Management of patients with a severe burn injury is a long-term, complex process, with treatment dependent on the degree and location of the burn and total body surface area (TBSA) affected. In adverse conditions with limited resources, efficient triage, stabilisation, and rapid transfer to a specialised intensive care burn centre is necessary to provide optimal outcomes. This initial lag time and the form of primary treatment initiated, from injury to specialist care, is crucial for the burn patient. This study aims to investigate the efficacy of a novel visco-elastic burn dressing with a proprietary bio-stimulatory marine mineral complex (MXC) as a primary care treatment to initiate a healthy healing process prior to specialist care. METHODS: A new versatile emergency burn dressing saturated in a >90% translucent water-based, sterile, oil-free gel and carrying a unique bio-stimulatory marine mineral complex (MXC) was developed. This dressing was tested using LabSkin as a burn model platform. LabSkin a novel cellular 3D-dermal organotypic full thickness human skin equivalent, incorporating fully-differentiated dermal and epidermal components that functionally models skin. Cell and molecular analysis was carried out by in vitro Real-Time Cellular Analysis (RTCA), thermal analysis, and focused transcriptomic array profiling for quantitative gene expression analysis, interrogating both wound healing and fibrosis/scarring molecular pathways. In vivo analysis was also performed to assess the bio-mechanical and physiological effects of this novel dressing on human skin. RESULTS: This hybrid emergency burn dressing (EBD) with MXC was hypoallergenic, and improved the barrier function of skin resulting in increased hydration up to 24 h. It was demonstrated to effectively initiate cooling upon application, limiting the continuous burn effect and preventing local tissue from damage and necrosis. xCELLigence RTCA® on primary human dermal cells (keratinocyte, fibroblast and micro-vascular endothelial) demonstrated improved cellular function with respect to tensegrity, migration, proliferation and cell-cell contact (barrier formation) [1]. Quantitative gene profiling supported the physiological and cellular function finding. A beneficial quid pro quo regulation of genes involved in wound healing and fibrosis formation was observed at 24 and 48 h time points. CONCLUSION: Utilisation of this EBD + MXC as a primary treatment is an effective and easily applicable treatment in cases of burn injury, proving both a cooling and hydrating environment for the wound. It regulates inflammation and promotes healing in preparation for specialised secondary burn wound management. Moreover, it promotes a healthy remodelling phenotype that may potentially mitigate scarring. Based on our findings, this EBD + MXC is ideal for use in all pre-hospital, pre-surgical and resource limited settings.


Asunto(s)
Vendas Hidrocoloidales , Quemaduras , Cicatriz , Productos Biológicos/uso terapéutico , Quemaduras/patología , Quemaduras/terapia , Cicatriz/patología , Humanos , Técnicas In Vitro , Piel/patología , Cicatrización de Heridas
11.
Eur J Appl Physiol ; 106(4): 555-62, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19347350

RESUMEN

Triglyceride-rich postprandial lipoproteins are known to activate endothelial cells in vitro, contributing to atherosclerosis. Endothelial microparticles (EMP) are membranous vesicles released into the circulation from vascular endothelial cells that permit cell activation to be monitored in vivo. The objective of the study was to examine changes in EMP following a high fat meal, consumed with and without prior exercise. Eight recreationally active young men underwent two oral fat tolerance tests following either 100 min exercise at 70% VO(2)peak (EX trial) or no exercise (CON trial) on the previous evening. Postprandial triglycerides were reduced (1.97 +/- 0.31 vs. 1.17 +/- 0.13 mmol L(-1), p < 0.05) and HDL-cholesterol (HDL-C) increased (1.20 +/- 0.07 vs. 1.30 +/- 0.08 mmol L(-1), p < 0.05) in the EX compared to CON trial. EMP (CD31+/42b-) increased postprandially (p < 0.05). However, counts were not different between trials (postprandial CON and EX trial counts x 10(3 )microL(-1), 3.10 +/- 0.14 vs. 3.26 +/- 0.37). There were no changes in sICAM-1 or sVCAM-1 postprandially and no differences between trials. Interleukin-6 (IL-6) and leukocytes increased postprandially (p < 0.05). IL-6 values were not different between trials. Leukocytes were higher at 0 h in the EX trial with CON and EX trial values similar at 6 h. EMP, but not sICAM-1 or sVCAM-1, increase in response to a high fat meal. However, EMP are not attenuated by acute exercise, despite a considerable reduction in postprandial lipemia and an increase in HDL-C.


Asunto(s)
Moléculas de Adhesión Celular/sangre , Micropartículas Derivadas de Células/metabolismo , HDL-Colesterol/sangre , Grasas de la Dieta/metabolismo , Endotelio Vascular/fisiología , Esfuerzo Físico/fisiología , Triglicéridos/sangre , Adulto , Endotelio Vascular/citología , Humanos , Masculino
12.
Brain Res ; 1193: 84-92, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18177846

RESUMEN

Basolateral condition of the brain microvascular endothelium is believed to influence blood-brain barrier (BBB) phenotype, although the precise transcriptional and post-translational mechanisms involved are poorly defined. In vivo, the basolateral surface of the blood-brain endothelium is bathed in serum-free interstitial fluid and encompassed by astrocytic end-feet. We hypothesized that these conditions impact on BBB function by directly modulating expression and biochemical properties of tight junctions. To investigate this, an in vitro transwell culture model was employed to selectively modify the basolateral environment of bovine brain microvascular endothelial cells (BBMvECs). In the absence of basolateral (but not apical) serum, we observed higher levels of expression, association and plasma membrane localization for the tight junction proteins, occludin and zonula occludens-1 (ZO-1), in parallel with elevated transendothelial electrical resistance (TEER) and reduced (14)[C]-sucrose permeability of BBMvEC monolayers. We further examined the effects of non-contact co-culture with basolateral astrocytes (C6 glioma) on indices of BBMvEC barrier function in both the presence and absence of serum. Astrocyte co-culture with serum led to enhanced occludin protein expression, occludin/ZO-1 association, and ZO-1 membrane localization, in parallel with increased TEER of BBMvEC monolayers. Astrocyte co-culture in the absence of serum (i.e. basolateral conditions most consistent with in vivo BBB physiology) however, gave the highest increases in BBMvEC barrier indices. Thus, we can conclude that factors influencing condition of the basolateral environment of the brain microvasculature can directly, and independently, modify BBB properties by regulating the expression and biochemical properties of the tight junction proteins, occludin and ZO-1.


Asunto(s)
Encéfalo/anatomía & histología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Uniones Estrechas/fisiología , Análisis de Varianza , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Permeabilidad Capilar/fisiología , Bovinos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Impedancia Eléctrica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ocludina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ratas , Suero/fisiología , Factores de Tiempo , Proteína de la Zonula Occludens-1
13.
Mech Ageing Dev ; 174: 76-85, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29155255

RESUMEN

The cardiovascular system is responsible for transport of blood and nutrients to tissues, and is pivotal to the physiological health and longevity. Epigenetic modification is a natural, age-associated process resulting in highly contextualised gene expression with clear implications for cell differentiation and disease onset. Biological/epigenetic age is independent of chronological age, constituting a highly reflective snapshot of an individual's overall health. Accelerated vascular ageing is of major concern, effectively lowering disease threshold. Age-related chronic illness involves a complex interplay between many biological processes and is modulated by non-modifiable and modifiable risk factors. These alter the static genome by a number of epigenetic mechanisms, which change gene expression in an age and lifestyle dependent manner. This 'epigenetic drift' impacts health and contributes to the etiology of chronic illness. Lifestyle factors may cause acceleration of this epigenetic "clock", pre-disposing individuals to cardiovascular disease. Nutrition and physical activity are modifiable lifestyle choices, synergistically contributing to cardiovascular health. They represent a powerful potential epigenetic intervention point for effective cardiovascular protective and management strategies. Thus, together with traditional risk factors, monitoring the epigenetic signature of ageing may prove beneficial for tailoring lifestyle to fit biology - supporting the increasingly popular concept of "ageing well".


