Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 35(11): 2354-65, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26338297

RESUMEN

OBJECTIVE: Collateral arteriogenesis, the growth of existing arterial vessels to a larger diameter, is a fundamental adaptive response that is often critical for the perfusion and survival of tissues downstream of chronic arterial occlusion(s). Shear stress regulates arteriogenesis; however, the arteriogenic significance of reversed flow direction, occurring in numerous collateral artery segments after femoral artery ligation, is unknown. Our objective was to determine if reversed flow direction in collateral artery segments differentially regulates endothelial cell signaling and arteriogenesis. APPROACH AND RESULTS: Collateral segments experiencing reversed flow direction after femoral artery ligation in C57BL/6 mice exhibit increased pericollateral macrophage recruitment, amplified arteriogenesis (30% diameter and 2.8-fold conductance increases), and remarkably permanent (12 weeks post femoral artery ligation) remodeling. Genome-wide transcriptional analyses on human umbilical vein endothelial cells exposed to reversed flow conditions mimicking those occurring in vivo yielded 10-fold more significantly regulated transcripts, as well as enhanced activation of upstream regulators (nuclear factor κB [NFκB], vascular endothelial growth factor, fibroblast growth factor-2, and transforming growth factor-ß) and arteriogenic canonical pathways (protein kinase A, phosphodiesterase, and mitogen-activated protein kinase). Augmented expression of key proarteriogenic molecules (Kruppel-like factor 2 [KLF2], intercellular adhesion molecule 1, and endothelial nitric oxide synthase) was also verified by quantitative real-time polymerase chain reaction, leading us to test whether intercellular adhesion molecule 1 or endothelial nitric oxide synthase regulate amplified arteriogenesis in flow-reversed collateral segments in vivo. Interestingly, enhanced pericollateral macrophage recruitment and amplified arteriogenesis was attenuated in flow-reversed collateral segments after femoral artery ligation in intercellular adhesion molecule 1(-/-) mice; however, endothelial nitric oxide synthase(-/-) mice showed no such differences. CONCLUSIONS: Reversed flow leads to a broad amplification of proarteriogenic endothelial signaling and a sustained intercellular adhesion molecule 1-dependent augmentation of arteriogenesis. Further investigation of the endothelial mechanotransduction pathways activated by reversed flow may lead to more effective and durable therapeutic options for arterial occlusive diseases.


Asunto(s)
Arterias/fisiopatología , Circulación Colateral , Isquemia/fisiopatología , Mecanotransducción Celular , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Animales , Arterias/metabolismo , Arterias/patología , Velocidad del Flujo Sanguíneo , Células Cultivadas , Modelos Animales de Enfermedad , Arteria Femoral/fisiopatología , Arteria Femoral/cirugía , Regulación de la Expresión Génica , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Ligadura , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Remodelación Vascular
2.
Arterioscler Thromb Vasc Biol ; 35(10): 2185-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26293464

RESUMEN

OBJECTIVES: The predictive value of animal and in vitro systems for drug development is limited, particularly for nonhuman primate studies as it is difficult to deduce the drug mechanism of action. We describe the development of an in vitro cynomolgus macaque vascular system that reflects the in vivo biology of healthy, atheroprone, or advanced inflammatory cardiovascular disease conditions. APPROACH AND RESULTS: We compare the responses of the in vitro human and cynomolgus vascular systems to 4 statins. Although statins exert beneficial pleiotropic effects on the human vasculature, the mechanism of action is difficult to investigate at the tissue level. Using RNA sequencing, we quantified the response to statins and report that most statins significantly increased the expression of genes that promote vascular health while suppressing inflammatory cytokine gene expression. Applying computational pathway analytics, we identified statin-regulated biological themes, independent of cholesterol lowering, that provide mechanisms for off-target effects, including thrombosis, cell cycle regulation, glycogen metabolism, and ethanol degradation. CONCLUSIONS: The cynomolgus vascular system described herein mimics the baseline and inflammatory regional biology of the human vasculature, including statin responsiveness, and provides mechanistic insight not achievable in vivo.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Evaluación Preclínica de Medicamentos/métodos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lipoproteínas LDL/efectos de los fármacos , Animales , Enfermedades Cardiovasculares/sangre , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Humanos , Técnicas In Vitro , Lipoproteínas LDL/metabolismo , Macaca fascicularis , Modelos Cardiovasculares , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Especificidad de la Especie
3.
J Cell Biol ; 176(5): 719-27, 2007 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-17312022

RESUMEN

Elevated permeability of the endothelium is thought to be crucial in atherogenesis because it allows circulating lipoproteins to access subendothelial monocytes. Both local hemodynamics and cytokines may govern endothelial permeability in atherosclerotic plaque. We recently found that p21-activated kinase (PAK) regulates endothelial permeability. We now report that onset of fluid flow, atherogenic flow profiles, oxidized LDL, and proatherosclerotic cytokines all stimulate PAK phosphorylation and recruitment to cell-cell junctions. Activation of PAK is higher in cells plated on fibronectin (FN) compared to basement membrane proteins in all cases. In vivo, PAK is activated in atherosclerosis-prone regions of arteries and correlates with FN in the subendothelium. Inhibiting PAK in vivo reduces permeability in atherosclerosis-prone regions. Matrix-specific PAK activation therefore mediates elevated vascular permeability in atherogenesis.


Asunto(s)
Aterosclerosis/enzimología , Permeabilidad Capilar , Matriz Extracelular/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Bovinos , Citocinas/farmacología , Activación Enzimática , Fibronectinas/metabolismo , Uniones Intercelulares/enzimología , Lipoproteínas LDL/farmacología , Fosforilación , Transducción de Señal , Quinasas p21 Activadas
4.
Circ Res ; 106(11): 1703-11, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20378855

RESUMEN

RATIONALE: The extracellular matrix protein fibronectin (FN) is focally deposited in regions of atherosclerosis, where it contributes to inflammatory signaling. OBJECTIVE: To elucidate the mechanism by which FN deposition is regulated by local shear stress patterns, its dependence on platelet-endothelial cell adhesion molecule (PECAM)-1 mechanotransduction and the role this pathway plays in sustaining an atheroprone/proinflammatory phenotype. METHODS AND RESULTS: Human endothelial cells were exposed in vitro to atheroprone or atheroprotective shear stress patterns derived from human carotid arteries. Onset of atheroprotective flow induced a transient increase in FN deposition, whereas atheroprone flow caused a steady increase in FN expression and integrin activation over time, leading to a significant and sustained increase in FN deposition relative to atheroprotective conditions. Comparing FN staining in ApoE(-/-) and ApoE(-/-)PECAM(-/-) mice showed that PECAM-1 was essential for FN accumulation in atheroprone regions of the aortic arch. In vitro, small interfering RNA against PECAM-1 blocked the induction of FN and the activation of nuclear factor (NF)-kappaB by atheroprone flow, which was rescued by the addition of exogenous FN. Additionally, blocking NF-kappaB activation attenuated the flow-induced FN expression. Small interfering RNA against FN significantly reduced NF-kappaB activity, which was rescued by the addition of exogenous FN. CONCLUSIONS: These results indicate that FN gene expression and assembly into matrix fibrils is induced by atheroprone fluid shear stress. This effect is mediated at least in part by the transcription factor NF-kappaB. Additionally, because FN promotes activation of NF-kappaB, atheroprone shear stress creates a positive feedback to maintain inflammation.


Asunto(s)
Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Fibronectinas/metabolismo , Hemodinámica , Inflamación/metabolismo , Mecanotransducción Celular , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Retroalimentación Fisiológica , Fibronectinas/genética , Humanos , Inflamación/genética , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Flujo Pulsátil , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Factores de Tiempo , Transfección , Regulación hacia Arriba
5.
Arterioscler Thromb Vasc Biol ; 31(7): 1625-33, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21527747

RESUMEN

OBJECTIVE: The goal of this study was to assess the activity of ß-catenin/T-cell-specific transcription factor (TCF) signaling in atherosclerosis development and its regulation of fibronectin in vascular endothelium. METHODS AND RESULTS: Histological staining identified preferential nuclear localization of ß-catenin in the endothelium of atheroprone aorta before and during lesion development. Transgenic reporter studies revealed that increased levels of TCF transcriptional activity in endothelium correlated anatomically with ß-catenin nuclear localization and fibronectin deposition. Exposure of endothelial cells to human-derived atheroprone shear stress induced nuclear localization of ß-catenin, transcriptional activation of TCF, and expression of fibronectin. Activation of fibronectin expression required ß-catenin, TCF, and the transcriptional coactivator CRBP-binding protein. Finally, we identified platelet endothelial cell adhesion molecule-1 as a critical regulator of constitutive ß-catenin and glycogen synthase kinase-3ß activities. CONCLUSIONS: These data reveal novel constitutive activation of the endothelial ß-catenin/TCF signaling pathway in atherosclerosis and regulation of fibronectin through hemodynamic shear stress.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Fibronectinas/metabolismo , Hemodinámica , Inflamación/metabolismo , Transducción de Señal , Factores de Transcripción TCF/metabolismo , beta Catenina/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Núcleo Celular/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Interferencia de ARN , Estrés Mecánico , Factores de Transcripción TCF/genética , Factores de Tiempo , Activación Transcripcional , Transfección , beta Catenina/genética
6.
Dev Cell ; 10(1): 11-20, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16399074

RESUMEN

Essentially all organisms from bacteria to humans are mechanosensitive. Physical forces regulate a large array of physiological processes, and dysregulation of mechanical responses contributes to major human diseases. A survey of both specialized and widely expressed mechanosensitive systems suggests that physical forces provide a general means of altering protein conformation to generate signals. Specialized systems differ mainly in having acquired efficient mechanisms for transferring forces to the mechanotransducers.


Asunto(s)
Adaptación Fisiológica/fisiología , Mecanotransducción Celular/fisiología , Transducción de Señal/fisiología , Animales , Ciclo Celular/fisiología , Humanos , Hipertensión/patología , Hipertensión/fisiopatología , Pulmón/fisiología , Modelos Biológicos , Músculo Liso Vascular/fisiopatología , Miocardio , Neoplasias/fisiopatología , Estimulación Física , Estructura Terciaria de Proteína/fisiología , Estrés Mecánico
7.
Magn Reson Med ; 66(5): 1382-90, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21656547

RESUMEN

Atherosclerosis is a complex disease whose spatial distribution is hypothesized to be influenced by the local hemodynamic environment. The use of transgenic mice provides a mechanism to study the relationship between hemodynamic forces, most notably wall shear stress (WSS), and the molecular factors that influence the disease process. Phase contrast MRI using rectilinear trajectories has been used to measure boundary conditions for use in computational fluid dynamic models. However, the unique flow environment of the mouse precludes use of standard imaging techniques in complex, curved flow regions such as the aortic arch. In this article, two-dimensional and three-dimensional spiral cine phase contrast sequences are presented that enable measurement of velocity profiles in curved regions of the mouse vasculature. WSS is calculated directly from the spatial velocity gradient, enabling WSS calculation with a minimal set of assumptions. In contrast to the outer radius of the aortic arch, the inner radius has a lower time-averaged longitudinal WSS (7.06 ± 0.76 dyne/cm(2) vs. 18.86 ± 1.27 dyne/cm(2) ; P < 0.01) and higher oscillatory shear index (0.14 ± 0.01 vs. 0.08 ± 0.01; P < 0.01). This finding is in agreement with humans, where WSS is lower and more oscillatory along the inner radius, an atheroprone region, than the outer radius, an atheroprotective region.


Asunto(s)
Aorta Torácica/anatomía & histología , Imagen por Resonancia Cinemagnética/métodos , Animales , Humanos , Aumento de la Imagen/métodos , Imagenología Tridimensional/métodos , Matemática , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Resistencia al Corte
8.
J Vasc Res ; 47(2): 168-80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19851078

RESUMEN

Vascular smooth muscle cell (SMC) phenotypic modulation plays a key role in atherosclerosis and is classically defined as a switch from a 'contractile' phenotype to a 'synthetic' phenotype, whereby genes that define the contractile SMC phenotype are suppressed and proliferation and/or migratory mechanisms are induced. There is also evidence that SMCs may take on a 'proinflammatory' phenotype, whereby SMCs secrete cytokines and express cell adhesion molecules, e.g. IL-8, IL-6, and VCAM-1, respectively, which may functionally regulate monocyte and macrophage adhesion and other processes during atherosclerosis. Factors that drive the inflammatory phenotype are not limited to cytokines but also include hemodynamic forces imposed on the blood vessel wall and intimate interaction of endothelial cells with SMCs, as well as changes in matrix composition in the vessel wall. However, it is critical to recognize that our understanding of the complex interaction of these multiple signal inputs has only recently begun to shed light on mechanisms that regulate the inflammatory SMC phenotype, primarily through models that attempt to recreate this environment ex vivo. The goal of this review is to summarize our current knowledge in this area and identify some of the key unresolved challenges and questions requiring further study.


Asunto(s)
Aterosclerosis/inmunología , Inflamación/inmunología , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Hemodinámica , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , Fenotipo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Estrés Mecánico , Transcripción Genética
9.
Circ Res ; 103(6): 671-9, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18669917

RESUMEN

Disturbed blood flow induces inflammatory gene expression in endothelial cells, which promotes atherosclerosis. Flow stimulates the proinflammatory transcription factor nuclear factor (NF)-kappaB through integrin- and Rac-dependent production of reactive oxygen species (ROS). Previous work demonstrated that NF-kappaB activation by flow is matrix-specific, occurring in cells on fibronectin but not collagen. Activation of p21-activated kinase (PAK) followed the same matrix-dependent pattern. We now show that inhibiting PAK in cells on fibronectin blocked NF-kappaB activation by both laminar and oscillatory flow in vitro and at sites of disturbed flow in vivo. Constitutively active PAK rescued flow-induced NF-kappaB activation in cells on collagen. Surprisingly, PAK was not required for flow-induced ROS production. Instead, PAK modulated the ability of ROS to activate the NF-kappaB pathway. These data demonstrate that PAK controls NF-kappaB activation by modulating the sensitivity of cells to ROS.


Asunto(s)
FN-kappa B/metabolismo , Oxidantes/fisiología , Transducción de Señal/fisiología , Quinasas p21 Activadas/fisiología , Animales , Velocidad del Flujo Sanguíneo/fisiología , Bovinos , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Resistencia al Corte
10.
Arterioscler Thromb Vasc Biol ; 29(5): 725-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19229069

RESUMEN

OBJECTIVE: Interleukin-8 (IL-8) is a soluble human-specific chemokine implicated in the development of the chronic inflammatory disease atherosclerosis. Recently, we showed that atheroprone hemodynamics induced IL-8 secretion from endothelial cells (ECs) concurrent with increased EC/smooth muscle cell (SMC) VCAM-1 expression in a human hemodynamic coculture model. Despite an IL-8 association with inflammation, we show here that blocking IL-8 activity during atheroprone flow resulted in increased levels of EC/SMC VCAM-1 expression. We tested the hypothesis that IL-8 limits SMC VCAM-1 expression in response to inflammatory stimuli, either atheroprone flow or cytokine interleukin-1beta (IL-1beta) addition. METHODS AND RESULTS: Atheroprone flow increased monocyte adhesion in both EC/SMCs, concurrent with the induction of VCAM-1 protein. VCAM-1 antisera attenuated this response. IL-1beta upregulated VCAM-1 in SMCs by 3-fold, a response inhibited by the addition of IL-8 at 24 hours. Neither IL-1beta nor IL-8 induced proliferation or migration. Neutralization of the IL-8 receptor, CXCR2, further induced VCAM-1 in the presence of IL-1beta, and phospho-p38 was required for NF-kappaB activation and VCAM-1 expression. Additionally, IL-8 reduced p38 activation and NF-kappaB activity induced by IL-1beta alone. CONCLUSIONS: Together, these findings provide evidence for a novel role whereby IL-8 limits the inflammatory response in ECs/SMCs via VCAM-1 modulation.


Asunto(s)
Aterosclerosis/fisiopatología , Células Endoteliales/metabolismo , Interleucina-8/fisiología , Miocitos del Músculo Liso/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Humanos , Túnica Íntima/metabolismo , Venas Umbilicales/citología
11.
Am J Pathol ; 173(4): 1220-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18772338

RESUMEN

Endothelial cells acquire distinctive molecular signatures in their transformation to an angiogenic phenotype that are indicative of changes in cell behavior and function. Using a rat mesentery model of inflammation-induced angiogenesis and a panel of known endothelial markers (CD31, VE-cadherin, BS-I lectin), we identified a capillary sprout-specific endothelial phenotype that is characterized by the marked down-regulation of CD36, a receptor for the anti-angiogenic molecule thrombospondin-1 (TSP-1). TSP-1/CD36 interactions were shown to regulate angiogenesis in this model as application of TSP-1 inhibited angiogenesis and blockade of both TSP-1 and CD36 accelerated angiogenesis. Vascular endothelial growth factor, which was up-regulated in the in vivo model, elicited a dose- and time-dependent down-regulation of CD36 (ie, to a CD36 low phenotype) in cultured human umbilical vein endothelial cells. Human umbilical vein endothelial cells that had been conditioned to a CD36 low phenotype with VEGF were found to be refractory to anti-proliferative TSP-1 signaling via a CD36-dependent mechanism. The loss of exposure to wall shear stress, which occurs in vivo when previously quiescent cells begin to sprout, also generated a CD36 low phenotype. Ultimately, our results identified the regulation of endothelial cell CD36 expression as a novel mechanism through which VEGF stimulates and sustains capillary sprouting in the presence of TSP-1. Additionally, CD36 was shown to function as a potential molecular linkage through which wall shear stress may regulate both microvessel sprouting and quiescence.


Asunto(s)
Antígenos CD36/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Transducción de Señal , Trombospondina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Inflamación , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fenotipo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estrés Mecánico , Trombospondina 1/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
J Muscle Res Cell Motil ; 30(1-2): 41-55, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19205907

RESUMEN

Lipoma preferred partner (LPP) localizes to focal adhesions/dense bodies, is selectively expressed in smooth muscle cells (SMC) and enhances cell migration. SMCs cultured on denatured collagen or on a rigid substrate, up regulated expression of LPP, its partner palladin, tenascin C (TN-C), phosphorylated focal adhesion kinase (pFAK) and exhibited robust stress fibers. In an endothelial (EC)/SMC hemodynamic flow system, shear stress waveforms mimicking atheroprone flow, applied to the EC layer, significantly decreased expression of SMC LPP and palladin. They were also down regulated with TN-C, in an ApoE murine model of atherosclerosis and with oxidative stress but up regulated in an arterial injury model in response to upstream sequential changes in pFAK, Prx1 and TN-C. In conclusion, expression of LPP and palladin are modulated by a mix of mechanical cues, oxidative stress and substrate composition which translate into their up or down regulation in vessel wall injury and early atherogenesis.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas del Citoesqueleto/biosíntesis , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Fosfoproteínas/biosíntesis , Resistencia al Corte , Animales , Aorta/lesiones , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/genética , Adhesión Celular , Movimiento Celular , Colágeno/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Proteínas con Dominio LIM , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Fosfoproteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Fibras de Estrés/metabolismo , Tenascina/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 28(8): 1534-41, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18556570

RESUMEN

OBJECTIVE: The initiation of atherosclerosis is in part dependent on the hemodynamic shear stress environment promoting a proinflammatory phenotype of the endothelium. Previous studies demonstrated increased expression of ER stress protein and unfolded protein response (UPR) regulator, GRP78, within all vascular cells in atherosclerotic lesions and its regulation in the endothelium by several atherosclerotic stressors; however, regulation of GRP78 by shear stress directly has not been established. METHOD AND RESULTS: Using an in vitro model to simulate human arterial shear stress waveforms, atheroprone or atheroprotective flow was applied to human endothelial cells. GRP78 was found to be significantly upregulated (3-fold) in a sustained manner under atheroprone, but not atheroprotective flow up to 24 hours. This response was dependent on both sustained activation of p38, as well integrin alpha2beta1. Increased GRP78 correlated with the activation of the ER stress sensing element (ERSE1) promoter by atheroprone flow as a marker of the UPR. Shear stress regulated GRP78 through increased protein stability when compared to other flow regulated proteins, such as connexin-43 and vascular cell adhesion molecule (VCAM)-1. Increased endothelial expression of GRP78 was also observed in atheroprone versus atheroprotective regions of C57BL6 mice. CONCLUSIONS: This study supports a role of the hemodynamic environment in preferentially inducing GRP78 and the UPR in atheroprone regions, before lesion development, and suggests a potential atheroprotective (ie, prosurvival), compensatory effect in response to ER stress within atherosclerotic lesions.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Velocidad del Flujo Sanguíneo/fisiología , Chaperón BiP del Retículo Endoplásmico , Hemorreología , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Transducción de Señal , Venas Umbilicales/citología , Venas Umbilicales/fisiología , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
14.
Arterioscler Thromb Vasc Biol ; 28(11): 2003-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18688018

RESUMEN

OBJECTIVE: Platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) has recently been shown to form an essential element of a mechanosensory complex that mediates endothelial responses to fluid shear stress. The aim of this study was to determine the in vivo role of PECAM-1 in atherosclerosis. METHODS AND RESULTS: We crossed C57BL/6 Pecam1(-/-) mice with apolipoprotein E-deficient (Apoe(-/-)) mice. On a Western diet, Pecam1(-/-)Apoe(-/-) mice showed reduced atherosclerotic lesion size compared to Apoe(-/-) mice. Striking differences were observed in the lesser curvature of the aortic arch, an area of disturbed flow, but not in the descending thoracic or abdominal aorta. Vascular cell adhesion molecule-1 (VCAM-1) expression, macrophage infiltration, and endothelial nuclear NF-kappaB were all reduced in Pecam1(-/-)Apoe(-/-) mice. Bone marrow transplantation suggested that endothelial PECAM-1 is the main determinant of atherosclerosis in the aortic arch, but that hematopoietic PECAM-1 promotes lesions in the abdominal aorta. In vitro data show that siRNA-based knockdown of PECAM-1 attenuates endothelial NF-kappaB activity and VCAM-1 expression under conditions of atheroprone flow. CONCLUSIONS: These results indicate that endothelial PECAM-1 contributes to atherosclerotic lesion formation in regions of disturbed flow by regulating NF-kappaB-mediated gene expression.


Asunto(s)
Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aorta Torácica/metabolismo , Aorta Torácica/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Grasas de la Dieta , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/patología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Flujo Sanguíneo Regional , Estrés Mecánico , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Stem Cells Transl Med ; 6(8): 1673-1683, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28628273

RESUMEN

Human induced pluripotent stem cells (iPSCs) can be differentiated into vascular endothelial (iEC) and smooth muscle (iSMC) cells. However, because iECs and iSMCs are not derived from an intact blood vessel, they represent an immature phenotype. Hemodynamics and heterotypic cell:cell communication play important roles in vascular cell phenotypic modulation. Here we tested the hypothesis that hemodynamic exposure of iECs in coculture with iSMCs induces an in vivo-like phenotype. iECs and iSMCs were cocultured under vascular region-specific blood flow hemodynamics, and compared to hemodynamic cocultures of blood vessel-derived endothelial (pEC) and smooth muscle (pSMC) cells. Hemodynamic flow-induced gene expression positively correlated between pECs and iECs as well as pSMCs and iSMCs. While endothelial nitric oxide synthase 3 protein was lower in iECs than pECs, iECs were functionally mature as seen by acetylated-low-density lipoprotein (LDL) uptake. SMC contractile protein markers were also positively correlated between pSMCs and iSMCs. Exposure of iECs and pECs to atheroprone hemodynamics with oxidized-LDL induced an inflammatory response in both. Dysfunction of the transforming growth factor ß (TGFß) pathway is seen in several vascular diseases, and iECs and iSMCs exhibited a transcriptomic prolife similar to pECs and pSMCs, respectively, in their responses to LY2109761-mediated transforming growth factor ß receptor I/II (TGFßRI/II) inhibition. Although there are differences between ECs and SMCs derived from iPSCs versus blood vessels, hemodynamic coculture restores a high degree of similarity in their responses to pathological stimuli associated with vascular diseases. Thus, iPSC-derived vascular cells exposed to hemodynamics may provide a viable system for modeling rare vascular diseases and testing new therapeutic approaches. Stem Cells Translational Medicine 2017;6:1673-1683.


Asunto(s)
Diferenciación Celular , Células Endoteliales/citología , Hemodinámica , Células Madre Pluripotentes Inducidas/citología , Miocitos del Músculo Liso/citología , Fenotipo , Transcriptoma , Células Cultivadas , Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Tissue Eng ; 12(7): 1903-13, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16889520

RESUMEN

Engineering of implantable tissues requires rapid induction of angiogenesis to meet the significant oxygen and nutrient demands of cells during tissue repair. To this end, our laboratories have utilized medicinal chemistry to synthesize non-peptide-based inducers of angiogenesis to aid tissue engineering. In this study, we describe the evaluation of SC-3-149, a small molecule compound with proliferative effects on vascular endothelial cells. Specifically, exogenous exposure of SC-3-149 induced an 18-fold increase in proliferation of human microvascular endothelial cells in vitro at low micromolar potency by day 14 in culture. Moreover, SC-3-149 significantly increased the formation of endothelial cord and tubelike structures in vitro, and improved endothelial scratch wound healing within 24 h. SC-3-149 also significantly inhibited vascular endothelial cell death owing to serum deprivation and high acidity (pH 6). Concurrent incubation of SC-3-149 with vascular endothelial growth factor increased cell survivability under serum-deprived conditions by an additional 7%. In addition, in vivo injection of SC-3-149 into the rat mesentery produced qualitative increases in microvessel length density. Taken together, our studies suggest that SC-3-149 and its analogs may serve as promising new angiogenic agents for targeted drug delivery and therapeutic angiogenesis in tissue engineering.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Proliferación Celular/efectos de los fármacos , Células Endoteliales/fisiología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Indoles/síntesis química , Neovascularización Fisiológica/efectos de los fármacos , Inductores de la Angiogénesis/síntesis química , Animales , Bioprótesis , Células Cultivadas , Células Endoteliales/citología , Femenino , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Humanos , Indoles/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Ingeniería de Tejidos , Cicatrización de Heridas/efectos de los fármacos
17.
Thromb Res ; 143: 34-9, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27179130

RESUMEN

An experimental in vitro model of the hemodynamics that occur in atrial fibrillation (AFib) in the left atrial appendage (LAA) was developed to study changes in human endothelial cell thrombotic potential. We applied human-derived sinus rhythm and AFib hemodynamic shear stress patterns to primary human endothelial cells (ECs) in culture. We found that ECs exposed to AFib hemodynamics have increased thrombotic potential as measured by increased expression of pro-thrombotic gene markers and fibrin deposition on the endothelium. Treatment with the factor Xa inhibitor, apixaban, attenuated fibrin deposition thickness while increasing fibrin density at the endothelial cell surface. This study suggests that altered hemodynamics associated with AFib play a key role in driving the thrombotic potential of the LAA endothelium.


Asunto(s)
Apéndice Atrial/patología , Fibrilación Atrial/sangre , Fibrilación Atrial/complicaciones , Células Endoteliales/patología , Hemodinámica , Trombosis/sangre , Trombosis/etiología , Fibrilación Atrial/patología , Células Cultivadas , Fibrina/análisis , Humanos , Trombosis/patología
18.
JCI Insight ; 1(20): e90954, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27942596

RESUMEN

A barrier to drug development for nonalcoholic steatohepatitis (NASH) is the absence of translational preclinical human-relevant systems. An in vitro liver model was engineered to incorporate hepatic sinusoidal flow, transport, and lipotoxic stress risk factors (glucose, insulin, free fatty acids) with cocultured primary human hepatocytes, hepatic stellate cells (HSCs), and macrophages. Transcriptomic, lipidomic, and functional endpoints were evaluated and compared with clinical data from NASH patient biopsies. The lipotoxic milieu promoted hepatocyte lipid accumulation (4-fold increase, P < 0.01) and a lipidomics signature similar to NASH biopsies. Hepatocyte glucose output increased with decreased insulin sensitivity. These changes were accompanied by increased inflammatory analyte secretion (e.g., IL-6, IL-8, alanine aminotransferase). Fibrogenic activation markers increased with lipotoxic conditions, including secreted TGF-ß (>5-fold increase, P < 0.05), extracellular matrix gene expression, and HSC activation. Significant pathway correlation existed between this in vitro model and human biopsies. Consistent with clinical trial data, 0.5 µM obeticholic acid in this model promoted a healthy lipidomic signature, reduced inflammatory and fibrotic secreted factors, but also increased ApoB secretion, suggesting a potential adverse effect on lipoprotein metabolism. Lipotoxic stress activates similar biological signatures observed in NASH patients in this system, which may be relevant for interrogating novel therapeutic approaches to treat NASH.


Asunto(s)
Técnicas de Cocultivo , Células Estrelladas Hepáticas/citología , Hepatocitos/citología , Macrófagos/citología , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Animales , Glucosa/metabolismo , Humanos , Inflamación , Resistencia a la Insulina , Lípidos/análisis , Hígado , Metaboloma , Ratones Endogámicos C57BL , Modelos Biológicos , Transcriptoma
19.
Chem Biol Interact ; 255: 31-44, 2016 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26626330

RESUMEN

Drug induced liver injury (DILI), a major cause of pre- and post-approval failure, is challenging to predict pre-clinically due to varied underlying direct and indirect mechanisms. Nevirapine, a non-nucleoside reverse transcriptase inhibitor (NNRTI) and Ritonavir, a protease inhibitor, are antiviral drugs that cause clinical DILI with different phenotypes via different mechanisms. Assessing DILI in vitro in hepatocyte cultures typically requires drug exposures significantly higher than clinical plasma Cmax concentrations, making clinical interpretations of mechanistic pathway changes challenging. We previously described a system that uses liver-derived hemodynamic blood flow and transport parameters to restore primary human hepatocyte biology, and drug responses at concentrations relevant to in vivo or clinical exposure levels. Using this system, primary hepatocytes from 5 human donors were exposed to concentrations approximating clinical therapeutic and supra-therapeutic levels of Nevirapine (11.3 and 175.0 µM) and Ritonavir (3.5 and 62.4 µM) for 48 h. Whole genome transcriptomics was performed by RNAseq along with functional assays for metabolic activity and function. We observed effects at both doses, but a greater number of genes were differentially expressed with higher probability at the toxic concentrations. At the toxic doses, both drugs showed direct cholestatic potential with Nevirapine increasing bile synthesis and Ritonavir inhibiting bile acid transport. Clear differences in antigen presentation were noted, with marked activation of MHC Class I by Nevirapine and suppression by Ritonavir. This suggests CD8+ T cell involvement for Nevirapine and possibly NK Killer cells for Ritonavir. Both compounds induced several drug metabolizing genes (including CYP2B6, CYP3A4 and UGT1A1), mediated by CAR activation in Nevirapine and PXR in Ritonavir. Unlike Ritonavir, Nevirapine did not increase fatty acid synthesis or activate the respiratory electron chain with simultaneous mitochondrial uncoupling supporting clinical reports of a lower propensity for steatosis. This in vitro study offers insights into the disparate direct and immune-mediated toxicity mechanisms underlying Nevirapine and Ritonavir toxicity in the clinic.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Hepatocitos/efectos de los fármacos , Nevirapina/toxicidad , Ritonavir/toxicidad , Transcriptoma , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Evaluación Preclínica de Medicamentos/métodos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología
20.
Nat Commun ; 4: 1525, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23443553

RESUMEN

Haemodynamic variations are inherent to blood vessel geometries (such as bifurcations) and correlate with regional development of inflammation and atherosclerosis. However, the complex frequency spectrum characteristics from these haemodynamics have never been exploited to test whether frequency variations are critical determinants of endothelial inflammatory phenotype. Here we utilize an experimental Fourier transform analysis to systematically manipulate individual frequency harmonics from human carotid shear stress waveforms applied in vitro to human endothelial cells. The frequency spectrum, specifically the 0 th and 1st harmonics, is a significant regulator of inflammation, including NF-κB activity and downstream inflammatory phenotype. Further, a harmonic-based regression-model predicts eccentric NF-κB activity observed in the human internal carotid artery. Finally, short interfering RNA-knockdown of the mechanosensor PECAM-1 reverses frequency-dependent regulation of NF-κB activity. Thus, PECAM-1 may have a critical role in the endothelium's exquisite sensitivity to complex shear stress frequency harmonics and provide a mechanism for the focal development of vascular inflammation.


Asunto(s)
Células Endoteliales/patología , Hemodinámica/fisiología , Inflamación/patología , Inflamación/fisiopatología , Velocidad del Flujo Sanguíneo , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Circulación Coronaria , Células Endoteliales/metabolismo , Análisis de Fourier , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Modelos Cardiovasculares , Mutación/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA