Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Cell Biol ; 84: 102213, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37531894

RESUMO

Blood vessels are lined by a single layer of endothelial cells that provide a barrier between circulating plasma and the underlying tissue. Permeability of endothelial cells is tightly regulated, and increased permeability is associated with a number of diseases including atherosclerosis. Endothelial cells are continuously exposed to mechanical forces exerted by flowing blood and are particularly sensitive to shear stress, which is a key determinant of endothelial function. Undisturbed flow promotes endothelial resilience and reduces permeability to macromolecules whereas disturbed flow promotes endothelial dysfunction and barrier disruption. This review will outline recent advances in our understanding of how disturbed and undisturbed flow regulate paracellular and transcellular permeability and will highlight potential cellular targets that could form the basis of therapies to limit the development of cardiovascular disease.


Assuntos
Células Endoteliais , Endotélio Vascular , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo
2.
J Cell Sci ; 136(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36846872

RESUMO

Multidirectional or disturbed flow promotes endothelial dysfunction and is associated with early atherogenesis. Here we investigated the role of Wnt signalling in flow-mediated endothelial dysfunction. The expression of Frizzled-4 was higher in cultured human aortic endothelial cells (ECs) exposed to disturbed flow compared to that seen for undisturbed flow, obtained using an orbital shaker. Increased expression was also detected in regions of the porcine aortic arch exposed to disturbed flow. The increased Frizzled-4 expression in cultured ECs was abrogated following knockdown of R-spondin-3. Disturbed flow also increased the nuclear localisation and activation of ß-catenin, an effect that was dependent on Frizzled-4 and R-spondin-3. Inhibition of ß-catenin using the small-molecule inhibitor iCRT5 or knockdown of Frizzled-4 or R-spondin-3 resulted in reduced expression of pro-inflammatory genes in ECs exposed to disturbed flow, as did inhibition of WNT5A signalling. Inhibition of the canonical Wnt pathway had no effect. Inhibition of ß-catenin also reduced endothelial paracellular permeability; this was associated with altered junctional and focal adhesion organisation and cytoskeletal remodelling. These data suggest the presence of an atypical Frizzled-4-ß-catenin pathway that promotes endothelial dysfunction in response to disturbed flow.


Assuntos
Células Endoteliais , beta Catenina , Animais , Humanos , beta Catenina/genética , beta Catenina/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Permeabilidade , Suínos , Via de Sinalização Wnt , Receptores Frizzled/metabolismo
3.
Biochem Biophys Res Commun ; 642: 90-96, 2023 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-36566567

RESUMO

Calcific aortic valve disease affects the aortic side of the valve, exposed to low magnitude multidirectional ("disturbed) blood flow, more than it affects the ventricular side, exposed to high magnitude uniaxial flow. Overt disease is preceded by endothelial dysfunction and inflammation. Here we investigate the potential role of the transforming growth factor-ß (TGF-ß) receptor ALK5 in this process. Although ECs are always subject to shear stress due to blood flow, and their responses to shear stress are important in healthy valve development and homeostasis, low magnitude multidirectional flow can induce pathophysiological changes. Previous work has shown ALK5 to be an important mechanosensor. ALK5 transduces mechanically sensed signals via the activation of the SMAD2/3 transcriptional modulators. However, it is currently unclear precisely how ALK5-mediated shear stress responses translate into pathological changes under conditions of chronically disturbed flow. Here, we demonstrate that ALK5 mechanosensory signalling influences flow-induced endothelial leukocyte adhesion and paracellular permeability. Low magnitude multidirectional flow resulted in downregulation of the receptor, accompanied by increased SMAD2 phosphorylation, in human umbilical vein endothelial cell (HUVEC) monolayers. These changes correlated with elevated monocyte adhesion and significantly increased transendothelial transport of an albumin-sized tracer. These effects were abolished by inhibition of ALK5 kinase activity. Analysis of ALK5 expression patterns in porcine aortic valve tissue corroborated the findings from cell-based experiments. Together, these results suggest that ALK5 has a role in shear stress-associated cardiovascular disease pathology, emphasising the importance of further mechanistic investigations and supporting it as a potential therapeutic target.


Assuntos
Proteínas Serina-Treonina Quinases , Receptores de Fatores de Crescimento Transformadores beta , Animais , Humanos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Suínos
4.
Int J Mol Sci ; 23(15)2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35897652

RESUMO

Haemodynamic wall shear stress varies from site to site within the arterial system and is thought to cause local variation in endothelial permeability to macromolecules. Our aim was to investigate mechanisms underlying the changes in paracellular permeability caused by different patterns of shear stress in long-term culture. We used the swirling well system and a substrate-binding tracer that permits visualisation of transport at the cellular level. Permeability increased in the centre of swirled wells, where flow is highly multidirectional, and decreased towards the edge, where flow is more uniaxial, compared to static controls. Overall, there was a reduction in permeability. There were also decreases in early- and late-stage apoptosis, proliferation and mitosis, and there were significant correlations between the first three and permeability when considering variation from the centre to the edge under flow. However, data from static controls did not fit the same relation, and a cell-by-cell analysis showed that <5% of uptake under shear was associated with each of these events. Nuclear translocation of NF-κB p65 increased and then decreased with the duration of applied shear, as did permeability, but the spatial correlation between them was not significant. Application of an NO synthase inhibitor abolished the overall decrease in permeability caused by chronic shear and the difference in permeability between the centre and the edge of the well. Hence, shear and paracellular permeability appear to be linked by NO synthesis and not by apoptosis, mitosis or inflammation. The effect was mediated by an increase in transport through tricellular junctions.


Assuntos
Endotélio Vascular , Mitose , Humanos , Inflamação , Permeabilidade , Estresse Mecânico
5.
Tissue Barriers ; 9(4): 1959243, 2021 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-34542010

RESUMO

Atherosclerosis is characterized by focal accumulations of lipid within the arterial wall, thought to arise from effects of hemodynamic wall shear stress (WSS) on endothelial permeability. Identifying pathways that mediate the effects of shear on permeability could therefore provide new therapeutic opportunities. Here, we consider whether the sphingosine-1-phosphate (S1P) pathway could constitute such a route. We review effects of S1P in endothelial barrier function, the influence of WSS on S1P production and signaling, the results of trials investigating S1P in experimental atherosclerosis in mice, and associations between S1P levels and cardiovascular disease in humans. Although it seems clear that S1P reduces endothelial permeability and responds to WSS, the evidence that it influences atherosclerosis is equivocal. The effects of specifically pro- and anti-atherosclerotic WSS profiles on the S1P pathway require investigation, as do influences of S1P on the vesicular pathways likely to dominate low-density lipoprotein transport across endothelium.


Assuntos
Aterosclerose , Animais , Endotélio , Hemodinâmica , Lisofosfolipídeos , Camundongos , Permeabilidade , Esfingosina/análogos & derivados
6.
Front Cell Dev Biol ; 9: 706143, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34291056

RESUMO

Elevated serum concentrations of leucine-rich α-2-glycoprotein (LRG1) have been reported in patients with inflammatory, autoimmune, and cardiovascular diseases. This study aims to investigate the role of LRG1 in endothelial activation. LRG1 in endothelial cells (ECs) of arteries and serum of patients with critical limb ischemia (CLI) was assessed by immunohistochemistry and ELISA, respectively. LRG1 expression in sheared and tumor necrosis factor-α (TNF-α)-treated ECs was analyzed. The mechanistic role of LRG1 in endothelial activation was studied in vitro. Plasma of 37-week-old Lrg1 -/- mice was used to investigate causality between LRG1 and tumor necrosis factor receptor 1 (TNFR1) shedding. LRG1 was highly expressed in ECs of stenotic but not normal arteries. LRG1 concentrations in serum of patients with CLI were elevated compared to healthy controls. LRG1 expression was shear dependent. It could be induced by TNF-α, and the induction of its expression was mediated by NF-κB activation. LRG1 inhibited TNF-α-induced activation of NF-κB signaling, expression of VCAM-1 and ICAM-1, and monocyte capture, firm adhesion, and transendothelial migration. Mechanistically, LRG1 exerted its function by causing the shedding of TNFR1 via the ALK5-SMAD2 pathway and the subsequent activation of ADAM10. Consistent with this mechanism, LRG1 and sTNFR1 concentrations were correlated in the serum of CLI patients. Causality between LRG1 and TNFR1 shedding was established by showing that Lrg1 -/- mice had lower plasma sTNFR1 concentrations than wild type mice. Our results demonstrate a novel role for LRG1 in endothelial activation and its potential therapeutic role in inflammatory diseases should be investigated further.

7.
Cell Death Dis ; 11(6): 493, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32606304

RESUMO

Increased endothelial cell (EC) apoptosis is associated with the development of atherosclerotic plaques that develop predominantly at sites exposed to disturbed flow (DF). Strategies to promote EC survival may therefore represent a novel therapeutic approach in cardiovascular disease. Nitric oxide (NO) and ß-catenin have both been shown to promote cell survival and they interact in ECs as we previously demonstrated. Here we investigated the physiological role of ß-catenin as a mediator of NO-induced cell survival in ECs. We found that ß-catenin depleted human umbilical vein ECs (HUVEC) stimulated with pharmacological activators of endothelial NO synthase (eNOS) showed a reduction in eNOS phosphorylation (Ser1177) as well as reduced intracellular cyclic guanosine monophosphate levels compared to control cells in static cultures. In addition, ß-catenin depletion abrogated the protective effects of the NO donor, S-nitroso-N-acetylpenicillamine, during TNFα- and H2O2-induced apoptosis. Using an orbital shaker to generate shear stress, we confirmed eNOS and ß-catenin interaction in HUVEC exposed to undisturbed flow and DF and showed that ß-catenin depletion reduced eNOS phosphorylation. ß-catenin depletion promoted apoptosis exclusively in HUVEC exposed to DF as did inhibition of soluble guanylate cyclase (sGC) or ß-catenin transcriptional activity. The expression of the pro-survival genes, Bcl-2 and survivin was also reduced following inhibition of ß-catenin transcriptional activity, as was the expression of eNOS. In conclusion, our data demonstrate that ß-catenin is a positive regulator of eNOS activity and cell survival in human ECs. sGC activity and ß-catenin-dependent transcription of Bcl-2, survivin, BIRC3 and eNOS are essential to maintain cell survival in ECs under DF.


Assuntos
Apoptose/genética , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Reologia , beta Catenina/metabolismo , Animais , Sobrevivência Celular/genética , GMP Cíclico/metabolismo , Regulação para Baixo/genética , Guanilato Quinases/metabolismo , Humanos , Camundongos , Óxido Nítrico/metabolismo , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Estresse Mecânico , Survivina/genética , Survivina/metabolismo , Transcrição Gênica
8.
Atherosclerosis ; 285: 170-177, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31096159

RESUMO

A striking feature of atherosclerosis is its highly non-uniform distribution within the arterial tree. This has been attributed to variation in the haemodynamic wall shear stress (WSS) experienced by endothelial cells, but the WSS characteristics that are important and the mechanisms by which they lead to disease remain subjects of intensive investigation despite decades of research. In vivo evidence suggests that multidirectional WSS is highly atherogenic. This possibility is increasingly being studied by culturing endothelial cells in wells that are swirled on an orbital shaker. The method is simple and cost effective, has high throughput and permits chronic exposure, but interpretation of the results can be difficult because the fluid mechanics are complex; hitherto, their description has largely been restricted to the engineering literature. Here we review the findings of such studies, which indicate that putatively atherogenic flow characteristics occur at the centre of the well whilst atheroprotective ones occur towards the edge, and we describe simple mathematical methods for choosing experimental variables that avoid resonance, wave breaking and uncovering of the cells. We additionally summarise a large number of studies showing that endothelium cultured at the centre of the well expresses more pro-inflammatory and fewer homeostatic genes, has higher permeability, proliferation, apoptosis and senescence, and shows more endothelial-to-mesenchymal transition than endothelium at the edge. This simple method, when correctly interpreted, has the potential to greatly increase our understanding of the homeostatic and pathogenic mechanobiology of endothelial cells and may help identify new therapeutic targets in vascular disease.


Assuntos
Fenômenos Biofísicos , Células Endoteliais/fisiologia , Aterosclerose/etiologia , Células Cultivadas , Endotélio Vascular/citologia , Humanos , Estresse Mecânico
9.
Emerg Top Life Sci ; 2(5): 701-712, 2018 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-33530666

RESUMO

Mechanical forces play an important role in regulating tissue development and homeostasis in multiple cell types including bone, joint, epithelial and vascular cells, and are also implicated in the development of diseases, e.g. osteoporosis, cardiovascular disease and osteoarthritis. Defining the mechanisms by which cells sense and respond to mechanical forces therefore has important implications for our understanding of tissue function in health and disease and may lead to the identification of targets for therapeutic intervention. Mechanoactivation of the Wnt signalling pathway was first identified in osteoblasts with a key role for ß-catenin demonstrated in loading-induced osteogenesis. Since then, mechanoregulation of the Wnt pathway has also been observed in stem cells, epithelium, chondrocytes and vascular and lymphatic endothelium. Wnt can signal through both canonical and non-canonical pathways, and evidence suggests that both can mediate responses to mechanical strain, stretch and shear stress. This review will discuss our current understanding of the activation of the Wnt pathway in response to mechanical forces.

10.
Arterioscler Thromb Vasc Biol ; 37(1): 130-143, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27834691

RESUMO

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.


Assuntos
Apoptose , Aterosclerose/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Mecanotransdução Celular/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Células Cultivadas , Embrião não Mamífero/irrigação sanguínea , Células Endoteliais/patologia , Feminino , Perfilação da Expressão Gênica/métodos , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Camundongos , Fenótipo , Interferência de RNA , Fluxo Sanguíneo Regional , Estresse Mecânico , Suínos , Transcriptoma , Transfecção , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
11.
Br J Pharmacol ; 172(14): 3548-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25824964

RESUMO

BACKGROUND AND PURPOSE: There are conflicting data regarding whether netrin-1 retards or accelerates atherosclerosis progression, as it can lead either to monocyte repulsion from or retention within plaques depending on its cellular source. We investigated the effect of aspirin, which is widely used in cardiovascular prophylaxis, on the synthesis of different isoforms of netrin-1 by endothelial cells under pro-inflammatory conditions, and defined the net effect of aspirin-dependent systemic modulation of netrin-1 on atherosclerosis progression. EXPERIMENTAL APPROACH: Netrin-1 synthesis was studied in vitro using human endothelial cells stimulated with TNF-α, with or without aspirin treatment. In vivo experiments were conducted in ApoE(-/-) mice fed with a high-fat diet (HFD), receiving either aspirin or clopidogrel. KEY RESULTS: TNF-α-induced NF-κB activation up-regulated the nuclear isoform of netrin-1, while simultaneously reducing secreted netrin-1. Down-regulation of the secreted isoform compromised the chemorepellent action of the endothelium against monocyte chemotaxis. Aspirin counteracted TNF-α-mediated effects on netrin-1 synthesis by endothelial cells through COX-dependent inhibition of NF-κB and concomitant histone hyperacetylation. Administration of aspirin to ApoE(-/-) mice on HFD increased blood and arterial wall levels of netrin-1 independently of its effects on platelets, accompanied by reduced plaque size and content of monocytes/macrophages, compared with untreated or clopidogrel-treated mice. In vivo blockade of netrin-1 enhanced monocyte plaque infiltration in aspirin-treated ApoE(-/-) mice. CONCLUSIONS AND IMPLICATIONS: Aspirin counteracts down-regulation of secreted netrin-1 induced by pro-inflammatory stimuli in endothelial cells. The aspirin-dependent increase of netrin-1 in ApoE(-/-) mice exerts anti-atherogenic effects by preventing arterial accumulation of monocytes.


Assuntos
Aspirina/farmacologia , Histonas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Fatores de Crescimento Neural/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Acetilação/efeitos dos fármacos , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aspirina/administração & dosagem , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Fatores de Crescimento Neural/biossíntese , Netrina-1 , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/biossíntese
12.
Cardiovasc Res ; 104(1): 116-26, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25062958

RESUMO

AIMS: ß-catenin has been shown to be regulated by inducible nitric oxide synthase (NOS) in endothelial cells. We investigated here whether ß-catenin interacts with and regulates endothelial NOS (eNOS) and whether eNOS activation promotes ß-catenin signalling. METHODS AND RESULTS: We identified ß-catenin as a novel eNOS binding protein in human umbilical vein endothelial cells (HUVECs) by mass spectroscopy and western blot analyses of ß-catenin and eNOS immunoprecipitates. This was confirmed by in situ proximity ligation assay. eNOS activity, assessed by cGMP production and eNOS phosphorylation (Ser1177), was enhanced in ß-catenin(-/-) mouse pulmonary endothelial cells (MPECs) relative to wild-type MPECs. eNOS activation (using adenosine, salbutamol, thrombin, or histamine), or application of an NO donor (spermine NONOate) or cGMP-analogue (8-bromo-cGMP) caused nuclear translocation of ß-catenin in HUVEC as shown by western blotting of nuclear extracts. Exposure to spermine NONOate, 8-bromo-cGMP, or sildenafil (a phosphodiesterase type 5 inhibitor) also increased the expression of ß-catenin-dependent transcripts, IL-8, and cyclin D1. Stimulation of wild-type MPECs with basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), spermine NONOate, 8-bromo-cGMP, or sildenafil increased tube length relative to controls in an angiogenesis assay. These responses were abrogated in ß-catenin(-/-) MPECs, with the exception of that to bFGF which is NO-independent. In C57BL/6 mice, subcutaneous VEGF-supplemented Matrigel plugs containing ß-catenin(-/-) MPECs exhibited reduced angiogenesis compared with plugs containing wild-type MPECs. Angiogenesis was not altered in bFGF-supplemented Matrigel. CONCLUSION: These data reveal bidirectional cross-talk and regulation between the NO-cGMP and ß-catenin signalling pathways.


Assuntos
Células Endoteliais da Veia Umbilical Humana/enzimologia , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , beta Catenina/metabolismo , Animais , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Quinase 3 da Glicogênio Sintase , Glicogênio Sintase Quinase 3 beta , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Inibidores da Fosfodiesterase 5/farmacologia , Fosforilação , Ligação Proteica , Serina , Transdução de Sinais , beta Catenina/deficiência , beta Catenina/genética
13.
Atherosclerosis ; 235(2): 613-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24956536

RESUMO

OBJECTIVE: The c-Jun N-terminal kinase (JNK) family regulates fundamental physiological processes including apoptosis and metabolism. Although JNK2 is known to promote foam cell formation during atherosclerosis, the potential role of JNK1 is uncertain. We examined the potential influence of JNK1 and its negative regulator, MAP kinase phosphatase-1 (MKP-1), on endothelial cell (EC) injury and early lesion formation using hypercholesterolemic LDLR(-/-) mice. METHODS AND RESULTS: To assess the function of JNK1 in early atherogenesis, we measured EC apoptosis and lesion formation in LDLR(-/-) or LDLR(-/-)/JNK1(-/-) mice exposed to a high fat diet for 6 weeks. En face staining using antibodies that recognise active, cleaved caspase-3 (apoptosis) or using Sudan IV (lipid deposition) revealed that genetic deletion of JNK1 reduced EC apoptosis and lesion formation in hypercholesterolemic mice. By contrast, although EC apoptosis was enhanced in LDLR(-/-)/MKP-1(-/-) mice compared to LDLR(-/-) mice, lesion formation was unaltered. CONCLUSION: We conclude that JNK1 is required for EC apoptosis and lipid deposition during early atherogenesis. Thus pharmacological inhibitors of JNK may reduce atherosclerosis by preventing EC injury as well as by influencing foam cell formation.


Assuntos
Células Endoteliais/patologia , Hipercolesterolemia/fisiopatologia , Proteína Quinase 8 Ativada por Mitógeno/deficiência , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Dieta Hiperlipídica , Fosfatase 1 de Especificidade Dupla/deficiência , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/metabolismo
14.
Arterioscler Thromb Vasc Biol ; 34(5): 985-95, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24651677

RESUMO

OBJECTIVE: Although atherosclerosis is associated with systemic risk factors such as age, high cholesterol, and obesity, plaque formation occurs predominately at branches and bends that are exposed to disturbed patterns of blood flow. The molecular mechanisms that link disturbed flow-generated mechanical forces with arterial injury are uncertain. To illuminate them, we investigated the effects of flow on endothelial cell (EC) senescence. APPROACH AND RESULTS: LDLR(-/-) (low-density lipoprotein receptor(-/-)) mice were exposed to a high-fat diet for 2 to 12 weeks (or to a normal chow diet as a control) before the assessment of cellular senescence in aortic ECs. En face staining revealed that senescence-associated ß-galactosidase activity and p53 expression were elevated in ECs at sites of disturbed flow in response to a high-fat diet. By contrast, ECs exposed to undisturbed flow did not express senescence-associated ß-galactosidase or p53. Studies of aortae from healthy pigs (aged 6 months) also revealed enhanced senescence-associated ß-galactosidase staining at sites of disturbed flow. These data suggest that senescent ECs accumulate at disturbed flow sites during atherogenesis. We used in vitro flow systems to examine whether a causal relationship exists between flow and EC senescence. Exposure of cultured ECs to flow (using either an orbital shaker or a syringe-pump flow bioreactor) revealed that disturbed flow promoted EC senescence compared with static conditions, whereas undisturbed flow reduced senescence. Gene silencing studies demonstrated that disturbed flow induced EC senescence via a p53-p21 signaling pathway. Disturbed flow-induced senescent ECs exhibited reduced migration compared with nonsenescent ECs in a scratch wound closure assay, and thus may be defective for arterial repair. However, pharmacological activation of sirtuin 1 (using resveratrol or SRT1720) protected ECs from disturbed flow-induced senescence. CONCLUSIONS: Disturbed flow promotes endothelial senescence via a p53-p21-dependent pathway which can be inhibited by activation of sirtuin 1. These observations support the principle that pharmacological activation of sirtuin 1 may promote cardiovascular health by suppressing EC senescence at atheroprone sites.


Assuntos
Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Senescência Celular , Células Endoteliais/metabolismo , Mecanotransdução Celular , Proteína Supressora de Tumor p53/metabolismo , Animais , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Reatores Biológicos , Movimento Celular , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Mecanotransdução Celular/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Fluxo Sanguíneo Regional , Sirtuína 1/metabolismo , Estresse Mecânico , Suínos , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/genética , Cicatrização
15.
Cell Mol Bioeng ; 5(2): 184-193, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33968262

RESUMO

The permeability of cultured endothelial monolayers is higher than the permeability of endothelium in vivo. Co-culture with astrocytes can induce a tight, blood-brain-barrier phenotype in aortic endothelium in vitro. We hypothesised that dendritic cells, which reside in the intima of non-cerebral arteries and have features in common with astrocytes, may also reduce the permeability of cultured aortic endothelium. The permeability of porcine aortic endothelial monolayers was reduced by non-contact co-culture with dendritic cells (but not with the peripheral blood monocytes from which they were derived) and by dendritic cell conditioned medium, indicating a soluble mediator. The reduction in permeability was similar to that obtained by co-culture with astrocytes; however, dendritic cells did not up-regulate P-glycoprotein and there was no synergy with the effect of chronic shear stress on permeability, contrary to observations with astrocytes. Endothelial permeability was reduced by sphingosine-1-phosphate, which mediates the barrier-tightening effect of platelets, but inhibitors of sphingosine-1-phosphate receptors did not block the effect of dendritic cells. Rates of endothelial mitosis and apoptosis were also unaffected by co-culture. Hence dendritic cells reduce permeability by different mechanisms from those mediating barrier-tightening effects of astrocytes and platelets, although factors mediating the permeability-lowering effects of chronic shear stress may be involved. We speculate that dendritic cells influence endothelial permeability in vivo.

16.
F1000 Med Rep ; 3: 5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21654925

RESUMO

Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids and inflammatory cells along the inner walls of arteries, and is an underlying cause of cardiovascular disease. Atherosclerotic lesions develop predominantly at branches, bends, and bifurcations in the arterial tree because these sites are exposed to low or disturbed blood flow, which exerts low/oscillatory shear stress on the vessel wall. This mechanical environment alters endothelial cell physiology by enhancing inflammatory activation. In contrast, regions of the arterial tree that are exposed to uniform, unidirectional blood flow and experience high shear stress are protected from inflammation and lesion development. Shear stress is sensed by the endothelium via mechanoreceptors and is subsequently transduced into biochemical signals resulting in modulation of proinflammatory signaling pathways. In this article, we address the molecular mechanisms behind the spatial localization of vascular inflammation and atherosclerosis, with particular focus on studies by our own group of two key proinflammatory signaling pathways, the mitogen-activated protein kinase pathway and the nuclear factor-kappa-B pathway.

17.
Arterioscler Thromb Vasc Biol ; 31(2): 384-91, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21127291

RESUMO

OBJECTIVE: The goal of this study was to examine the effect of chronic heterogeneous shear stress, applied using an orbital shaker, on endothelial cell morphology and the expression of cyclooxygenases 1 and 2. METHODS AND RESULTS: Porcine aortic endothelial cells were plated on fibronectin-coated Transwell plates. Cells were cultured for up to 7 days either under static conditions or on an orbital shaker that generated a wave of medium inducing shear stress over the cells. Cells were fixed and stained for the endothelial surface marker CD31 or cyclooxygenases 1 and 2. En face confocal microscopy and scanning ion conductance microscopy were used to show that endothelial cells were randomly oriented at the center of the well, aligned with shear stress nearer the periphery, and expressed cyclooxygenase-1 under all conditions. Lipopolysaccharide induced cyclooxygenase-2 and the production of 6-keto-prostaglandin F(1α) in all cells. CONCLUSIONS: Cyclooxygenase-1 is expressed in endothelial cells cultured under chronic shear stress of high or low directionality.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Estresse Mecânico , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Aorta Torácica/citologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Contagem de Células , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Isoenzimas/metabolismo , Lipopolissacarídeos/farmacologia , Modelos Animais , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Suínos
18.
Am J Physiol Heart Circ Physiol ; 298(6): H1850-6, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20363882

RESUMO

Endothelial properties are affected by mechanical stresses. Several studies have shown that an acute application of shear stress increases the permeability of endothelial monolayers in culture. We investigated whether more prolonged application of shear has the opposite effect. Porcine aortic endothelial cells were cultured on Transwell filters to assess monolayer permeability to albumin. The medium above the cells was swirled using an orbital shaker; resultant shears were computed to lie within the physiological range. Acute application of shear increased permeability, but chronic application reduced it. The effect of chronic but not acute shear was reversed by inhibiting nitric oxide (NO) synthesis. The effect of chronic shear was also reversed by inhibiting phosphatidylinositol 3-OH kinase (PI3K) and soluble guanylyl cyclase. None of these interventions affected permeability under static conditions, and inhibition of cyclooxygenase was without effect. Chronic shear decreased mitosis rates by a fraction comparable to the reduction in permeability, but this effect was not reversed by inhibiting NO synthesis. We conclude that chronic application of shear stress reduces endothelial permeability to macromolecules by a PI3K-NO-cGMP-dependent mechanism. Since atherosclerosis can be triggered by excessive entry of plasma macromolecules into the arterial wall, the phenomenon may help explain the atheroprotective effects of shear and NO.


Assuntos
Albuminas/farmacocinética , Permeabilidade da Membrana Celular/fisiologia , Endotélio Vascular/fisiopatologia , Substâncias Macromoleculares/farmacocinética , Estresse Mecânico , Animais , Aorta Torácica/citologia , Aorta Torácica/fisiopatologia , Proliferação de Células , Células Cultivadas , GMP Cíclico/antagonistas & inibidores , GMP Cíclico/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Modelos Animais , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Prostaglandina-Endoperóxido Sintases/metabolismo , Suínos
19.
Invest Ophthalmol Vis Sci ; 50(3): 1319-28, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18997095

RESUMO

PURPOSE: The accumulation of advanced glycation end products (AGEs) within the retina in diabetes is associated with a chronic increase in retinal microvascular permeability. Isolated perfused retinas were used to examine the acute effects of AGEs on retinal microvascular permeability. METHODS: Retinas were dissected from eyes obtained from male Wistar rats, pinned out flat, and perfused with the low-molecular-weight fluorescent dye sulforhodamine B. Microvascular permeability was determined from the rate of decrease in fluorescence gradient across a vessel under conditions of zero flow. The production of reactive oxygen species (ROS) in JG2.1 retinal endothelial cells was also assessed with a fluorescent probe working solution. RESULTS: A 30-second application of AGE-modified bovine serum albumin (AGE-BSA) to the abluminal surface of the retinal vasculature produced a rapid dose-dependent increase in retinal capillary permeability that was inhibited by pretreatment with anti-RAGE IgG. The permeability response also required ROS generated by NADPH oxidase because pretreatment with apocynin and the free radical scavengers superoxide dismutase and catalase significantly reduced the response. Pretreatment with calphostin C, SKF-96365, and U-73122 also significantly reduced the permeability response. In addition, the permeability response to bradykinin increased permeability through ROS and was potentiated after pretreatment with AGE-BSA. This potentiation was blocked by apocynin. CONCLUSIONS: Acute activation of NADPH oxidase by phospholipase C-mediated activation of Ca(2+)-dependent PKC occurs downstream of RAGE activation to acutely increase retinal capillary permeability in the isolated perfused rat retina.


Assuntos
Permeabilidade Capilar/fisiologia , NADPH Oxidases/fisiologia , Receptores Imunológicos/metabolismo , Vasos Retinianos/enzimologia , Acetofenonas/farmacologia , Animais , Bradicinina/farmacologia , Cálcio/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Catalase/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Corantes Fluorescentes/metabolismo , Produtos Finais de Glicação Avançada/farmacologia , Masculino , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptor para Produtos Finais de Glicação Avançada , Vasos Retinianos/efeitos dos fármacos , Rodaminas/metabolismo , Soroalbumina Bovina/farmacologia , Superóxido Dismutase/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...