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/prevención & control , Fenómenos Fisiológicos Nutricionales del Anciano , Epigénesis Genética , Ejercicio Físico , Animales , Enfermedades Cardiovasculares/metabolismo , Humanos
14.
Arterioscler Thromb Vasc Biol ; 26(1): 62-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16269664

RESUMEN

OBJECTIVE: The vascular endothelium constitutes a highly effective fluid/solute barrier through the regulated apposition of intercellular tight junction complexes. Because endothelium-mediated functions and pathology are driven by hemodynamic forces (cyclic strain and shear stress), we hypothesized a dynamic regulatory link between endothelial tight junction assembly/function and hemodynamic stimuli. We, therefore, examined the effects of cyclic strain on the expression, modification, and function of 2 pivotal endothelial tight junction components, occludin and ZO-1. METHODS AND RESULTS: For these studies, bovine aortic endothelial cells were subjected to physiological levels of equibiaxial cyclic strain (5% strain, 60 cycles/min, 24 hours). In response to strain, both occludin and ZO-1 protein expression increased by 2.3+/-0.1-fold and 2.0+/-0.3-fold, respectively, concomitant with a strain-dependent increase in occludin (but not ZO-1) mRNA levels. These changes were accompanied by reduced occludin tyrosine phosphorylation (75.7+/-8%) and increased ZO-1 serine/threonine phosphorylation (51.7+/-9% and 82.7+/-25%, respectively), modifications that could be completely blocked with tyrosine phosphatase and protein kinase C inhibitors (dephostatin and rottlerin, respectively). In addition, there was a significant strain-dependent increase in endothelial occludin/ZO-1 association (2.0+/-0.1-fold) in parallel with increased localization of both occludin and ZO-1 to the cell-cell border. These events could be completely blocked by dephostatin and rottlerin, and they correlated with a strain-dependent reduction in transendothelial permeability to FITC-dextran. CONCLUSIONS: Overall, these findings indicate that cyclic strain modulates both the expression and phosphorylation state of occludin and ZO-1 in vascular endothelial cells, with putative consequences for endothelial tight junction assembly and barrier integrity.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/fisiología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Uniones Estrechas/fisiología , Acetofenonas/farmacología , Animales , Aorta/citología , Benzopiranos/farmacología , Permeabilidad Capilar/fisiología , Bovinos , Dextranos/farmacocinética , Inhibidores Enzimáticos/farmacología , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Expresión Génica/fisiología , Hidroquinonas/farmacología , Técnicas In Vitro , Proteínas de la Membrana/genética , Ocludina , Fosfoproteínas/genética , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , ARN Mensajero/metabolismo , Estrés Mecánico , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1
15.
Data Brief ; 15: 600-605, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29085875

RESUMEN

The data presented herein are connected to our research article (doi: 10.1016/j.biocel.2017.04.012) [1], in which we investigated the functional connections between the urokinase receptor (uPAR), and the ezrin/radixin/moesin (ERM) proteins, moesin and merlin [1]. Firstly, a model of action is proposed that enlightens how uPAR regulates distal integrins. In addition, data show the effects of expressing wild-type moesin or permanently active T558D mutant of moesin on angiogenesis and morphology of human aortic endothelial cells (HAEC). Additional data compare the effects of urokinase (uPA, the main ligand of uPAR) on the same cells. Lastly, we provide technical data demonstrating the effects of specific siRNA for moesin and merlin on moesin and merlin expression, respectively.

16.
Int J Biochem Cell Biol ; 88: 14-22, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28473293

RESUMEN

The glycosyl-phosphatidyl-inositol (GPI)-anchored urokinase receptor (uPAR) has no intracellular domain, but nevertheless initiates signalling through proximal interactions with other membrane receptors including integrins. The relationships between uPAR and ezrin/radixin/moesin (ERM) proteins, moesin and merlin have never been explored. Moesin and merlin are versatile membrane-actin links and regulators of receptors signalling, respectively. We show that uPAR controls moesin and merlin, which propagate uPAR-initiated signals and modulate integrin functions, thereby regulating uPAR activity. uPAR rapidly de-phosphorylates moesin and phosphorylates merlin inactivating both proteins, and enhancing cell migration and angiogenesis. Moesin behaves as a molecular switch turning either on or off uPAR signalling through cycles of de-activation/activation, or sustained activation, respectively. Furthermore, moesin is at the crossroads of uPAR-initiated outside-in and inside-out signalling promoting integrin-dependent cell adhesion suggesting that uPAR also activates integrins distally through moesin. Knocking down merlin expression enhanced cell migration and adhesion through different regulation of fibronectin- and vitronectin-binding integrins.


Asunto(s)
Adhesión Celular , Quimiotaxis , Células Endoteliales/citología , Proteínas de Microfilamentos/metabolismo , Neovascularización Fisiológica , Neurofibromina 2/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Células Endoteliales/metabolismo , Humanos
17.
Curr Clin Pharmacol ; 11(1): 28-42, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26725878

RESUMEN

Microvesicles (MVs) are submicron intact particles released from the cellular membrane of eukaryotic cells. MVs can be sub-categorised into microparticles (MPs), which are between 100nm- 1micron in size, and exosomes, measuring less than 100nm. Once thought to be cellular debris, MPs are now known to play important biological effector functions. Their biogenesis and release are as a result highly regulated processes in response to cellular activation or stress, and apoptosis. MPs are now known to play a crucial role in maintaining physiological homeostasis and have been demonstrated to be involved in numerous biological processes, including inflammation, cardiovascular disease, immune response, cancer dissemination, coagulation and angiogenesis. Consequently, there is active interest in studying MPs, and their 'cause and effect' in the initiation and potentiation of various pathologies. Circulating levels, both quantitative and qualitative, of MPs is thought to be a reflective index of cardiovascular competence. Therefore, studies to understand the biological relevance of the various permutations and combinations of circulating MPs, their cellular origin and bioactive cargo may lead to increased understanding of the sequelae of CVD and associated diseases. This review synopsizes our current understanding of the role of MPs in cardiovascular disease, their biogenesis and effector function, and their future use as both diagnostic and prognostic indices of cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Micropartículas Derivadas de Células/metabolismo , Endotelio Vascular/metabolismo , Homeostasis/fisiología , Animales , Apoptosis/fisiología , Enfermedades Cardiovasculares/patología , Micropartículas Derivadas de Células/patología , Endotelio Vascular/patología , Humanos
18.
Int J Biochem Cell Biol ; 77(Pt A): 171-180, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27339040

RESUMEN

INTRODUCTION: Receptor activator of nuclear factor kappa beta-ligand (RANKL) is thought to promote vascular calcification (VC) by inducing osteoblastic behaviour in vascular smooth muscle cells (VSMC) in an ill-defined process. The present study assessed whether RANKL affects pro-osteoblastic paracrine signalling between human aortic endothelial cells (HAEC) and human aortic smooth muscle cells (HASMC) using both conditioned media transfer and cell co-culture experimental approaches. METHODS AND RESULTS: For initial experiments (6-well format), HAEC-conditioned media was harvested following 72h exposure to RANKL, and transferred to reporter HASMCs with/without noggin, an inhibitor of pro-osteoblastic bone morphogenetic protein (BMP) paracrine signalling. In further experiments, HAECs and HASMCs were co-cultured within the CellMax(®) Duo, a perfusing bioreactor unit that mimics the flow-mediated co-interaction of these cells within the arterial wall, and RANKL was added to the perfusing media for 72h. At the conclusion of each experiment markers of osteoblastic activity were measured in HASMCs, including alkaline phosphatase (ALP) activity, mRNA levels of ALP and Runx2, as well as BMP-2 and BMP-4 concentrations. RANKL increased BMP-2 release from HAECs, while exposure of HASMCs to RANKL-treated HAEC-conditioned media induced osteoblastic behaviour in HASMCs, an effect prevented by noggin. Within the CellMax(®) Duo bioreactor, the addition of RANKL to the intraluminal HAECs also produced an increase in BMP-2 and increased osteoblastic behaviour within the co-cultured HASMC population. CONCLUSIONS: RANKL promotes VC by inducing BMP-2 release from HAECs, which in turn appears to act in a paracrine fashion on the adjacent HASMC population to increase osteoblastic activity.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Células Endoteliales/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/diagnóstico por imagen , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Ligando RANK/farmacología , Adulto , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Masculino , Adulto Joven
19.
Atherosclerosis ; 247: 87-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878368

RESUMEN

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein that belongs to the tumour necrosis factor (TNF) cytokine superfamily. TRAIL is expressed by numerous cell types including vascular cells, immune cells and adipocytes. Although originally thought to induce apoptosis in malignant or transformed cells only, it is now known that TRAIL can bind up to 5 distinct receptors to activate complex signalling pathways, and is capable of exerting pleiotropic effects in non-transformed cells. In this respect, a number of clinical and animal studies point to the potential vasoprotective influence of TRAIL, with TRAIL deficiency being linked to accelerated atherosclerosis and vascular calcification. Moreover, exogenous TRAIL administration has been shown to exhibit anti-atherosclerotic activity in-vivo. In-vitro studies on TRAIL in this context have yielded conflicting results however, with evidence of both pro-atherogenic and vasoprotective effects ascribed to TRAIL. Notwithstanding these various studies, mechanistic information on the precise nature of TRAIL-mediated injury/protection within the vasculature, as well as the identity of the downstream molecular/cellular targets of TRAIL, is still quite limited. In this review, we will summarize our current knowledge of TRAIL regulation, signalling mechanisms, and its apparent involvement in CVD pathogenesis as a prelude to examining the existing evidence for TRAIL-mediated vasoprotection. To this end, extensive in vitro, in vivo, and clinical studies will be reviewed and critical findings highlighted.


Asunto(s)
Aterosclerosis/patología , Vasos Sanguíneos/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Fármacos Cardiovasculares/uso terapéutico , Humanos , Factores Protectores , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
20.
Biomed Res Int ; 2016: 2465763, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26942189

RESUMEN

Biomarkers encompass a wide range of different measurable indicators, representing a tangible link to physiological changes occurring within the body. Accessibility, sensitivity, and specificity are significant factors in biomarker suitability. New biomarkers continue to be discovered, and questions over appropriate selection and assessment of their usefulness remain. If traditional markers of inflammation are not sufficiently robust in their specificity, then perhaps alternative means of detection may provide more information. Epigenetic drift (epigenetic modifications as they occur as a direct function with age), and its ancillary elements, including platelets, secreted microvesicles (MVs), and microRNA (miRNA), may hold enormous predictive potential. The majority of epigenetic drift observed in blood is independent of variations in blood cell composition, addressing concerns affecting traditional blood-based biomarker efficacy. MVs are found in plasma and other biological fluids in healthy individuals. Altered MV/miRNA profiles may also be found in individuals with various diseases. Platelets are also highly reflective of physiological and lifestyle changes, making them extremely sensitive biomarkers of human health. Platelets release increased levels of MVs in response to various stimuli and under a plethora of disease states, which demonstrate a functional effect on other cell types.


Asunto(s)
Biomarcadores de Tumor/genética , Epigénesis Genética/genética , Inflamación/genética , MicroARNs/genética , Envejecimiento/sangre , Envejecimiento/patología , Biomarcadores de Tumor/sangre , Plaquetas , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/patología , Humanos , Inflamación/sangre , Inflamación/patología , MicroARNs/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA