Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Stem Cells ; 41(4): 341-353, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36639926

RESUMEN

Human induced pluripotent stem cells (hiPSCs) not only provide an abundant source of vascular cells for potential therapeutic applications in vascular disease but also constitute an excellent model for understanding the mechanisms that regulate the differentiation and the functionality of vascular cells. Here, we reported that myocyte enhancer factor 2C (MEF2C) transcription factor, but not any other members of the MEF2 family, was robustly upregulated during the differentiation of vascular progenitors and endothelial cells (ECs) from hiPSCs. Vascular endothelial growth factors (VEGF) strongly induced MEF2C expression in endothelial lineage cells. The specific upregulation of MEF2C during the commitment of endothelial lineage was dependent on the extracellular signal regulated kinase (ERK). Moreover, knockdown of MEF2C with shRNA in hiPSCs did not affect the differentiation of ECs from these hiPSCs, but greatly reduced the migration and tube formation capacity of the hiPSC-derived ECs. Through a chromatin immunoprecipitation-sequencing, genome-wide RNA-sequencing, quantitative RT-PCR, and immunostaining analyses of the hiPSC-derived endothelial lineage cells with MEF2C inhibition or knockdown compared to control hiPSC-derived ECs, we identified TNF-related apoptosis inducing ligand (TRAIL) and transmembrane protein 100 (TMEM100) as novel targets of MEF2C. This study demonstrates an important role for MEF2C in regulating human EC functions and highlights MEF2C and its downstream effectors as potential targets to treat vascular malfunction-associated diseases.


Asunto(s)
Células Endoteliales , Células Madre Pluripotentes Inducidas , Humanos , Células Endoteliales/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Diferenciación Celular/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética
2.
J Immunol ; 205(10): 2806-2820, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055281

RESUMEN

Intermediate monocytes (iMo; CD14+CD16+) increase in number in the circulation of patients with unstable coronary artery disease (CAD), and their recruitment to inflamed arteries is implicated in events leading to mortality following MI. Monocyte recruitment to inflamed coronary arteries is initiated by high affinity ß2-integrin (CD11c/CD18) that activates ß1-integrin (VLA-4) to bind endothelial VCAM-1. How integrin binding under shear stress mechanosignals a functional shift in iMo toward an inflammatory phenotype associated with CAD progression is unknown. Whole blood samples from patients treated for symptomatic CAD including non-ST elevation MI, along with healthy age-matched subjects, were collected to assess chemokine and integrin receptor levels on monocytes. Recruitment on inflamed human aortic endothelium or rVCAM-1 under fluid shear stress was assessed using a microfluidic-based artery on a chip (A-Chip). Membrane upregulation of high affinity CD11c correlated with concomitant activation of VLA-4 within focal adhesive contacts was required for arrest and diapedesis across inflamed arterial endothelium to a greater extent in non-ST elevation MI compared with stable CAD patients. The subsequent conversion of CD11c from a high to low affinity state under fluid shear activated phospho-Syk- and ADAM17-mediated proteolytic cleavage of CD16. This marked the conversion of iMo to an inflammatory phenotype associated with nuclear translocation of NF-κB and production of IL-1ß+ We conclude that CD11c functions as a mechanoregulator that activates an inflammatory state preferentially in a majority of iMo from cardiac patients but not healthy patients.


Asunto(s)
Antígeno CD11c/metabolismo , Enfermedad de la Arteria Coronaria/inmunología , Endotelio Vascular/inmunología , Monocitos/inmunología , Infarto del Miocardio sin Elevación del ST/inmunología , Adulto , Anciano , Regulación Alostérica/inmunología , Aorta/citología , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Línea Celular , Membrana Celular/metabolismo , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/cirugía , Vasos Coronarios/citología , Vasos Coronarios/inmunología , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Femenino , Humanos , Integrina alfa4beta1/metabolismo , Dispositivos Laboratorio en un Chip , Masculino , Técnicas Analíticas Microfluídicas/instrumentación , Persona de Mediana Edad , Infarto del Miocardio sin Elevación del ST/sangre , Infarto del Miocardio sin Elevación del ST/cirugía , Intervención Coronaria Percutánea , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
3.
Am J Respir Cell Mol Biol ; 65(6): 646-657, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34251297

RESUMEN

Compromised endothelial-cell (EC) barrier function is a hallmark of inflammatory diseases. mTOR inhibitors, widely applied as clinical therapies, cause pneumonitis through mechanisms that are not yet fully understood. This study aimed to elucidate the EC mechanisms underlying the pathogenesis of pneumonitis caused by mTOR inhibition (mTORi). Mice with EC-specific deletion of mTOR complex components (Mtor, Rptor or Rictor) were administered LPS to induce pulmonary injury. Cultured ECs were treated with pharmacologic inhibitors, siRNA, or overexpression plasmids. EC barrier function was evaluated in vivo with Evans blue assay and in vitro by measurement of transendothelial electrical resistance and albumin flux. mTORi increased basal and TNFα-induced EC permeability, which was caused by myosin light chain (MLC) phosphorylation-dependent cell contraction. Inactivation of mTOR kinase activity by mTORi triggered PKCδ/p38/NF-κB signaling that significantly upregulated TNFα-induced MLCK (MLC kinase) expression, whereas Raptor promoted the phosphorylation of PKCα/MYPT1 independently of its interaction with mTOR, leading to suppression of MLCP (MLC phosphatase) activity. EC-specific deficiency in mTOR, Raptor or Rictor aggravated lung inflammation in LPS-treated mice. These findings reveal that mTORi induces PKC-dependent endothelial MLC phosphorylation, contraction, and hyperpermeability that promote pneumonitis.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores mTOR/efectos adversos , Neumonía/enzimología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Lipopolisacáridos/toxicidad , Inhibidores mTOR/farmacología , Ratones , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Permeabilidad , Fosforilación/efectos de los fármacos , Neumonía/inducido químicamente , Serina-Treonina Quinasas TOR/metabolismo
4.
J Mol Cell Cardiol ; 140: 30-41, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32087218

