Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 8(35): eabo7958, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36044575

RESUMEN

Endothelial cell (EC) sensing of disturbed blood flow triggers atherosclerosis, a disease of arteries that causes heart attack and stroke, through poorly defined mechanisms. The Notch pathway plays a central role in blood vessel growth and homeostasis, but its potential role in sensing of disturbed flow has not been previously studied. Here, we show using porcine and murine arteries and cultured human coronary artery EC that disturbed flow activates the JAG1-NOTCH4 signaling pathway. Light-sheet imaging revealed enrichment of JAG1 and NOTCH4 in EC of atherosclerotic plaques, and EC-specific genetic deletion of Jag1 (Jag1ECKO) demonstrated that Jag1 promotes atherosclerosis at sites of disturbed flow. Mechanistically, single-cell RNA sequencing in Jag1ECKO mice demonstrated that Jag1 suppresses subsets of ECs that proliferate and migrate. We conclude that JAG1-NOTCH4 sensing of disturbed flow enhances atherosclerosis susceptibility by regulating EC heterogeneity and that therapeutic targeting of this pathway may treat atherosclerosis.


Asunto(s)
Aterosclerosis , Proteína Jagged-1 , Placa Aterosclerótica , Receptor Notch4 , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Ratones , Placa Aterosclerótica/metabolismo , Receptor Notch4/genética , Receptor Notch4/metabolismo , Transducción de Señal , Porcinos
2.
Int J Mol Sci ; 23(11)2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35682592

RESUMEN

(1) Background: Systemic infection is associated with increased neuroinflammation and accelerated cognitive decline in AD patients. Activated neutrophils produce neutrophil-derived microvesicles (NMV), which are internalised by human brain microvascular endothelial cells and increase their permeability in vitro, suggesting that NMV play a role in blood-brain barrier (BBB) integrity during infection. The current study investigated whether microRNA content of NMV from AD patients is significantly different compared to healthy controls and could impact cerebrovascular integrity. (2) Methods: Neutrophils isolated from peripheral blood samples of five AD and five healthy control donors without systemic infection were stimulated to produce NMV. MicroRNAs isolated from NMV were analysed by RNA-Seq, and online bioinformatic tools were used to identify significantly differentially expressed microRNAs in the NMV. Target and pathway analyses were performed to predict the impact of the candidate microRNAs on vascular integrity. (3) Results: There was no significant difference in either the number of neutrophils (p = 0.309) or the number of NMV (p = 0.3434) isolated from AD donors compared to control. However, 158 microRNAs were significantly dysregulated in AD NMV compared to controls, some of which were associated with BBB dysfunction, including miR-210, miR-20b-5p and miR-126-5p. Pathway analysis revealed numerous significantly affected pathways involved in regulating vascular integrity, including the TGFß and PDGFB pathways, as well as Hippo, IL-2 and DNA damage signalling. (4) Conclusions: NMV from AD patients contain miRNAs that may alter the integrity of the BBB and represent a novel neutrophil-mediated mechanism for BBB dysfunction in AD and the accelerated cognitive decline seen as a result of a systemic infection.


Asunto(s)
Enfermedad de Alzheimer , MicroARNs , Enfermedad de Alzheimer/metabolismo , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Humanos , MicroARNs/metabolismo , Neutrófilos/metabolismo , RNA-Seq
3.
Am J Respir Crit Care Med ; 205(8): 903-916, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35044899

RESUMEN

Rationale: Patients with chronic obstructive pulmonary disease (COPD) experience excess cardiovascular morbidity and mortality, and exacerbations further increase the risk of such events. COPD is associated with persistent blood and airway neutrophilia and systemic and tissue hypoxia. Hypoxia augments neutrophil elastase release, enhancing capacity for tissue injury. Objective: To determine whether hypoxia-driven neutrophil protein secretion contributes to endothelial damage in COPD. Methods: The healthy human neutrophil secretome generated under normoxic or hypoxic conditions was characterized by quantitative mass spectrometry, and the capacity for neutrophil-mediated endothelial damage was assessed. Histotoxic protein concentrations were measured in normoxic versus hypoxic neutrophil supernatants and plasma from patients experiencing COPD exacerbation and healthy control subjects. Measurements and Main Results: Hypoxia promoted PI3Kγ-dependent neutrophil elastase secretion, with greater release seen in neutrophils from patients with COPD. Supernatants from neutrophils incubated under hypoxia caused pulmonary endothelial cell damage, and identical supernatants from COPD neutrophils increased neutrophil adherence to endothelial cells. Proteomics revealed differential neutrophil protein secretion under hypoxia and normoxia, and hypoxia augmented secretion of a subset of histotoxic granule and cytosolic proteins, with significantly greater release seen in COPD neutrophils. The plasma of patients with COPD had higher content of hypoxia-upregulated neutrophil-derived proteins and protease activity, and vascular injury markers. Conclusions: Hypoxia drives a destructive "hypersecretory" neutrophil phenotype conferring enhanced capacity for endothelial injury, with a corresponding signature of neutrophil degranulation and vascular injury identified in plasma of patients with COPD. Thus, hypoxic enhancement of neutrophil degranulation may contribute to increased cardiovascular risk in COPD. These insights may identify new therapeutic opportunities for endothelial damage in COPD.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , Lesiones del Sistema Vascular , Células Endoteliales/metabolismo , Humanos , Hipoxia/metabolismo , Elastasa de Leucocito/metabolismo , Neutrófilos/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Lesiones del Sistema Vascular/metabolismo
4.
Elife ; 102021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34633287

RESUMEN

Fibrinogen is essential for blood coagulation. The C-terminus of the fibrinogen α-chain (αC-region) is composed of an αC-domain and αC-connector. Two recombinant fibrinogen variants (α390 and α220) were produced to investigate the role of subregions in modulating clot stability and resistance to lysis. The α390 variant, truncated before the αC-domain, produced clots with a denser structure and thinner fibres. In contrast, the α220 variant, truncated at the start of the αC-connector, produced clots that were porous with short, stunted fibres and visible fibre ends. These clots were mechanically weak and susceptible to lysis. Our data demonstrate differential effects for the αC-subregions in fibrin polymerisation, clot mechanical strength, and fibrinolytic susceptibility. Furthermore, we demonstrate that the αC-subregions are key for promoting longitudinal fibre growth. Together, these findings highlight critical functions of the αC-subregions in relation to clot structure and stability, with future implications for development of novel therapeutics for thrombosis.


Asunto(s)
Coagulación Sanguínea/fisiología , Fibrinógeno/química , Fibrinógeno/metabolismo , Fibrinólisis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Animales , Células CHO , Cricetulus , Fibrina/química , Humanos , Ratones Noqueados , Proteínas Recombinantes/química
5.
Int J Biochem Cell Biol ; 141: 106097, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34655813

RESUMEN

Microvesicles are formed through shedding from the plasma membrane, a process shared by almost all human cells. Microvesicles are highly abundant and have been detected in blood, urine, cerebrospinal fluid, and saliva. They contain a library of cargo derived from their parental cell during formation, including proteases, micro-RNAs and lipids and delivery of this parental cell-derived cargo to other cells can alter target cell function and drive disease. Cell specific molecules on the surface of microvesicles, obtained during microvesicle formation, allows their parental cell to be identified and populations of microvesicles to be investigated for roles in the pathogenesis of various diseases. For instance, recent work by our group has identified a role for neutrophil microvesicles in atherosclerosis. Microvesicle profiles could in future be associated with certain diseases and act as a biomarker to allow for earlier diagnosis. This short review will discuss some of the processes central to all microvesicles before focusing on neutrophil microvesicles, their potential role in cardiovascular disease and the mechanisms that may underpin this.


Asunto(s)
Membrana Celular , Neutrófilos , Aterosclerosis , Comunicación Celular , Micropartículas Derivadas de Células
6.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34183396

RESUMEN

The onset of venous thromboembolism, including pulmonary embolism, represents a significant health burden affecting more than 1 million people annually worldwide. Current treatment options are based on anticoagulation, which is suboptimal for preventing further embolic events. In order to develop better treatments for thromboembolism, we sought to understand the structural and mechanical properties of blood clots and how this influences embolism in vivo. We developed a murine model in which fibrin γ-chain cross-linking by activated Factor XIII is eliminated (FGG3X) and applied methods to study thromboembolism at whole-body and organ levels. We show that FGG3X mice have a normal phenotype, with overall coagulation parameters and platelet aggregation and function largely unaffected, except for total inhibition of fibrin γ-chain cross-linking. Elimination of fibrin γ-chain cross-linking resulted in thrombi with reduced strength that were prone to fragmentation. Analysis of embolism in vivo using Xtreme optical imaging and light sheet microscopy demonstrated that the elimination of fibrin γ-chain cross-linking resulted in increased embolization without affecting clot size or lysis. Our findings point to a central previously unrecognized role for fibrin γ-chain cross-linking in clot stability. They also indirectly indicate mechanistic targets for the prevention of thrombosis through selective modulation of fibrin α-chain but not γ-chain cross-linking by activated Factor XIII to reduce thrombus size and burden, while maintaining clot stability and preventing embolism.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Factor XIIIa/metabolismo , Fibrinógeno/metabolismo , Embolia Pulmonar/etiología , Embolia Pulmonar/patología , Vena Cava Inferior/patología , Trombosis de la Vena/complicaciones , Animales , Coagulación Sanguínea , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Imagen Óptica , Embolia Pulmonar/sangre , Trombosis de la Vena/sangre
7.
Sci Rep ; 10(1): 3870, 2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32099026

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Nat Commun ; 11(1): 214, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924781

RESUMEN

Neutrophils are implicated in the pathogenesis of atherosclerosis but are seldom detected in atherosclerotic plaques. We investigated whether neutrophil-derived microvesicles may influence arterial pathophysiology. Here we report that levels of circulating neutrophil microvesicles are enhanced by exposure to a high fat diet, a known risk factor for atherosclerosis. Neutrophil microvesicles accumulate at disease-prone regions of arteries exposed to disturbed flow patterns, and promote vascular inflammation and atherosclerosis in a murine model. Using cultured endothelial cells exposed to disturbed flow, we demonstrate that neutrophil microvesicles promote inflammatory gene expression by delivering miR-155, enhancing NF-κB activation. Similarly, neutrophil microvesicles increase miR-155 and enhance NF-κB at disease-prone sites of disturbed flow in vivo. Enhancement of atherosclerotic plaque formation and increase in macrophage content by neutrophil microvesicles is dependent on miR-155. We conclude that neutrophils contribute to vascular inflammation and atherogenesis through delivery of microvesicles carrying miR-155 to disease-prone regions.


Asunto(s)
Aterosclerosis/metabolismo , Endotelio/metabolismo , MicroARNs/metabolismo , Neutrófilos/metabolismo , Animales , Aterosclerosis/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales , Endotelio/patología , Regulación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Ratones , Ratones Noqueados para ApoE , MicroARNs/genética , FN-kappa B/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
9.
Nat Rev Cardiol ; 17(1): 52-63, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31366922

RESUMEN

Flowing blood generates a frictional force called shear stress that has major effects on vascular function. Branches and bends of arteries are exposed to complex blood flow patterns that exert low or low oscillatory shear stress, a mechanical environment that promotes vascular dysfunction and atherosclerosis. Conversely, physiologically high shear stress is protective. Endothelial cells are critical sensors of shear stress but the mechanisms by which they decode complex shear stress environments to regulate physiological and pathophysiological responses remain incompletely understood. Several laboratories have advanced this field by integrating specialized shear-stress models with systems biology approaches, including transcriptome, methylome and proteome profiling and functional screening platforms, for unbiased identification of novel mechanosensitive signalling pathways in arteries. In this Review, we describe these studies, which reveal that shear stress regulates diverse processes and demonstrate that multiple pathways classically known to be involved in embryonic development, such as BMP-TGFß, WNT, Notch, HIF1α, TWIST1 and HOX family genes, are regulated by shear stress in arteries in adults. We propose that mechanical activation of these pathways evolved to orchestrate vascular development but also drives atherosclerosis in low shear stress regions of adult arteries.


Asunto(s)
Aterosclerosis/genética , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mecanotransducción Celular/genética , Animales , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Endotelio Vascular/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Neovascularización Fisiológica/genética , Fenotipo , Flujo Sanguíneo Regional , Factores de Riesgo , Estrés Mecánico , Remodelación Vascular/genética
10.
Cardiovasc Res ; 116(7): 1300-1310, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31504243

RESUMEN

AIMS: Atherosclerosis develops near branches and bends of arteries that are exposed to disturbed blood flow which exerts low wall shear stress (WSS). These mechanical conditions alter endothelial cells (EC) by priming them for inflammation and by inducing turnover. Homeobox (Hox) genes are developmental genes involved in the patterning of embryos along their anterior-posterior and proximal-distal axes. Here we identified Hox genes that are regulated by WSS and investigated their functions in adult arteries. METHODS AND RESULTS: EC were isolated from inner (low WSS) and outer (high WSS) regions of the porcine aorta and the expression of Hox genes was analysed by quantitative real-time PCR. Several Hox genes (HoxA10, HoxB4, HoxB7, HoxB9, HoxD8, HoxD9) were significantly enriched at the low WSS compared to the high WSS region. Similarly, studies of cultured human umbilical vein EC (HUVEC) or porcine aortic EC revealed that the expression of multiple Hox genes (HoxA10, HoxB9, HoxD8, HoxD9) was enhanced under low (4 dyn/cm2) compared to high (13 dyn/cm2) WSS conditions. Gene silencing studies identified Hox genes (HoxB9, HoxD8, HoxD9) that are positive regulators of inflammatory molecule expression in EC exposed to low WSS, and others (HoxB9, HoxB7, HoxB4) that regulated EC turnover. We subsequently focused on HoxB9 because it was strongly up-regulated by low WSS and, uniquely, was a driver of both inflammation and proliferation. At a mechanistic level, we demonstrate using cultured EC and murine models that bone morphogenic protein 4 (BMP4) is an upstream regulator of HoxB9 which elicits inflammation via induction of numerous inflammatory mediators including TNF and downstream NF-κB activation. Moreover, the BMP4-HoxB9-TNF pathway was potentiated by hypercholesterolaemic conditions. CONCLUSIONS: Low WSS induces multiple Hox genes that control the activation state and turnover of EC. Notably, low WSS activates a BMP4-HoxB9-TNF signalling pathway to initiate focal arterial inflammation, thereby demonstrating integration of the BMP and Hox systems in vascular pathophysiology.


Asunto(s)
Aorta Torácica/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/metabolismo , Placa Aterosclerótica , Animales , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Proteína Morfogenética Ósea 4/genética , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Flujo Sanguíneo Regional , Transducción de Señal , Estrés Mecánico , Sus scrofa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
11.
Int J Mol Sci ; 20(20)2019 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-31652502

RESUMEN

The blood-brain barrier (BBB), composed of brain microvascular endothelial cells (BMEC) that are tightly linked by tight junction (TJ) proteins, restricts the movement of molecules between the periphery and the central nervous system. Elevated systemic levels of neutrophils have been detected in patients with altered BBB function, but the role of neutrophils in BMEC dysfunction is unknown. Neutrophils are key players of the immune response and, when activated, produce neutrophil-derived microvesicles (NMV). NMV have been shown to impact the integrity of endothelial cells throughout the body and we hypothesize that NMV released from circulating neutrophils interact with BMEC and induce endothelial cell dysfunction. Therefore, the current study investigated the interaction of NMV with human BMEC and determined whether they altered gene expression and function in vitro. Using flow cytometry and confocal imaging, NMV were shown to be internalized by the human cerebral microvascular endothelial cell line hCMEC/D3 via a variety of energy-dependent mechanisms, including endocytosis and macropinocytosis. The internalization of NMV significantly altered the transcriptomic profile of hCMEC/D3, specifically inducing the dysregulation of genes associated with TJ, ubiquitin-mediated proteolysis and vesicular transport. Functional studies confirmed NMV significantly increased permeability and decreased the transendothelial electrical resistance (TEER) of a confluent monolayer of hCMEC/D3. These findings indicate that NMV interact with and affect gene expression of BMEC as well as impacting their integrity. We conclude that NMV may play an important role in modulating the permeability of BBB during an infection.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Neutrófilos/metabolismo , Barrera Hematoencefálica/citología , Permeabilidad Capilar , Células Cultivadas , Endocitosis , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos
12.
J Cell Sci ; 132(11)2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31076511

RESUMEN

Endothelial cell (EC) sensing of fluid shear stress direction is a critical determinant of vascular health and disease. Unidirectional flow induces EC alignment and vascular homeostasis, whereas bidirectional flow has pathophysiological effects. ECs express several mechanoreceptors that respond to flow, but the mechanism for sensing shear stress direction is poorly understood. We determined, by using in vitro flow systems and magnetic tweezers, that ß1 integrin is a key sensor of force direction because it is activated by unidirectional, but not bidirectional, shearing forces. ß1 integrin activation by unidirectional force was amplified in ECs that were pre-sheared in the same direction, indicating that alignment and ß1 integrin activity has a feedforward interaction, which is a hallmark of system stability. En face staining and EC-specific genetic deletion studies in the murine aorta revealed that ß1 integrin is activated and is essential for EC alignment at sites of unidirectional flow but is not activated at sites of bidirectional flow. In summary, ß1 integrin sensing of unidirectional force is a key mechanism for decoding blood flow mechanics to promote vascular homeostasis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Aorta/fisiología , Integrina beta1/metabolismo , Flujo Sanguíneo Regional/fisiología , Animales , Línea Celular , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Mecanorreceptores/fisiología , Ratones , Ratones Noqueados , Estrés Fisiológico/fisiología
14.
Arterioscler Thromb Vasc Biol ; 37(11): 2087-2101, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28882872

RESUMEN

OBJECTIVE: Atherosclerosis develops near branches and bends of arteries that are exposed to low shear stress (mechanical drag). These sites are characterized by excessive endothelial cell (EC) proliferation and inflammation that promote lesion initiation. The transcription factor HIF1α (hypoxia-inducible factor 1α) is canonically activated by hypoxia and has a role in plaque neovascularization. We studied the influence of shear stress on HIF1α activation and the contribution of this noncanonical pathway to lesion initiation. APPROACH AND RESULTS: Quantitative polymerase chain reaction and en face staining revealed that HIF1α was expressed preferentially at low shear stress regions of porcine and murine arteries. Low shear stress induced HIF1α in cultured EC in the presence of atmospheric oxygen. The mechanism involves the transcription factor nuclear factor-κB that induced HIF1α transcripts and induction of the deubiquitinating enzyme Cezanne that stabilized HIF1α protein. Gene silencing revealed that HIF1α enhanced proliferation and inflammatory activation in EC exposed to low shear stress via induction of glycolysis enzymes. We validated this observation by imposing low shear stress in murine carotid arteries (partial ligation) that upregulated the expression of HIF1α, glycolysis enzymes, and inflammatory genes and enhanced EC proliferation. EC-specific genetic deletion of HIF1α in hypercholesterolemic apolipoprotein E-defecient mice reduced inflammation and endothelial proliferation in partially ligated arteries, indicating that HIF1α drives inflammation and vascular dysfunction at low shear stress regions. CONCLUSIONS: Mechanical low shear stress activates HIF1α at atheroprone regions of arteries via nuclear factor-κB and Cezanne. HIF1α promotes atherosclerosis initiation at these sites by inducing excessive EC proliferation and inflammation via the induction of glycolysis enzymes.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/metabolismo , Mecanotransducción Celular , Placa Aterosclerótica , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endopeptidasas/metabolismo , Células Endoteliales/patología , Inducción Enzimática , Femenino , Predisposición Genética a la Enfermedad , Glucólisis , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ratones Noqueados , FN-kappa B/metabolismo , Oxígeno/metabolismo , Fenotipo , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Sus scrofa , Factores de Tiempo , Transfección , Ubiquitinación , Regulación hacia Arriba
15.
Sci Rep ; 7(1): 3375, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28611395

RESUMEN

Blood flow influences atherosclerosis by generating wall shear stress, which alters endothelial cell (EC) physiology. Low shear stress induces dedifferentiation of EC through a process termed endothelial-to-mesenchymal transition (EndMT). The mechanisms underlying shear stress-regulation of EndMT are uncertain. Here we investigated the role of the transcription factor Snail in low shear stress-induced EndMT. Studies of cultured EC exposed to flow revealed that low shear stress induced Snail expression. Using gene silencing it was demonstrated that Snail positively regulated the expression of EndMT markers (Slug, N-cadherin, α-SMA) in EC exposed to low shear stress. Gene silencing also revealed that Snail enhanced the permeability of endothelial monolayers to macromolecules by promoting EC proliferation and migration. En face staining of the murine aorta or carotid arteries modified with flow-altering cuffs demonstrated that Snail was expressed preferentially at low shear stress sites that are predisposed to atherosclerosis. Snail was also expressed in EC overlying atherosclerotic plaques in coronary arteries from patients with ischemic heart disease implying a role in human arterial disease. We conclude that Snail is an essential driver of EndMT under low shear stress conditions and may promote early atherogenesis by enhancing vascular permeability.


Asunto(s)
Arterias Carótidas/patología , Endotelio Vascular/patología , Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica , Placa Aterosclerótica/patología , Factores de Transcripción de la Familia Snail/metabolismo , Estrés Mecánico , Animales , Aorta/metabolismo , Aorta/patología , Arterias Carótidas/metabolismo , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Nucleares/fisiología , Placa Aterosclerótica/metabolismo , Receptor TIE-1/fisiología , Factores de Transcripción de la Familia Snail/genética , Porcinos , Proteína 1 Relacionada con Twist/fisiología
16.
Thromb Haemost ; 117(7): 1296-1316, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28569921

RESUMEN

Microvesicles are members of the family of extracellular vesicles shed from the plasma membrane of activated or apoptotic cells. Microvesicles were initially characterised by their pro-coagulant activity and described as "microparticles". There is mounting evidence revealing a role for microvesicles in intercellular communication, with particular relevance to hemostasis and vascular biology. Coupled with this, the potential of microvesicles as meaningful biomarkers is under intense investigation. This Position Paper will summarise the current knowledge on the mechanisms of formation and composition of microvesicles of endothelial, platelet, red blood cell and leukocyte origin. This paper will also review and discuss the different methods used for their analysis and quantification, will underline the potential biological roles of these vesicles with respect to vascular homeostasis and thrombosis and define important themes for future research.


Asunto(s)
Aterosclerosis/sangre , Micropartículas Derivadas de Células/fisiología , Transporte Biológico Activo , Biomarcadores/sangre , Plaquetas/patología , Plaquetas/fisiología , Comunicación Celular , Micropartículas Derivadas de Células/patología , Células Endoteliales/fisiología , Células Endoteliales/ultraestructura , Eritrocitos/patología , Eritrocitos/fisiología , Homeostasis , Humanos , Inflamación/sangre , Leucocitos/patología , Leucocitos/fisiología , Membrana Dobles de Lípidos/sangre , Neovascularización Fisiológica , Fosfatidilserinas/sangre , Trombosis/sangre , Enfermedades Vasculares/sangre
17.
Sci Rep ; 7(1): 2332, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28539655

RESUMEN

Angiopoietins are a family of growth factors that are ligands for the tyrosine kinase receptor, Tie2. Angiopoietin 1 (Ang-1) is agonistic for Tie2, plays a key role in blood vessel maturation and stability and has been shown to possess anti-inflammatory properties. However, Tie2 expression has been demonstrated on human neutrophils and the observation that neutrophils migrate in response to Ang-1 in vitro has confounded research into its exact role in inflammation as well as its potential use as a therapeutic agent. We used a mouse model of peritoneal neutrophilic inflammation to determine if Ang-1 could stimulate neutrophil migration in vivo. Tie2 expression was demonstrated on mouse neutrophils. In addition, recombinant human Ang-1 induced significant chemotaxis of isolated mouse neutrophils in a Tie2- and CD18-dependent manner. Subsequently, co-immunoprecipitation of Ang-1 and CD18 demonstrated their interaction. Intraperitoneal injection of an engineered angiopoietin-1, MAT.Ang-1, induced significant neutrophil migration into the peritoneum and a significant increase in the levels of CCL4 in peritoneal lavage fluid. Depletion of resident peritoneal macrophages prior to, or concomitant injections of an anti-CCL4 antibody with MAT.Ang-1 resulted in a significant reduction in neutrophil recruitment. These data indicate a pro-inflammatory role for Ang-1 with respect to neutrophil recruitment.


Asunto(s)
Angiopoyetina 1/genética , Antígenos CD18/genética , Quimiocina CCL4/genética , Inflamación/genética , Receptor TIE-2/genética , Angiopoyetina 1/administración & dosificación , Animales , Movimiento Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Lavado Peritoneal , Peritoneo/efectos de los fármacos , Peritoneo/metabolismo , Peritoneo/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Transducción de Señal/efectos de los fármacos
18.
Arterioscler Thromb Vasc Biol ; 37(1): 130-143, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27834691

RESUMEN

OBJECTIVE: Atherosclerosis is initiated at branches and bends of arteries exposed to disturbed blood flow that generates low shear stress. This mechanical environment promotes lesions by inducing endothelial cell (EC) apoptosis and dysfunction via mechanisms that are incompletely understood. Although transcriptome-based studies have identified multiple shear-responsive genes, most of them have an unknown function. To address this, we investigated whether zebrafish embryos can be used for functional screening of mechanosensitive genes that regulate EC apoptosis in mammalian arteries. APPROACH AND RESULTS: First, we demonstrated that flow regulates EC apoptosis in developing zebrafish vasculature. Specifically, suppression of blood flow in zebrafish embryos (by targeting cardiac troponin) enhanced that rate of EC apoptosis (≈10%) compared with controls exposed to flow (≈1%). A panel of candidate regulators of apoptosis were identified by transcriptome profiling of ECs from high and low shear stress regions of the porcine aorta. Genes that displayed the greatest differential expression and possessed 1 to 2 zebrafish orthologues were screened for the regulation of apoptosis in zebrafish vasculature exposed to flow or no-flow conditions using a knockdown approach. A phenotypic change was observed in 4 genes; p53-related protein (PERP) and programmed cell death 2-like protein functioned as positive regulators of apoptosis, whereas angiopoietin-like 4 and cadherin 13 were negative regulators. The regulation of perp, cdh13, angptl4, and pdcd2l by shear stress and the effects of perp and cdh13 on EC apoptosis were confirmed by studies of cultured EC exposed to flow. CONCLUSIONS: We conclude that a zebrafish model of flow manipulation coupled to gene knockdown can be used for functional screening of mechanosensitive genes in vascular ECs, thus providing potential therapeutic targets to prevent or treat endothelial injury at atheroprone sites.


Asunto(s)
Apoptosis , Aterosclerosis/genética , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mecanotransducción Celular/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Células Cultivadas , Embrión no Mamífero/irrigación sanguínea , Células Endoteliales/patología , Femenino , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Fenotipo , Interferencia de ARN , Flujo Sanguíneo Regional , Estrés Mecánico , Porcinos , Transcriptoma , Transfección , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
19.
Cardiovasc Res ; 112(3): 689-701, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27671802

RESUMEN

AIMS: Stent deployment causes endothelial cells (EC) denudation, which promotes in-stent restenosis and thrombosis. Thus endothelial regrowth in stented arteries is an important therapeutic goal. Stent struts modify local hemodynamics, however the effects of flow perturbation on EC injury and repair are incompletely understood. By studying the effects of stent struts on flow and EC migration, we identified an intervention that promotes endothelial repair in stented arteries. METHODS AND RESULTS: In vitro and in vivo models were developed to monitor endothelialization under flow and the influence of stent struts. A 2D parallel-plate flow chamber with 100 µm ridges arranged perpendicular to the flow was used. Live cell imaging coupled to computational fluid dynamic simulations revealed that EC migrate in the direction of flow upstream from the ridges but subsequently accumulate downstream from ridges at sites of bidirectional flow. The mechanism of EC trapping by bidirectional flow involved reduced migratory polarity associated with altered actin dynamics. Inhibition of Rho-associated protein kinase (ROCK) enhanced endothelialization of ridged surfaces by promoting migratory polarity under bidirectional flow (P < 0.01). To more closely mimic the in vivo situation, we cultured EC on the inner surface of polydimethylsiloxane tubing containing Coroflex Blue stents (65 µm struts) and monitored migration. ROCK inhibition significantly enhanced EC accumulation downstream from struts under flow (P < 0.05). We investigated the effects of ROCK inhibition on re-endothelialization in vivo using a porcine model of EC denudation and stent placement. En face staining and confocal microscopy revealed that inhibition of ROCK using fasudil (30 mg/day via osmotic minipump) significantly increased re-endothelialization of stented carotid arteries (P < 0.05). CONCLUSIONS: Stent struts delay endothelial repair by generating localized bidirectional flow which traps migrating EC. ROCK inhibitors accelerate endothelial repair of stented arteries by enhancing EC polarity and migration through regions of bidirectional flow.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Angioplastia de Balón/instrumentación , Arterias Carótidas/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Repitelización/efectos de los fármacos , Stents , Quinasas Asociadas a rho/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Animales , Arterias Carótidas/enzimología , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Células Cultivadas , Simulación por Computador , Células Endoteliales/enzimología , Células Endoteliales/patología , Hemodinámica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Hidrodinámica , Masculino , Modelos Animales , Modelos Cardiovasculares , Cadenas Ligeras de Miosina/metabolismo , Fenotipo , Diseño de Prótesis , Flujo Sanguíneo Regional , Transducción de Señal/efectos de los fármacos , Sus scrofa , Factores de Tiempo , Quinasas Asociadas a rho/metabolismo
20.
Circ Res ; 119(3): 450-62, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27245171

RESUMEN

RATIONALE: Blood flow-induced shear stress controls endothelial cell (EC) physiology during atherosclerosis via transcriptional mechanisms that are incompletely understood. The mechanosensitive transcription factor TWIST is expressed during embryogenesis, but its role in EC responses to shear stress and focal atherosclerosis is unknown. OBJECTIVE: To investigate whether TWIST regulates endothelial responses to shear stress during vascular dysfunction and atherosclerosis and compare TWIST function in vascular development and disease. METHODS AND RESULTS: The expression and function of TWIST1 was studied in EC in both developing vasculature and during the initiation of atherosclerosis. In zebrafish, twist was expressed in early embryonic vasculature where it promoted angiogenesis by inducing EC proliferation and migration. In adult porcine and murine arteries, TWIST1 was expressed preferentially at low shear stress regions as evidenced by quantitative polymerase chain reaction and en face staining. Moreover, studies of experimental murine carotid arteries and cultured EC revealed that TWIST1 was induced by low shear stress via a GATA4-dependent transcriptional mechanism. Gene silencing in cultured EC and EC-specific genetic deletion in mice demonstrated that TWIST1 promoted atherosclerosis by inducing inflammation and enhancing EC proliferation associated with vascular leakiness. CONCLUSIONS: TWIST expression promotes developmental angiogenesis by inducing EC proliferation and migration. In addition to its role in development, TWIST is expressed preferentially at low shear stress regions of adult arteries where it promotes atherosclerosis by inducing EC proliferation and inflammation. Thus, pleiotropic functions of TWIST control vascular disease and development.


Asunto(s)
Aterosclerosis/metabolismo , Velocidad del Flujo Sanguíneo/fisiología , Endotelio Vascular/metabolismo , Proteínas Nucleares/biosíntesis , Proteína 1 Relacionada con Twist/biosíntesis , Animales , Aterosclerosis/patología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Porcinos , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA