Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Biomech ; 50: 11-19, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27916240

ABSTRACT

Despite substantial evidence for the central role of hemodynamic shear stress in the functional integrity of vascular endothelial cells, hemodynamic and molecular regulation of the endocardial endothelium lining the heart chambers remains understudied. We propose that regional differences in intracardiac hemodynamics influence differential endocardial gene expression leading to phenotypic heterogeneity of this cell layer. Measurement of intracardiac hemodynamics was performed using 4-dimensional flow MRI in healthy humans (n=8) and pigs (n=5). Local wall shear stress (WSS) and oscillatory shear indices (OSI) were calculated in three distinct regions of the LV - base, mid-ventricle (midV), and apex. In both the humans and pigs, WSS values were significantly lower in the apex and midV relative to the base. Additionally, both the apex and midV had greater oscillatory shear indices (OSI) than the base. To investigate regional phenotype, endocardial endothelial cells (EEC) were isolated from an additional 8 pigs and RNA sequencing was performed. A false discovery rate of 0.10 identified 1051 differentially expressed genes between the base and apex, and 321 between base and midV. Pathway analyses revealed apical upregulation of genes associated with translation initiation. Furthermore, tissue factor pathway inhibitor (TFPI; mean 50-fold) and prostacyclin synthase (PTGIS; 5-fold), genes prominently associated with antithrombotic protection, were consistently upregulated in LV apex. These spatio-temporal WSS values in defined regions of the left ventricle link local hemodynamics to regional heterogeneity in endocardial gene expression.


Subject(s)
Endothelial Cells/physiology , Endothelium, Vascular/physiology , Adult , Animals , Endothelium, Vascular/diagnostic imaging , Female , Heart Ventricles/diagnostic imaging , Hemodynamics , Humans , Magnetic Resonance Imaging , Male , Phenotype , Stress, Mechanical , Swine , Young Adult
2.
J Biomech ; 50: 3-10, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27865480

ABSTRACT

Epigenetics is the regulation of gene expression (transcription) in response to changes in the cell environment through genomic modifications that largely involve the non-coding fraction of the human genome and that cannot be attributed to modification of the primary DNA sequence. Epigenetics is dominant in establishing cell fate and positioning during programmed embryonic development. However the same pathways are used by mature postnatal and adult mammalian cells during normal physiology and are implicated in disease mechanisms. Recent research demonstrates that blood flow and pressure are cell environments that can influence transcription via epigenetic pathways. The principal epigenetic pathways are chemical modification of cytosine residues of DNA (DNA methylation) and of the amino tails of histone proteins associated with DNA in nucleosomes. They also encompass the post-transcriptional degradation of mRNA transcripts by non-coding RNAs (ncRNA). In vascular endothelium, epigenetic pathways respond to temporal and spatial variations of flow and pressure, particularly hemodynamic disturbed blood flow, with important consequences for gene expression. The biofluid environment is linked by mechanotransduction and solute transport to cardiovascular cell phenotypes via signaling pathways and epigenetic regulation for which there is an adequate interdisciplinary infrastructure with robust tools and methods available. Epigenetic mechanisms may be less familiar than acute genomic signaling to Investigators at the interface of biofluids, biomechanics and cardiovascular biology. Here we introduce a biofluids / cellular biomechanics readership to the principal epigenetic pathways and provide a contextual overview of endothelial epigenetic plasticity in the regulation of flow-responsive transcription.


Subject(s)
Endothelial Cells/metabolism , Epigenesis, Genetic , Gene Expression , Animals , Blood Circulation , DNA Methylation , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Humans
3.
J Am Heart Assoc ; 5(4): e003170, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27091183

ABSTRACT

BACKGROUND: Unlike arteries, in which regionally distinct hemodynamics are associated with phenotypic heterogeneity, the relationships between endocardial endothelial cell phenotype and intraventricular flow remain largely unexplored. We investigated regional differences in left ventricular wall shear stress and their association with endocardial endothelial cell gene expression. METHODS AND RESULTS: Local wall shear stress was calculated from 4-dimensional flow magnetic resonance imaging in 3 distinct regions of human (n=8) and pig (n=5) left ventricle: base, adjacent to the outflow tract; midventricle; and apex. In both species, wall shear stress values were significantly lower in the apex and midventricle relative to the base; oscillatory shear index was elevated in the apex. RNA sequencing of the endocardial endothelial cell transcriptome in pig left ventricle (n=8) at a false discovery rate ≤10% identified 1051 genes differentially expressed between the base and the apex and 327 between the base and the midventricle; no differentially expressed genes were detected at this false discovery rate between the apex and the midventricle. Enrichment analyses identified apical upregulation of genes associated with translation initiation including mammalian target of rapamycin, and eukaryotic initiation factor 2 signaling. Genes of mitochondrial dysfunction and oxidative phosphorylation were also consistently upregulated in the left ventricular apex, as were tissue factor pathway inhibitor (mean 50-fold) and prostacyclin synthase (5-fold)-genes prominently associated with antithrombotic protection. CONCLUSIONS: We report the first spatiotemporal measurements of wall shear stress within the left ventricle and linked regional hemodynamics to heterogeneity in ventricular endothelial gene expression, most notably to translation initiation and anticoagulation properties in the left ventricular apex, in which oscillatory shear index is increased and wall shear stress is decreased.


Subject(s)
Endocardium/metabolism , Heart Ventricles/metabolism , RNA/genetics , Shear Strength/physiology , Animals , Cardiac Imaging Techniques , Endocardium/diagnostic imaging , Endocardium/physiology , Female , Gene Expression Profiling , Genomic Library , Heart Ventricles/diagnostic imaging , Hemodynamics , Humans , Magnetic Resonance Imaging , Male , Swine , Ventricular Function, Left/physiology , Young Adult
4.
J Clin Invest ; 125(8): 2995-3007, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26214523

ABSTRACT

Fluid shear forces have established roles in blood vascular development and function, but whether such forces similarly influence the low-flow lymphatic system is unknown. It has been difficult to test the contribution of fluid forces in vivo because mechanical or genetic perturbations that alter flow often have direct effects on vessel growth. Here, we investigated the functional role of flow in lymphatic vessel development using mice deficient for the platelet-specific receptor C-type lectin-like receptor 2 (CLEC2) as blood backfills the lymphatic network and blocks lymph flow in these animals. CLEC2-deficient animals exhibited normal growth of the primary mesenteric lymphatic plexus but failed to form valves in these vessels or remodel them into a structured, hierarchical network. Smooth muscle cell coverage (SMC coverage) of CLEC2-deficient lymphatic vessels was both premature and excessive, a phenotype identical to that observed with loss of the lymphatic endothelial transcription factor FOXC2. In vitro evaluation of lymphatic endothelial cells (LECs) revealed that low, reversing shear stress is sufficient to induce expression of genes required for lymphatic valve development and identified GATA2 as an upstream transcriptional regulator of FOXC2 and the lymphatic valve genetic program. These studies reveal that lymph flow initiates and regulates many of the key steps in collecting lymphatic vessel maturation and development.


Subject(s)
Lymph/physiology , Lymphatic Vessels/embryology , Muscle, Smooth, Vascular/embryology , Myocytes, Smooth Muscle/metabolism , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Shear Strength
5.
BMC Genomics ; 16: 506, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26148682

ABSTRACT

BACKGROUND: Atherosclerosis is a heterogeneously distributed disease of arteries in which the endothelium plays an important central role. Spatial transcriptome profiling of endothelium in pre-lesional arteries has demonstrated differential phenotypes primed for athero-susceptibility at hemodynamic sites associated with disturbed blood flow. DNA methylation is a powerful epigenetic regulator of endothelial transcription recently associated with flow characteristics. We investigated differential DNA methylation in flow region-specific aortic endothelial cells in vivo in adult domestic male and female swine. RESULTS: Genome-wide DNA methylation was profiled in endothelial cells (EC) isolated from two robust locations of differing patho-susceptibility:--an athero-susceptible site located at the inner curvature of the aortic arch (AA) and an athero-protected region in the descending thoracic (DT) aorta. Complete methylated DNA immunoprecipitation sequencing (MeDIP-seq) identified over 5500 endothelial differentially methylated regions (DMRs). DMR density was significantly enriched in exons and 5'UTR sequences of annotated genes, 60 of which are linked to cardiovascular disease. The set of DMR-associated genes was enriched in transcriptional regulation, pattern specification HOX loci, oxidative stress and the ER stress adaptive pathway, all categories linked to athero-susceptible endothelium. Examination of the relationship between DMR and mRNA in HOXA genes demonstrated a significant inverse relationship between CpG island promoter methylation and gene expression. Methylation-specific PCR (MSP) confirmed differential CpG methylation of HOXA genes, the ER stress gene ATF4, inflammatory regulator microRNA-10a and ARHGAP25 that encodes a negative regulator of Rho GTPases involved in cytoskeleton remodeling. Gender-specific DMRs associated with ciliogenesis that may be linked to defects in cilia development were also identified in AA DMRs. CONCLUSIONS: An endothelial methylome analysis identifies epigenetic DMR characteristics associated with transcriptional regulation in regions of atherosusceptibility in swine aorta in vivo. The data represent the first methylome blueprint for spatio-temporal analyses of lesion susceptibility predisposing to endothelial dysfunction in complex flow environments in vivo.


Subject(s)
Aorta/metabolism , DNA Methylation/genetics , Endothelium, Vascular/metabolism , Transcriptome/genetics , Animals , Atherosclerosis/genetics , CpG Islands/genetics , Endothelial Cells/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Male , Phenotype , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Spatio-Temporal Analysis , Swine
6.
ACS Nano ; 9(7): 6785-93, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26153796

ABSTRACT

Nanocarriers (NCs) coated with antibodies (Abs) to extracellular epitopes of the transmembrane glycoprotein PECAM (platelet endothelial cell adhesion molecule-1/CD31) enable targeted drug delivery to vascular endothelial cells. Recent studies revealed that paired Abs directed to adjacent, yet distinct epitopes of PECAM stimulate each other's binding to endothelial cells in vitro and in vivo ("collaborative enhancement"). This phenomenon improves targeting of therapeutic fusion proteins, yet its potential role in targeting multivalent NCs has not been addressed. Herein, we studied the effects of Ab-mediated collaborative enhancement on multivalent NC spheres coated with PECAM Abs (Ab/NC, ∼180 nm diameter). We found that PECAM Abs do mutually enhance endothelial cell binding of Ab/NC coated by paired, but not "self" Ab. In vitro, collaborative enhancement of endothelial binding of Ab/NC by paired Abs is modulated by Ab/NC avidity, epitope selection, and flow. Cell fixation, but not blocking of endocytosis, obliterated collaborative enhancement of Ab/NC binding, indicating that the effect is mediated by molecular reorganization of PECAM molecules in the endothelial plasmalemma. The collaborative enhancement of Ab/NC binding was affirmed in vivo. Intravascular injection of paired Abs enhanced targeting of Ab/NC to pulmonary vasculature in mice by an order of magnitude. This stimulatory effect greatly exceeded enhancement of Ab targeting by paired Abs, indicating that '"collaborative enhancement"' effect is even more pronounced for relatively large multivalent carriers versus free Abs, likely due to more profound consequences of positive alteration of epitope accessibility. This phenomenon provides a potential paradigm for optimizing the endothelial-targeted nanocarrier delivery of therapeutic agents.


Subject(s)
Blood Platelets/metabolism , Epitopes/immunology , Nanospheres/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Cell Membrane/metabolism , Female , Human Umbilical Vein Endothelial Cells , Mice , Mice, Inbred C57BL , Nanospheres/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Protein Binding
7.
J Control Release ; 210: 39-47, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-25966362

ABSTRACT

Targeting nanocarriers (NC) to endothelial cell adhesion molecules including Platelet-Endothelial Cell Adhesion Molecule-1 (PECAM-1 or CD31) improves drug delivery and pharmacotherapy of inflammation, oxidative stress, thrombosis and ischemia in animal models. Recent studies unveiled that hydrodynamic conditions modulate endothelial endocytosis of NC targeted to PECAM-1, but the specificity and mechanism of effects of flow remain unknown. Here we studied the effect of flow on endocytosis by human endothelial cells of NC targeted by monoclonal antibodies Ab62 and Ab37 to distinct epitopes on the distal extracellular domain of PECAM. Flow in the range of 1-8dyn/cm(2), typical for venous vasculature, stimulated the uptake of spherical Ab/NC (~180nm diameter) carrying ~50 vs 200 Ab62 and Ab37 per NC, respectively. Effect of flow was inhibited by disruption of cholesterol-rich plasmalemma domains and deletion of PECAM-1 cytosolic tail. Flow stimulated endocytosis of Ab62/NC and Ab37/NC via eliciting distinct signaling pathways mediated by RhoA/ROCK and Src Family Kinases, respectively. Therefore, flow stimulates endothelial endocytosis of Ab/NC in a PECAM-1 epitope specific manner. Using ligands of binding to distinct epitopes on the same target molecule may enable fine-tuning of intracellular delivery based on the hemodynamic conditions in the vascular area of interest.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Drug Carriers/administration & dosage , Epitopes/immunology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cells, Cultured , Drug Carriers/chemistry , Endocytosis , Human Umbilical Vein Endothelial Cells , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polystyrenes/administration & dosage , Polystyrenes/chemistry , Rheology , Stress, Mechanical
8.
Arterioscler Thromb Vasc Biol ; 35(6): 1317-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25838424

ABSTRACT

Arterial endothelial phenotype is regulated by local hemodynamic forces that are linked to regional susceptibility to atherogenesis. A complex hierarchy of transcriptional, translational, and post-translational mechanisms is greatly influenced by the characteristics of local arterial shear stress environments. We discuss the emerging role of localized disturbed blood flow on epigenetic mechanisms of endothelial responses to biomechanical stress, including transcriptional regulation by proximal promoter DNA methylation, and post-transcriptional and translational regulation of gene and protein expression by chromatin remodeling and noncoding RNA-based mechanisms. Dynamic responses to flow characteristics in vivo and in vitro include site-specific differentially methylated regions of swine and mouse endothelial methylomes, histone marks regulating chromatin conformation, microRNAs, and long noncoding RNAs. Flow-mediated epigenomic responses intersect with cis and trans factor regulation to maintain endothelial function in a shear-stressed environment and may contribute to localized endothelial dysfunctions that promote atherosusceptibility.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Epigenesis, Genetic , Hemodynamics/physiology , Stress, Mechanical , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Genes, Homeobox/physiology , Histones/metabolism , MicroRNAs/physiology , Phenotype , Protein Modification, Translational , RNA, Long Noncoding/physiology , Repressor Proteins/metabolism , Transcription, Genetic
9.
Eur Heart J ; 35(43): 3013-20, 3020a-3020d, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25230814

ABSTRACT

Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.


Subject(s)
Arteries/physiology , Atherosclerosis/physiopathology , Apoptosis/physiology , Biomarkers/metabolism , Biomechanical Phenomena/physiology , Cell Proliferation/physiology , Cellular Senescence/physiology , Disease Progression , Endothelial Cells/physiology , Endothelium, Vascular/physiology , Homeostasis/physiology , Humans , Mechanoreceptors/physiology , Plaque, Atherosclerotic/physiopathology , Rupture, Spontaneous/physiopathology , Signal Transduction/physiology , Stress, Mechanical , Vascular Remodeling/physiology
10.
Vascul Pharmacol ; 62(2): 88-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24874278

ABSTRACT

Atherosclerosis is a multi-focal disease; it is associated with arterial curvatures, asymmetries and branches/bifurcations where non-uniform arterial geometry generates patterns of blood flow that are considerably more complex than elsewhere, and are collectively referred to as disturbed flow. Such regions are predisposed to atherosclerosis and are the sites of 'athero-susceptible' endothelial cells that express regionally different cell phenotypes than endothelium in nearby athero-protected locations. The regulatory hierarchy of endothelial function includes control at the epigenetic level. MicroRNAs and histone modifications are established epigenetic regulators that respond to disturbed flow. However, very recent reports have linked transcriptional regulation by DNA methylation to endothelial gene expression in disturbed flow in vivo and in vitro. We outline these in the context of site-specific atherosusceptibility mediated by local hemodynamics.


Subject(s)
DNA Methylation/genetics , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Epigenesis, Genetic/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Humans , Phenotype
11.
Circ Res ; 115(1): 32-43, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24755985

ABSTRACT

RATIONALE: Hemodynamic disturbed flow (DF) is associated with susceptibility to atherosclerosis. Endothelial Kruppel-Like Factor 4 (KLF4) is an important anti-inflammatory atheroprotective transcription factor that is suppressed in regions of DF. OBJECTIVE: The plasticity of epigenomic KLF4 transcriptional regulation by flow-mediated DNA methylation was investigated in vitro and in arterial tissue. METHODS AND RESULTS: To recapitulate dominant flow characteristics of atheroprotected and atherosusceptible arteries, human aortic endothelial cells were subjected to pulsatile undisturbed flow or oscillatory DF containing a flow-reversing phase. Differential CpG site methylation was measured by methylation-specific polymerase chain reaction, bisulfite pyrosequencing, and restriction enzyme-polymerase chain reaction. The methylation profiles of endothelium from disturbed and undisturbed flow sites of adult swine aortas were also investigated. In vitro, DF increased DNA methylation of CpG islands within the KLF4 promoter that significantly contributed to suppression of KLF4 transcription; the effects were mitigated by DNA methyltransferase (DNMT) inhibitors and knockdown of DNMT3A. Contributory mechanisms included DF-induced increase of DNMT3A protein (1.7-fold), DNMT3A enrichment (11-fold) on the KLF4 promoter, and competitive blocking of a myocyte enhancer factor-2 binding site in the KLF4 promoter near the transcription start site. DF also induced DNMT-sensitive propathological expression of downstream KLF4 transcription targets nitric oxide synthase 3, thrombomodulin, and monocyte chemoattractant protein-1. In support of the in vitro findings, swine aortic endothelium isolated from DF regions expressed significantly lower KLF4 and nitric oxide synthase 3, and bisulfite sequencing of KLF4 promoter identified a hypermethylated myocyte enhancer factor-2 binding site. CONCLUSIONS: Hemodynamics influence endothelial KLF4 expression through DNMT enrichment/myocyte enhancer factor-2 inhibition mechanisms of KLF4 promoter CpG methylation with regional consequences for atherosusceptibility.


Subject(s)
Atherosclerosis/etiology , DNA Methylation , Hemodynamics , Kruppel-Like Transcription Factors/genetics , Promoter Regions, Genetic , Animals , Blood Circulation , Cells, Cultured , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methyltransferase 3A , Endothelium, Vascular/metabolism , Humans , Kruppel-Like Factor 4 , MEF2 Transcription Factors/genetics , Nitric Oxide Synthase Type III/genetics , Swine
12.
J R Soc Interface ; 11(94): 20131079, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24554575

ABSTRACT

Drug eluting stents are associated with late stent thrombosis (LST), delayed healing and prolonged exposure of stent struts to blood flow. Using macroscale disturbed and undisturbed fluid flow waveforms, we numerically and experimentally determined the effects of microscale model strut geometries upon the generation of prothrombotic conditions that are mediated by flow perturbations. Rectangular cross-sectional stent strut geometries of varying heights and corresponding streamlined versions were studied in the presence of disturbed and undisturbed bulk fluid flow. Numerical simulations and particle flow visualization experiments demonstrated that the interaction of bulk fluid flow and stent struts regulated the generation, size and dynamics of the peristrut flow recirculation zones. In the absence of endothelial cells, deposition of thrombin-generated fibrin occurred primarily in the recirculation zones. When endothelium was present, peristrut expression of anticoagulant thrombomodulin (TM) was dependent on strut height and geometry. Thinner and streamlined strut geometries reduced peristrut flow recirculation zones decreasing prothrombotic fibrin deposition and increasing endothelial anticoagulant TM expression. The studies define physical and functional consequences of macro- and microscale variables that relate to thrombogenicity associated with the most current stent designs, and particularly to LST.


Subject(s)
Fibrin/metabolism , Gene Expression Regulation , Hemodynamics , Human Umbilical Vein Endothelial Cells/metabolism , Models, Cardiovascular , Stents , Thrombomodulin/biosynthesis , Thrombosis/metabolism , Cells, Cultured , Human Umbilical Vein Endothelial Cells/pathology , Humans , Thrombosis/etiology , Thrombosis/pathology
13.
Cardiovasc Res ; 99(2): 315-27, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23619421

ABSTRACT

Atherosclerosis initiates at predictable focal sites and develops to a spatially regional disease with limited distribution. There is compelling evidence that links haemodynamics to the localized origin of atherosclerotic lesions. Arterial flow in vivo is unsteady, dynamically complex, and regionally variable. Sites susceptible to atherosclerosis near arterial branches and curves are associated with regions of disturbed blood flow that contain repetitive phases of flow reversal resulting in steep multidirectional temporal and spatial gradients of wall shear stresses. Endothelium in atherosusceptible regions relative to protected sites shows activation of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), the altered expression of pro-inflammatory Nuclear Factor kappa B (NFκB) and oxidant/antioxidant pathways, and low expression of major protective factors, notably endothelial nitric oxide synthase and Kruppel-like Factors KLF2 and KLF4. At some atherosusceptible locations, reactive oxygen species levels are significantly elevated. Here we describe flow-related phenotypes identified in steady-state in vivo and outline some of the molecular mechanisms that may contribute to pre-lesional atherosusceptibility as deduced from complementary cell experiments in vitro. We conclude that disturbed flow is a significant local risk factor for atherosclerosis that induces a chronic low-level inflammatory state, an adaptive response to ensure continued function at the expense of increased susceptibility to atherogenesis. Surprisingly, when challenged by short-term hypercholesterolaemia in vivo, atherosusceptible endothelial phenotype was resistant to greater pro-inflammatory expression, suggesting that sustained hyperlipidaemia is required to overcome these protective characteristics.


Subject(s)
Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Hemodynamics , Mechanotransduction, Cellular , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomechanical Phenomena , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Kruppel-Like Factor 4 , Phenotype , Regional Blood Flow , Stress, Mechanical
14.
ACS Nano ; 6(10): 8824-36, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-22957767

ABSTRACT

Intracellular delivery of nanocarriers (NC) is controlled by their design and target cell phenotype, microenvironment, and functional status. Endothelial cells (EC) lining the vascular lumen represent an important target for drug delivery. Endothelium in vivo is constantly or intermittently (as, for example, during ischemia-reperfusion) exposed to blood flow, which influences NC-EC interactions by changing NC transport properties, and by direct mechanical effects upon EC mechanisms involved in NC binding and uptake. EC do not internalize antibodies to marker glycoprotein PECAM(CD31), yet internalize multivalent NC coated with PECAM antibodies (anti-PECAM/NC) via a noncanonical endocytic pathway distantly related to macropinocytosis. Here we studied the effects of flow on EC uptake of anti-PECAM/NC spheres (~180 nm diameter). EC adaptation to chronic flow, manifested by cellular alignment with flow direction and formation of actin stress fibers, inhibited anti-PECAM/NC endocytosis consistent with lower rates of anti-PECAM/NC endocytosis in vivo in arterial compared to capillary vessels. Acute induction of actin stress fibers by thrombin also inhibited anti-PECAM/NC endocytosis, demonstrating that formation of actin stress fibers impedes EC endocytic machinery. In contrast, acute flow without stress fiber formation, stimulated anti-PECAM/NC endocytosis. Anti-PECAM/NC endocytosis did not correlate with the number of cell-bound particles under flow or static conditions. PECAM cytosolic tail deletion and disruption of cholesterol-rich plasmalemma domains abrogated anti-PECAM/NC endocytosis stimulation by acute flow, suggesting complex regulation of a flow-sensitive endocytic pathway in EC. The studies demonstrate the importance of the local flow microenvironment for NC uptake by the endothelium and suggest that cell culture models of nanoparticle uptake should reflect the microenvironment and phenotype of the target cells.


Subject(s)
Endothelial Cells/metabolism , Nanocapsules/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Humans , Materials Testing , Shear Strength
15.
Arterioscler Thromb Vasc Biol ; 32(4): 979-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22267480

ABSTRACT

OBJECTIVE: Endothelial transcription factors Krüppel-like factor 4 (KLF4) and KLF2 are implicated in protection against atherogenesis. Steady-state microRNA (miR) regulation of KLFs in vivo is accessible by screening region-specific endothelial miRs and their targets. METHODS AND RESULTS: A subset of differentially expressed endothelial miRs was identified in atherosusceptible versus protected regions of normal swine aorta. In silico analyses predicted highly conserved binding sites in the 3'-untranslated region (3'UTR) of KLF4 for 5 miRs of the subset (miR-26a, -26b, -29a, -92a, and -103) and a single binding site for a miR-92a complex in the 3'UTR of KLF2. Of these, only miR-92a knockdown and knock-in resulted in responses of KLF4 and KLF2 expression in human arterial endothelial cells. Dual luciferase reporter assays demonstrated functional interactions of miR-92a with full-length 3'UTR sequences of both KLFs and with the specific binding elements therein. Two evolutionarily conserved miR-92a sites in KLF4 3'UTR and 1 site in KLF2 3'UTR were functionally validated. Knockdown of miR-92a in vitro resulted in partial rescue from cytokine-induced proinflammatory marker expression (monocyte chemotactic protein 1, vascular cell adhesion molecule-1, E-selectin, and endothelial nitric oxide synthase) that was attributable to enhanced KLF4 expression. Leukocyte-human arterial endothelial cell adhesion experiments supported this conclusion. In swine aortic arch endothelium, a site of atherosusceptibility where miR-92a expression was elevated, both KLFs were expressed at low levels relative to protected thoracic aorta. CONCLUSIONS: miR-92a coregulates KLF4 and KLF2 expression in arterial endothelium and contributes to phenotype heterogeneity associated with regional atherosusceptibility and protection in vivo.


Subject(s)
Atherosclerosis/genetics , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Kruppel-Like Transcription Factors/genetics , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Aorta/immunology , Aorta/metabolism , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Base Sequence , Binding Sites , Cell Adhesion , Cells, Cultured , Coculture Techniques , Conserved Sequence , Endothelial Cells/immunology , Endothelium, Vascular/immunology , Genes, Reporter , Genetic Predisposition to Disease , Humans , Inflammation Mediators/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Phenotype , RNA Interference , RNA, Messenger/metabolism , Swine , Transfection , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
J Heart Valve Dis ; 20(2): 165-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21560815

ABSTRACT

Aortic valve sclerosis (AVS), an early form of aortic valve disease, develops preferentially on the aortic side of valve leaflets, a predilection that is reflected in an heterogeneous side-specific gene expression profile. It has been ascertained that hypercholesterolemia is sufficient to initiate the endothelial expression of activated leukocyte adhesion molecule (ALCAM; CD166), restricted to the aortic side of the leaflet. Intercellular adhesion molecule-1 (ICAM-1) or vascular cell adhesion molecule-1 (VCAM-1)--both of which are more typically associated with early arterial inflammation--are not differentially expressed. ALCAM up-regulation by hypercholesterolemia suggests a side-specific spatial role in the recruitment of leukocytes to AVS sites.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/metabolism , Aortic Valve/immunology , Endothelial Cells/immunology , Heart Valve Diseases/immunology , Hypercholesterolemia/complications , Inflammation Mediators/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Animals , Aortic Valve/pathology , Endothelial Cells/pathology , Gene Expression Profiling , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Hypercholesterolemia/genetics , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Immunohistochemistry , Intercellular Adhesion Molecule-1/metabolism , Sclerosis , Swine , Up-Regulation , Vascular Cell Adhesion Molecule-1/metabolism
17.
Circ Cardiovasc Genet ; 4(3): 243-52, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21493819

ABSTRACT

BACKGROUND: Endothelial function is central to the localization of atherosclerosis. The in vivo endothelial phenotypic footprints of arterial bed identity and site-specific atherosusceptibility are addressed. METHODS AND RESULTS: Ninety-eight endothelial cell samples from 13 discrete coronary and noncoronary arterial regions of varying susceptibilities to atherosclerosis were isolated from 76 normal swine. Transcript profiles were analyzed to determine the steady-state in vivo endothelial phenotypes. An unsupervised systems biology approach using weighted gene coexpression networks showed highly correlated endothelial genes. Connectivity network analysis identified 19 gene modules, 12 of which showed significant association with circulatory bed classification. Differential expression of 1300 genes between coronary and noncoronary artery endothelium suggested distinct coronary endothelial phenotypes, with highest significance expressed in gene modules enriched for biological functions related to endoplasmic reticulum (ER) stress and unfolded protein binding, regulation of transcription and translation, and redox homeostasis. Furthermore, within coronary arteries, comparison of endothelial transcript profiles of susceptible proximal regions to protected distal regions suggested the presence of ER stress conditions in susceptible sites. Accumulation of reactive oxygen species throughout coronary endothelium was greater than in noncoronary endothelium consistent with coronary artery ER stress and lower endothelial expression of antioxidant genes in coronary arteries. CONCLUSIONS: Gene connectivity analyses discriminated between coronary and noncoronary endothelial transcript profiles and identified differential transcript levels associated with increased ER and oxidative stress in coronary arteries consistent with enhanced susceptibility to atherosclerosis.


Subject(s)
Coronary Vessels/metabolism , Endoplasmic Reticulum/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , Gene Regulatory Networks , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Coronary Vessels/anatomy & histology , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Gene Expression , Microarray Analysis , Phenotype , Swine
19.
Methods Enzymol ; 489: 109-26, 2011.
Article in English | MEDLINE | ID: mdl-21266227

ABSTRACT

The endothelium is a monolayer of cells that lines the entire inner surface of the cardiovascular and lymphatic circulations where it controls normal physiological functions through both systemic and local regulation. Endothelial phenotypes are heterogeneous, dynamic and malleable, properties that in large- and medium-sized arteries lead to a central role in the development of focal and regional atherosclerosis. The endothelial phenotype in athero-susceptible sites is different from that in nearby athero-resistant regions. Understanding the in vivo gene, protein, and metabolic expression profiles of susceptible endothelium is, therefore, an important spatiotemporal challenge in atherosclerosis research. Recent studies have demonstrated that endoplasmic reticulum (ER) stress and the UPR are characteristics of susceptible endothelium. Here, we outline global genomic profiling, pathway analyses, and gene connectivity approaches to the identification of UPR and associated pathways as discrete markers of athero-susceptibility in arterial endothelium.


Subject(s)
Atherosclerosis/physiopathology , Stress, Physiological , Unfolded Protein Response , Animals , Aorta/physiopathology , Atherosclerosis/metabolism , Endoplasmic Reticulum/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Gene Expression Profiling , Genomics , Microarray Analysis/methods , Sus scrofa
20.
Prog Biophys Mol Biol ; 104(1-3): 49-76, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21040741

ABSTRACT

A key aim of the cardiac Physiome Project is to develop theoretical models to simulate the functional behaviour of the heart under physiological and pathophysiological conditions. Heart function is critically dependent on the delivery of an adequate blood supply to the myocardium via the coronary vasculature. Key to this critical function of the coronary vasculature is system dynamics that emerge via the interactions of the numerous constituent components at a range of spatial and temporal scales. Here, we focus on several components for which theoretical approaches can be applied, including vascular structure and mechanics, blood flow and mass transport, flow regulation, angiogenesis and vascular remodelling, and vascular cellular mechanics. For each component, we summarise the current state of the art in model development, and discuss areas requiring further research. We highlight the major challenges associated with integrating the component models to develop a computational tool that can ultimately be used to simulate the responses of the coronary vascular system to changing demands and to diseases and therapies.


Subject(s)
Heart/physiology , Models, Cardiovascular , Biomechanical Phenomena , Coronary Circulation/physiology , Coronary Vessels/physiology , Humans , Myocardium/cytology , Myocardium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...