RESUMEN

AIMS: Mammalian target of rapamycin (mTOR) inhibitors used in drug-eluting stents (DES) to control restenosis have been found to delay endothelialization and increase incidence of late-stent thrombosis through mechanisms not completely understood. We revealed that mTOR inhibition (mTORi) upregulated the expression of cell growth suppressor IRF-1 in primary human arterial endothelial cells (HAEC). This study aimed to examine how mTOR-regulated IRF-1 expression contributes to the suppressive effect of mTORi on arterial endothelial proliferation. METHODS AND RESULTS: Western blotting, quantitative PCR, and a dual-luciferase reporter assay indicated that mTOR inhibitors rapamycin and torin 1 upregulated IRF-1 expression and increased its transcriptional activity. IRF-1 in turn contributed to the suppressive effect of mTORi by mediating HAEC apoptosis and cell cycle arrest in part through upregulation of caspase 1 and downregulation of cyclin D3, as revealed by CCK-8 assay, Annexin V binding assay, measurement of activated caspase 3, BrdU incorporation assay, and matrigel tube formation assay. In a mouse model of femoral artery wire injury, administration of rapamycin inhibited EC recovery, an effect alleviated by EC deficiency of IRF-1. Chromatin immunoprecipitation assay with HAEC and rescue expression of wild type or dominant-negative IRF-1 in EC isolated from Irf1-/- mice confirmed transcriptional regulation of IRF-1 on the expression of CASP1 and CCND3. Furthermore, mTORi activated multiple PKC members, among which PKCζ was responsible for the growth-inhibitory effect on HAEC. Activated PKCζ increased IRF1 transcription through JAK/STAT-1 and NF-κB signaling. Finally, overexpression of wild type or mutant raptor incapable of binding mTOR indicated that mTOR-free raptor contributed to PKCζ activation in mTOR-inhibited HAEC. CONCLUSIONS: The study reveals an IRF-1-mediated mechanism that contributes to the suppressive effects of mTORi on HAEC proliferation. Further study may facilitate the development of effective strategies to reduce the side effects of DES used in coronary interventions.


Asunto(s)
Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Arteria Femoral/lesiones , Humanos , Factor 1 Regulador del Interferón/genética , Ratones , Ratones Noqueados , Naftiridinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Transfección
5.
FASEB J ; 33(11): 12888-12899, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31499005

RESUMEN

Endothelial up-regulation of VCAM-1 at susceptible sites in arteries modulates the recruitment efficiency of inflammatory monocytes that initiates atherosclerotic lesion formation. We reported that hydrodynamic shear stress (SS) mechanoregulates inflammation in human aortic endothelial cells through endoplasmic reticulum (ER) stress via activation of the transcription factor x-box binding protein 1 (XBP1). Here, a microfluidic flow channel that produces a linear gradient of SS along a continuous monolayer of endothelium was used to delve the mechanisms underlying transcriptional regulation of TNF-α-stimulated VCAM-1 expression. High-resolution immunofluorescence imaging enabled continuous detection of platelet endothelial cell adhesion molecule 1 (PECAM-1)-dependent, outside-in signaling as a function of SS magnitude. Differential expression of VCAM-1 and intercellular adhesion molecule 1 (ICAM-1) was regulated by the spatiotemporal activation of MAPKs, ER stress markers, and transcription factors, which was dependent on the mechanosensing of SS through PECAM-1 and PI3K. Inhibition of p38 specifically abrogated the rise to peak VCAM-1 at low SS (2 dyn/cm2), whereas inhibition of ERK1/2 attenuated peak ICAM-1 at high SS (12 dyn/cm2). A shear stress-regulated temporal rise in p38 phosphorylation activated the nuclear translocation of XBP1, which together with the transcription factor IFN regulatory factor 1, promoted maximum VCAM-1 expression. These data reveal a mechanism by which SS sensitizes the endothelium to a cytokine-induced ER stress response to spatially regulate inflammation promoting atherosclerosis.-Bailey, K. A., Moreno, E., Haj, F. G., Simon, S. I., Passerini, A. G. Mechanoregulation of p38 activity enhances endoplasmic reticulum stress-mediated inflammation by arterial endothelium.


Asunto(s)
Arterias/patología , Estrés del Retículo Endoplásmico , Endotelio Vascular/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Arterias/enzimología , Núcleo Celular/metabolismo , Células Cultivadas , Endotelio Vascular/enzimología , Femenino , Humanos , Factor 1 Regulador del Interferón/metabolismo , Masculino , Fosforilación , Transcripción Genética , Molécula 1 de Adhesión Celular Vascular/genética
6.
J Lipid Res ; 60(12): 1996-2005, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31641036

RESUMEN

Epoxyeicosatrienoic acids (EETs) are formed from the metabolism of arachidonic acid by cytochrome P450s. EETs promote angiogenesis linked to tumor growth in various cancer models that is attenuated in vivo by cyclooxygenase 2 (COX-2) inhibitors. This study further defines a role for COX-2 in mediating endothelial EET metabolism promoting angiogenesis. Using human aortic endothelial cells (HAECs), we quantified 8,9-EET-induced tube formation and cell migration as indicators of angiogenic potential in the presence and absence of a COX-2 inducer [phorbol 12,13-dibutyrate (PDBu)]. The angiogenic response to 8,9-EET in the presence of PDBu was 3-fold that elicited by 8,9-EET stabilized with a soluble epoxide hydrolase inhibitor (t-TUCB). Contributing to this response was the COX-2 metabolite of 8,9-EET, the 11-hydroxy-8,9-EET (8,9,11-EHET), which exogenously enhanced angiogenic responses in HAECs at levels comparable to those elicited by vascular endothelial growth factor (VEGF). In contrast, the 15-hydroxy-8,9-EET isomer was also formed but inactive. The 8,9,11-EHET also promoted expression of the VEGF family of tyrosine kinase receptors. These results indicate that 8,9-EET-stimulated angiogenesis is enhanced by COX-2 metabolism in the endothelium through the formation of 8,9,11-EHET. This alternative pathway for the metabolism of 8,9-EET may be particularly important in regulating angiogenesis under circumstances in which COX-2 is induced, such as in cancer tumor growth and inflammation.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Ciclooxigenasa 2/metabolismo , Cicloparafinas/farmacología , Eicosanoides/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 316(2): H408-H420, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462552

RESUMEN

Increased expression of vascular cell adhesion molecule 1 (VCAM-1) on the aortic endothelium is an early marker of atherogenesis, promoted in part by elevated levels of inflammatory cytokines such as TNF-α. Mammalian target of rapamycin (mTOR) is a ubiquitous signaling molecule that has been considered to contribute to diverse cellular processes through mTOR complex 1 (mTORC1) or complex 2 (mTORC2). This study aimed to elucidate the role of mTOR signaling in TNF-α-induced VCAM-1 expression by the arterial endothelium. Primary human aortic endothelial cells (HAECs) were treated with low-dose (0.1 ng/ml) TNF-α, and VCAM-1 expression was measured by real-time quantitative PCR, Western blot analysis, and flow cytometry. Inhibition of mTOR through siRNA-mediated depletion or treatment with chemical inhibitors rapamycin or torin 1 suppressed VCAM1 transcription, which translated to inhibition of VCAM-1 surface expression by HAECs and concomitant decreased adhesion of monocytes. A promoter luciferase assay and chromatin immunoprecipitation indicated that mTOR regulated VCAM1 transcription through a mechanism involving transcription factor GATA6. Activation of PKC-α and an increase in miR-200a-3p expression, caused by mTOR inhibition but not disruption of mTORC1 or mTORC2 singly or together, decreased TNF-α-induced GATA6 expression and its enrichment at the VCAM1 promoter. In conclusion, mTOR inhibition activates PKC-α independently of disruption of mTORC1 and/or mTORC2, which challenges the conventional wisdom regarding mTOR signaling. Moreover, mTOR signals through transcriptional and posttranscriptional mechanisms to elicit maximal cytokine-induced endothelial inflammation that precedes atherosclerosis. NEW & NOTEWORTHY Both mammalian target of rapamycin (mTOR) complex 1 (mTORC1) and mTORC2 contribute to PKC-α activation in the human aortic endothelium. Inhibition of mTOR is not equivalent to disruption of mTORC1 and/or mTORC2 in affecting human aortic endothelial cell signaling. Specifically, inhibition of mTOR causes PKC-α activation and miR-200a-3p upregulation, which independently suppresses TNF-α-induced transcription factor GATA6 expression and subsequently inhibits VCAM-1 expression and monocytic cell adhesion onto the aortic endothelium.


Asunto(s)
Aterosclerosis/metabolismo , Adhesión Celular , Células Endoteliales/metabolismo , Factor de Transcripción GATA6/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Aorta/citología , Aorta/metabolismo , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Factor de Transcripción GATA6/genética , Humanos , Monocitos/fisiología , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética
8.
Am J Physiol Heart Circ Physiol ; 309(7): H1130-40, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26254329

RESUMEN

Mechanisms of restenosis in type 2 diabetes mellitus (T2DM) are incompletely elucidated, but advanced glycation end-product (AGE)-induced vascular remodeling likely contributes. We tested the hypothesis that AGE-related collagen cross-linking (ARCC) leads to increased downstream vascular resistance and altered in-stent hemodynamics, thereby promoting neointimal hyperplasia (NH) in T2DM. We proposed that decreasing ARCC with ALT-711 (Alagebrium) would mitigate this response. Abdominal aortic stents were implanted in Zucker lean (ZL), obese (ZO), and diabetic (ZD) rats. Blood flow, vessel diameter, and wall shear stress (WSS) were calculated after 21 days, and NH was quantified. Arterial segments (aorta, carotid, iliac, femoral, and arterioles) were harvested to detect ARCC and protein expression, including transforming growth factor-ß (TGF-ß) and receptor for AGEs (RAGE). Downstream resistance was elevated (60%), whereas flow and WSS were significantly decreased (44% and 56%) in ZD vs. ZL rats. NH was increased in ZO but not ZD rats. ALT-711 reduced ARCC and resistance (46%) in ZD rats while decreasing NH and producing similar in-stent WSS across groups. No consistent differences in RAGE or TGF-ß expression were observed in arterial segments. ALT-711 modified lectin-type oxidized LDL receptor 1 but not RAGE expression by cells on decellularized matrices. In conclusion, ALT-711 decreased ARCC, increased in-stent flow rate, and reduced NH in ZO and ZD rats through RAGE-independent pathways. The study supports an important role for AGE-induced remodeling within and downstream of stent implantation to promote enhanced NH in T2DM.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Diabetes Mellitus/metabolismo , Oclusión de Injerto Vascular/metabolismo , Neointima/metabolismo , Obesidad/metabolismo , Stents , Estrés Mecánico , Tiazoles/farmacología , Resistencia Vascular/efectos de los fármacos , Animales , Aorta Abdominal/metabolismo , Colágeno/efectos de los fármacos , Colágeno/metabolismo , Productos Finales de Glicación Avanzada/efectos de los fármacos , Productos Finales de Glicación Avanzada/metabolismo , Masculino , Neointima/prevención & control , Ratas , Ratas Zucker , Receptor para Productos Finales de Glicación Avanzada/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Resistencia al Corte , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
9.
Circ Res ; 111(8): 1054-64, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22874466

RESUMEN

RATIONALE: A high-fat diet accompanied by hypertriglyceridemia increases an individual's risk for development of atherosclerosis. An early event in this process is monocyte recruitment through binding to vascular cell adhesion molecule 1 (VCAM-1) upregulated on inflamed arterial endothelium. Diets high in polyunsaturated fatty acids (PUFAs) may provide athero-protection by ameliorating this effect. OBJECTIVE: We investigated the acute regulation of VCAM-1 expression in human aortic endothelial cells (HAEC) in response to triglyceride-rich lipoproteins (TGRL) isolated from subjects after consumption of a high-fat meal. METHODS AND RESULTS: Postprandial TGRL isolated from 38 subjects were categorized as proatherogenic or antiatherogenic according to their capacity to alter the inflammatory response of HAEC. Proatherogenic TGRL increased expression of VCAM-1, intercellular adhesion molecule 1 (ICAM-1), and E-selectin by ≈20% compared with stimulation with tumor necrosis factor-α alone, whereas antiatherogenic TGRL decreased VCAM-1 expression by ≈20% while still upregulating ICAM-1. The relative atherogenicity of TGRL positively correlated with particle density of TG, apolipoprotein (Apo)CIII, ApoE, and cholesterol. Ω3-PUFA mimicked the effect of antiatherogenic TGRL by downregulating VCAM-1 expression. TGRL exerted this differential regulation of VCAM-1 by reciprocally modulating expression and activity of the transcription factor interferon regulatory factor 1 (IRF-1) and expression of microRNA 126 (miR-126). Overexpression or silencing of IRF-1 or miR-126 expression recapitulated the proatherogenic or antiatherogenic regulation of VCAM-1. CONCLUSIONS: In response to a high-fat meal, TGRL bias the inflammatory response of endothelium via transcriptional and posttranscriptional editing of VCAM-1. Subjects with an anti-inflammatory response to a meal produced TGRL that was enriched in nonesterified fatty acids, decreased IRF-1 expression, increased miR-126 activity, and diminished monocyte arrest.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Factor 1 Regulador del Interferón/metabolismo , MicroARNs/fisiología , Molécula 1 de Adhesión Celular Vascular/genética , Aorta/citología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Adhesión Celular/fisiología , Línea Celular , Grasas Insaturadas en la Dieta/administración & dosificación , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Factor 1 Regulador del Interferón/genética , Monocitos/metabolismo , FN-kappa B/metabolismo , Periodo Posprandial/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 305(8): H1149-57, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23934855

RESUMEN

Dyslipidemia is a primary risk factor for cardiovascular disease, but the specific mechanisms that determine the localization of atherosclerotic plaques in arteries are not well defined. Triglyceride-rich lipoproteins (TGRL) isolated from human plasma after a high-fat meal modulate TNF-α-induced VCAM-1 expression in cultured human aortic endothelial cells (HAECs) via an interferon regulatory factor (IRF)-1-dependent transcriptional mechanism. We examined whether fluid shear stress acts as a mediator of IRF-1-dependent VCAM-1 expression in response to cytokine and dietary lipids. IRF-1 and VCAM-1 were examined by immunofluorescence in TNF-α-stimulated HAEC monolayers exposed to TGRL and a linear gradient of shear stress ranging from 0 to 16 dyn/cm(2) in a microfluidic device. Shear stress alone modulated TNF-α-induced VCAM-1 expression, eliciting a 150% increase at low shear stress (2 dyn/cm(2)) and a 70% decrease at high shear stress (12 dyn/cm(2)) relative to static. These differences correlated with a 60% increase in IRF-1 expression under low shear stress and a 40% decrease under high shear stress. The addition of TGRL along with cytokine activated a fourfold increase in VCAM-1 expression and a twofold increase in IRF-1 expression. The combined effect of shear stress and TGRL on the upregulation of membrane VCAM-1 was abolished by transfection of HAECs with IRF-1-specific small interfering RNA. In a healthy swine model, elevated levels of endothelial IRF-1 were also observed within atherosusceptible regions of the aorta by Western blot analysis and immunohistochemistry, implicating arterial hemodynamics in the regulation of IRF-1 expression. These data demonstrate direct roles for fluid shear stress and postprandial TGRL from human serum in the regulation of IRF-1 expression and downstream inflammatory responses in HAECs.


Asunto(s)
Aorta/metabolismo , Grasas de la Dieta/farmacología , Células Endoteliales/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Estrés Mecánico , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Aorta/citología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Factor 1 Regulador del Interferón/efectos de los fármacos , Periodo Posprandial , Porcinos , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/efectos de los fármacos
11.
Transl Res ; 255: 1-13, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36384204

RESUMEN

Cell inflammation and death are closely linked processes contributing to endothelial dysfunction, which plays a critical role in atherogenesis. Activation of the NLRP3 inflammasome causes pyroptosis, the Gasdermin D (GSDMD)-mediated inflammatory cell death. The non-canonical NF-κB pathway has been implicated in inflammation; however, its role in NLRP3 inflammasome-mediated endothelial dysfunction has not been investigated. This study investigated a role for the non-canonical NF-κB pathway in regulating endothelial pyroptosis as it relates to atherogenesis. Immunohistochemistry indicated inflammasome activation in the endothelial cells (EC) of human atherosclerotic arteries. Flow cytometry and Western blot analysis revealed that oxidized low-density lipoprotein (oxLDL) activated the NLRP3 inflammasome, concomitant with the activation of non-canonical NF-κB in primary human aortic EC. Interference of NF-κB inducing kinase (NIK), the key regulator of the non-canonical pathway, significantly attenuated oxLDL- or LPS/ATP-induced NLRP3 inflammasome activation, pyroptosis, IL-1ß, and IL-18 secretion. In contrast, overexpression of NIK exacerbated these responses. Chromatin immunoprecipitation revealed that activation of the non-canonical NF-κB pathway upregulated the transcription factor IRF-1 through RelB/p52 binding to its promoter region at -782/-770. In addition to the known target CASP1, RNA sequencing further identified GSDMD as a target gene of IRF-1. IRF-1 but not RelB/p52 interacted with the GSDMD promoter at -526/-515 and the CASP1 promoter at -11/10 to promote the expression and CASP1-mediated activation of GSDMD. Consistent with the observations in cultured endothelium, endothelial-specific deficiency of NIK or IRF-1 attenuated atherosclerosis in high-fat diet-fed Apoe-null mice. These data demonstrate that the non-canonical NF-κB pathway contributes to NLRP3 inflammasome-mediated endothelial pyroptosis and the development of atherosclerosis through GSDMD activation in a manner dependent on IRF-1. Further investigation may facilitate the identification of specific therapeutic targets for atherosclerotic heart diseases.


Asunto(s)
Aterosclerosis , FN-kappa B , Ratones , Animales , Humanos , FN-kappa B/metabolismo , Inflamasomas/metabolismo , Piroptosis/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Células Endoteliales/metabolismo , Endotelio/metabolismo , Inflamación
12.
Am J Physiol Heart Circ Physiol ; 302(12): H2498-508, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22467309

RESUMEN

Atherosclerosis occurs preferentially at sites of disturbed blood flow despite the influence of risk factors contributing to systemic inflammation. The receptor for advanced glycation endproducts (RAGE) is a prominent mediator of inflammation in diabetes that is upregulated in atherosclerotic plaques. Our goal was to elucidate a role for arterial hemodynamics in the regulation of RAGE expression and activity. Endothelial RAGE expression was elevated at sites of flow disturbance in the aortas of healthy swine. To demonstrate a direct role for physiological shear stress (SS) in modulating RAGE expression, human aortic endothelial cells (HAEC) were exposed to high SS (HSS; 15 dyn/cm(2)), which downregulated RAGE by fourfold, or oscillatory SS (OSS; 0 ± 5 dyn/cm(2)), which upregulated RAGE by threefold, compared with static culture at 4 h. In a model of diabetes-induced metabolic stress, HAEC were chronically conditioned under high glucose (25 mM) and then simultaneously stimulated with TNF-α (0.5 ng/ml) and the RAGE ligand high mobility group box 1 (HMGB1). A 50% increase in VCAM-1 expression over TNF-α was associated with increased cytoplasmic and mitochondrial reactive oxygen species and NF-κB activity. This increase was RAGE-specific and NADPH oxidase dependent. In activated HAEC, OSS amplified HMGB1-induced VCAM-1 (3-fold) and RAGE (1.6-fold) expression and proportionally enhanced monocyte adhesion to HAEC in a RAGE-dependent manner, while HSS mitigated these increases to the level of TNF-α alone. We demonstrate that SS plays a fundamental role in regulating RAGE expression and inflammatory responses in the endothelium. These findings may provide mechanistic insight into how diabetes accelerates the nonrandom distribution of atherosclerosis in arteries.


Asunto(s)
Diabetes Mellitus/fisiopatología , Células Endoteliales/fisiología , Endotelio Vascular/fisiopatología , Inflamación/fisiopatología , Receptores Inmunológicos/metabolismo , Estrés Fisiológico/fisiología , Animales , Aorta/citología , Aorta/metabolismo , Aorta/fisiopatología , Diabetes Mellitus/metabolismo , Células Endoteliales/citología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Estrés Mecánico , Porcinos , Factor de Necrosis Tumoral alfa , Molécula 1 de Adhesión Celular Vascular/metabolismo
13.
Commun Biol ; 5(1): 726, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35869262

RESUMEN

Clinically used inhibitors of mammalian target of rapamycin (mTOR) negatively impacts endothelial-dependent vasodilatation (EDD) through unidentified mechanisms. Here we show that either the endothelium-specific deletion of Mtor to inhibit both mTOR complexes, or depletion of Raptor or Rictor to disrupt mTORC1 or mTORC2, causes impaired EDD, accompanied by reduced NO in the serum of mice. Consistently, inhibition of mTOR decreases NO production by human and mouse EC. Specifically, inhibition of mTORC1 suppresses eNOS gene expression, due to impairment in p70S6K-mediated posttranscriptional regulation of the transcription factor KLF2 expression. In contrast to mTORC1 inhibition, a positive-feedback between MAPK (p38 and JNK) activation and Nox2 upregulation contributes to the excessive generation of reactive oxygen species (ROS), which causes eNOS uncoupling and decreased NO bioavailability in mTORC2-inhibited EC. Adeno-associated virus-mediated EC-specific overexpression of KLF2 or suppression of Nox2 restores EDD function in endothelial mTORC1- or mTORC2-inhibited mice.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vasodilatación , Animales , Endotelio/metabolismo , Humanos , Mamíferos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Sirolimus/farmacología
14.
Am J Physiol Heart Circ Physiol ; 300(3): H784-91, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21169396

RESUMEN

A rise in postprandial serum triglycerides (PP-sTG) can potentiate inflammatory responses in vascular endothelial cells (ECs) and thus serves as an independent risk factor for predicting increased cardiovascular morbidity. We examined postprandial triglyceride-rich lipoproteins (PP-TGRLs) in subjects ranging from normal to hypertriglyceridemic for their capacity to alter EC acute inflammatory responses. Cultured human aortic ECs (HAECs) were conditioned with PP-TGRLs isolated from human serum at the peak after a moderately high-fat meal. VLDL particle size increased postprandially and varied directly with the subject's PP-sTG level and waist circumference. PP-TGRL particles bound to HAECs and were internalized via LDL receptor-mediated endocytosis. PP-TGRL alone did not induce an inflammatory response over the range of individuals studied. However, combined with low-dose TNF-α stimulation (0.3 ng/ml), it elicited a net 10-15% increase above cytokine alone in the membrane expression of VCAM-1, ICAM-1, and E-selectin, which was not observed with fasting TGRLs. In contrast to upregulation of ICAM-1 and E-selectin, VCAM-1 transcription and expression varied in direct proportion with individual PP-sTG and waist circumference. The extent of monocyte arrest on inflamed HAECs under shear stress also correlated closely with VCAM-1 expression induced by conditioning with PP-TGRL and TNF-α stimulation. This ex vivo approach provides a quantitative means to assess an individual's inflammatory potential, revealing a greater propensity for endothelial inflammation in hypertriglyceridemic individuals with abdominal obesity.


Asunto(s)
Grasas de la Dieta/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Lipoproteínas/metabolismo , Triglicéridos/metabolismo , Aorta/metabolismo , Células Cultivadas , Selectina E/metabolismo , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lipoproteínas/sangre , Masculino , Monocitos/metabolismo , Obesidad Abdominal/metabolismo , Periodo Posprandial , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/administración & dosificación , Molécula 1 de Adhesión Celular Vascular/metabolismo , Circunferencia de la Cintura , Adulto Joven
15.
Theranostics ; 11(11): 5605-5619, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33859766

RESUMEN

Increased expression of vascular cell adhesion molecule (VCAM)-1 on the activated arterial endothelial cell (EC) surface critically contributes to atherosclerosis which may in part be regulated by epigenetic mechanisms. This study investigated whether and how the clinically available histone deacetylases 1 and 2 (HDAC1/2) inhibitor drug Romidepsin epigenetically modulates VCAM-1 expression to suppress atherosclerosis. Methods: VCAM-1 expression was analyzed in primary human aortic EC (HAEC) treated with Romidepsin or transfected with HDAC1/2-targeting siRNA. Methylation of GATA6 promoter region was examined with methylation-specific PCR assay. Enrichment of STAT3 to GATA6 promoter was detected with chromatin immunoprecipitation. Lys685Arg mutation was constructed to block STAT3 acetylation. The potential therapeutic effect of Romidepsin on atherosclerosis was evaluated in Apoe-/- mice fed with a high-fat diet. Results: Romidepsin significantly attenuated TNFα-induced VCAM-1 expression on HAEC surface and monocyte adhesion through simultaneous inhibition of HDAC1/2. This downregulation of VCAM-1 was attributable to reduced expression of transcription factor GATA6. Romidepsin enhanced STAT3 acetylation and its binding to DNA methyltransferase 1 (DNMT1), leading to hypermethylation of the GATA6 promoter CpG-rich region at +140/+255. Blocking STAT3 acetylation at Lys685 disrupted DNMT1-STAT3 interaction, decreased GATA6 promoter methylation, and reversed the suppressive effects of HDAC1/2 inhibition on GATA6 and VCAM-1 expression. Finally, intraperitoneal administration of Romidepsin reduced diet-induced atherosclerotic lesion development in Apoe-/- mice, accompanied by a reduction in GATA6/VCAM-1 expression in the aorta. Conclusions: HDAC1/2 contributes to VCAM-1 expression and atherosclerosis by suppressing STAT3 acetylation-dependent GATA6 promoter methylation. These findings may provide a rationale for HDAC1/2-targeting therapy in atherosclerotic heart disease.


Asunto(s)
Aterosclerosis/genética , Células Endoteliales/metabolismo , Factor de Transcripción GATA6/genética , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Regiones Promotoras Genéticas/genética , Molécula 1 de Adhesión Celular Vascular/genética , Animales , Aorta/metabolismo , Apolipoproteínas E/genética , Células Cultivadas , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , Regulación hacia Abajo/genética , Humanos , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Factor de Transcripción STAT3/genética , Células THP-1/metabolismo
16.
Semin Thromb Hemost ; 36(3): 265-75, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20533180

RESUMEN

Endothelial phenotypic heterogeneity plays an important role in the susceptibility of the cardiovascular system to disease. Arteries and heart valves are susceptible to chronic inflammatory disease in regions of blood flow disturbance that implicates hemodynamic forces and transport characteristics as prominent influences on endothelial phenotype. By combining in vivo high-throughput genomics (discovery science) and in vitro mechanistic approaches (reductionist science), we present endothelial patho-susceptibility as an imbalance of multiple interrelated pathways that sensitize the cells to pathological change. The recently identified association of endoplasmic reticulum stress with endothelium in regions of flow disturbance is outlined as an important example of susceptible phenotype linked to proinflammatory and oxidative stress pathways.


Asunto(s)
Arterias/patología , Aterosclerosis/fisiopatología , Endotelio Vascular/fisiopatología , Hemorreología , Adaptación Fisiológica , Animales , Aterosclerosis/patología , Susceptibilidad a Enfermedades , Retículo Endoplásmico/fisiología , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Perfilación de la Expresión Génica , Humanos , Hipercolesterolemia/complicaciones , Modelos Biológicos , FN-kappa B/metabolismo , Estrés Oxidativo , Fenotipo , Resistencia al Corte , Porcinos , Respuesta de Proteína Desplegada , Vasculitis/patología , Vasculitis/fisiopatología
17.
Circ Res ; 100(3): 381-90, 2007 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-17234968

RESUMEN

High levels of triglyceride-rich lipoproteins (TGRLs) in blood are linked to development of atherosclerosis, yet the mechanisms by which these particles initiate inflammation of endothelium are unknown. TGRL isolated from human plasma during the postprandial state was examined for its capacity to bind to cultured human aortic endothelial cells (HAECs) and alter the acute inflammatory response to tumor necrosis factor-alpha. HAECs were repetitively incubated with dietary levels of freshly isolated TGRL for 2 hours per day for 1 to 3 days to mimic postprandial lipidemia. TGRL induced membrane upregulation of the low-density lipoprotein family receptors LRP and LR11, which was inhibited by the low-density lipoprotein receptor-associated protein-1. TGRLs alone did not elicit inflammation in HAECs but enhanced the inflammatory response via a 10-fold increase in sensitivity to cytokine stimulation. This was reflected by increased mitogen-activated protein kinase activation, nuclear translocation of NF-kappaB, amplified expression of endothelial selectin and VCAM-1, and a subsequent increase in monocyte-specific recruitment under shear flow as quantified in a microfabricated vascular mimetic device.


Asunto(s)
Enfermedades de la Aorta/etiología , Arteriosclerosis/etiología , Arteritis/etiología , Grasas de la Dieta/efectos adversos , Células Endoteliales/efectos de los fármacos , Hipertrigliceridemia/complicaciones , Proteínas Relacionadas con Receptor de LDL/metabolismo , Lipoproteínas HDL/toxicidad , Lipoproteínas LDL/toxicidad , Lipoproteínas VLDL/toxicidad , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Receptores de LDL/metabolismo , Triglicéridos/toxicidad , Factor de Necrosis Tumoral alfa/farmacología , Aorta , Apolipoproteína C-III/metabolismo , Apolipoproteína C-III/farmacología , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Quilomicrones/sangre , Grasas de la Dieta/administración & dosificación , Selectina E/biosíntesis , Selectina E/genética , Endocitosis , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Emulsiones Grasas Intravenosas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertrigliceridemia/sangre , Hipoglucemia , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/genética , Proteína Asociada a Proteínas Relacionadas con Receptor de LDL/farmacología , Proteínas Relacionadas con Receptor de LDL/efectos de los fármacos , Leucocitos/citología , Leucocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Lipoproteínas VLDL/sangre , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/efectos de los fármacos , Proteínas de Transporte de Membrana/efectos de los fármacos , Modelos Cardiovasculares , Monocitos/citología , Monocitos/efectos de los fármacos , FN-kappa B/metabolismo , Estrés Oxidativo , Receptores de LDL/efectos de los fármacos , Reología , Transducción de Señal/efectos de los fármacos , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/fisiología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Sci Rep ; 9(1): 8655, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31209255

RESUMEN

Elevated triglyceride-rich lipoproteins (TGRL) in circulation is a risk factor for atherosclerosis. TGRL from subjects consuming a high saturated fat test meal elicited a variable inflammatory response in TNFα-stimulated endothelial cells (EC) that correlated strongly with the polyunsaturated fatty acid (PUFA) content. This study investigates how the relative abundance of oxygenated metabolites of PUFA, oxylipins, is altered in TGRL postprandially, and how these changes promote endothelial inflammation. Human aortic EC were stimulated with TNFα and treated with TGRL, isolated from subjects' plasma at fasting and 3.5 hrs postprandial to a test meal high in saturated fat. Endothelial VCAM-1 surface expression stimulated by TNFα provided a readout for atherogenic inflammation. Concentrations of esterified and non-esterified fatty acids and oxylipins in TGRL were quantified by mass spectrometry. Dyslipidemic subjects produced TGRL that increased endothelial VCAM-1 expression by ≥35%, and exhibited impaired fasting lipogenesis activity and a shift in soluble epoxide hydrolase and lipoxygenase activity. Pro-atherogenic TGRL were enriched in eicosapentaenoic acid metabolites and depleted in esterified C18-PUFA-derived diols. Abundance of these metabolites was strongly predictive of VCAM-1 expression. We conclude the altered metabolism in dyslipidemic subjects produces TGRL with a unique oxylipin signature that promotes a pro-atherogenic endothelial phenotype.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Dislipidemias/sangre , Epóxido Hidrolasas/genética , Ácidos Grasos Insaturados/administración & dosificación , Lipoproteínas/sangre , Oxilipinas/administración & dosificación , Triglicéridos/sangre , Adulto , Anciano , Estudios de Casos y Controles , Línea Celular , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dislipidemias/genética , Dislipidemias/patología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epóxido Hidrolasas/metabolismo , Ayuno , Ácidos Grasos Insaturados/sangre , Ácidos Grasos Insaturados/clasificación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación , Lipooxigenasa/genética , Lipooxigenasa/metabolismo , Masculino , Comidas , Persona de Mediana Edad , Oxilipinas/sangre , Oxilipinas/clasificación , Periodo Posprandial , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
Sci Rep ; 7(1): 8196, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811527

RESUMEN

Atherosclerosis impacts arteries where disturbed blood flow renders the endothelium susceptible to inflammation. Cytokine activation of endothelial cells (EC) upregulates VCAM-1 receptors that target monocyte recruitment to atherosusceptible regions. Endoplasmic reticulum (ER) stress elicits EC dysregulation in metabolic syndrome. We hypothesized that ER plays a central role in mechanosensing of atherosusceptible shear stress (SS) by signaling enhanced inflammation. Aortic EC were stimulated with low-dose TNFα (0.3 ng/ml) in a microfluidic channel that produced a linear SS gradient over a 20mm field ranging from 0-16 dynes/cm2. High-resolution imaging of immunofluorescence along the monolayer provided a continuous spatial metric of EC orientation, markers of ER stress, VCAM-1 and ICAM-1 expression, and monocyte recruitment. VCAM-1 peaked at 2 dynes/cm2 and decreased to below static TNFα-stimulated levels at atheroprotective-SS of 12 dynes/cm2, whereas ICAM-1 rose to a maximum in parallel with SS. ER expansion and activation of the unfolded protein response also peaked at 2 dynes/cm2, where IRF-1-regulated VCAM-1 expression and monocyte recruitment also rose to a maximum. Silencing of PECAM-1 or key ER stress genes abrogated SS regulation of VCAM-1 transcription and monocyte recruitment. We report a novel role for ER stress in mechanoregulation at arterial regions of atherosusceptible-SS inflamed by low-dose TNFα.


Asunto(s)
Estrés del Retículo Endoplásmico , Endotelio Vascular/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Resistencia al Corte , Estrés Mecánico , Adulto , Estrés del Retículo Endoplásmico/genética , Células Endoteliales/metabolismo , Femenino , Humanos , Leucocitos/inmunología , Leucocitos/metabolismo , Masculino , Modelos Biológicos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adulto Joven
20.
Physiol Genomics ; 13(2): 147-56, 2003 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-12700361

RESUMEN

Although mRNA amplification is necessary for microarray analyses from limited amounts of cells and tissues, the accuracy of transcription profiles following amplification has not been well characterized. We tested the fidelity of differential gene expression following linear amplification by T7-mediated transcription in a well-established in vitro model of cytokine [tumor necrosis factor alpha (TNFalpha)]-stimulated human endothelial cells using filter arrays of 13,824 human cDNAs. Transcriptional profiles generated from amplified antisense RNA (aRNA) (from 100 ng total RNA, approximately 1 ng mRNA) were compared with profiles generated from unamplified RNA originating from the same homogeneous pool. Amplification accurately identified TNFalpha-induced differential expression in 94% of the genes detected using unamplified samples. Furthermore, an additional 1,150 genes were identified as putatively differentially expressed using amplified RNA which remained undetected using unamplified RNA. Of genes sampled from this set, 67% were validated by quantitative real-time PCR as truly differentially expressed. Thus, in addition to demonstrating fidelity in gene expression relative to unamplified samples, linear amplification results in improved sensitivity of detection and enhances the discovery potential of high-throughput screening by microarrays.


Asunto(s)
Endotelio Vascular/química , Endotelio Vascular/metabolismo , Perfilación de la Expresión Génica/métodos , Nanotecnología/métodos , Técnicas de Amplificación de Ácido Nucleico/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/metabolismo , Sesgo , Línea Celular , Bases de Datos Genéticas , Endotelio Vascular/citología , Regulación de la Expresión Génica/genética , Humanos , Internet , Sensibilidad y Especificidad , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA