Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Stem Cells Transl Med ; 6(8): 1673-1683, 2017 08.
Article in English | MEDLINE | ID: mdl-28628273

ABSTRACT

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


Subject(s)
Cell Differentiation , Endothelial Cells/cytology , Hemodynamics , Induced Pluripotent Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Phenotype , Transcriptome , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
2.
JCI Insight ; 1(20): e90954, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27942596

ABSTRACT

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


Subject(s)
Coculture Techniques , Hepatic Stellate Cells/cytology , Hepatocytes/cytology , Macrophages/cytology , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Glucose/metabolism , Humans , Inflammation , Insulin Resistance , Lipids/analysis , Liver , Metabolome , Mice, Inbred C57BL , Models, Biological , Transcriptome
3.
Thromb Res ; 143: 34-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27179130

ABSTRACT

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


Subject(s)
Atrial Appendage/pathology , Atrial Fibrillation/blood , Atrial Fibrillation/complications , Endothelial Cells/pathology , Hemodynamics , Thrombosis/blood , Thrombosis/etiology , Atrial Fibrillation/pathology , Cells, Cultured , Fibrin/analysis , Humans , Thrombosis/pathology
4.
Chem Biol Interact ; 255: 31-44, 2016 Aug 05.
Article in English | MEDLINE | ID: mdl-26626330

ABSTRACT

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


Subject(s)
Anti-HIV Agents/toxicity , Chemical and Drug Induced Liver Injury/genetics , Hepatocytes/drug effects , Nevirapine/toxicity , Ritonavir/toxicity , Transcriptome , Cell Culture Techniques/methods , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Drug Evaluation, Preclinical/methods , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology
5.
Arterioscler Thromb Vasc Biol ; 35(11): 2354-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26338297

ABSTRACT

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


Subject(s)
Arteries/physiopathology , Collateral Circulation , Ischemia/physiopathology , Mechanotransduction, Cellular , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Arteries/metabolism , Arteries/pathology , Blood Flow Velocity , Cells, Cultured , Disease Models, Animal , Femoral Artery/physiopathology , Femoral Artery/surgery , Gene Expression Regulation , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Ligation , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Regional Blood Flow , Stress, Mechanical , Time Factors , Vascular Remodeling
6.
Arterioscler Thromb Vasc Biol ; 35(10): 2185-95, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26293464

ABSTRACT

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


Subject(s)
Cardiovascular Diseases/drug therapy , Drug Evaluation, Preclinical/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/drug effects , Animals , Cardiovascular Diseases/blood , Cells, Cultured , Endothelial Cells/drug effects , Humans , In Vitro Techniques , Lipoproteins, LDL/metabolism , Macaca fascicularis , Models, Cardiovascular , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Species Specificity
7.
Nat Commun ; 4: 1525, 2013.
Article in English | MEDLINE | ID: mdl-23443553

ABSTRACT

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


Subject(s)
Endothelial Cells/pathology , Hemodynamics/physiology , Inflammation/pathology , Inflammation/physiopathology , Blood Flow Velocity , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Coronary Circulation , Endothelial Cells/metabolism , Fourier Analysis , Gene Expression Regulation , Humans , Inflammation/genetics , Models, Cardiovascular , Mutation/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stress, Mechanical
8.
J Clin Invest ; 122(12): 4727-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160196

ABSTRACT

The endothelium regulates vascular homeostasis, and endothelial dysfunction is a proximate event in the pathogenesis of atherothrombosis. Stimulation of the endothelium with proinflammatory cytokines or exposure to hemodynamic-induced disturbed flow leads to a proadhesive and prothrombotic phenotype that promotes atherothrombosis. In contrast, exposure to arterial laminar flow induces a gene program that confers a largely antiadhesive, antithrombotic effect. The molecular basis for this differential effect on endothelial function remains poorly understood. While recent insights implicate Kruppel-like factors (KLFs) as important regulators of vascular homeostasis, the in vivo role of these factors in endothelial biology remains unproven. Here, we show that endothelial KLF4 is an essential determinant of atherogenesis and thrombosis. Using in vivo EC-specific KLF4 overexpression and knockdown murine models, we found that KLF4 induced an antiadhesive, antithrombotic state. Mechanistically, we demonstrated that KLF4 differentially regulated pertinent endothelial targets via competition for the coactivator p300. These observations provide cogent evidence implicating endothelial KLFs as essential in vivo regulators of vascular function in the adult animal.


Subject(s)
Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/physiology , Thrombosis/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Protein Interaction Domains and Motifs , Thrombosis/genetics , Vasculitis/metabolism , p300-CBP Transcription Factors/metabolism
9.
Expert Opin Drug Metab Toxicol ; 8(8): 999-1014, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22632603

ABSTRACT

INTRODUCTION: The frequent failure of high-throughput screening cell-based tools to accurately predict in vivo responses, coupled with limitations of animal models in predicting human safety or drug efficacy, impairs the de-risking process for biotechnology/pharmaceutical companies as they make important decisions to enter human clinical trials. Organotypic systems strive to fill the gap between these screening and in vivo studies and provide a solution. AREAS COVERED: The authors examine the various approaches to recreate physiological response on the bench and trace the evolution of organotypic systems, while discussing intrinsic challenges and opportunities that lie ahead. Furthermore, they cite literature that is the foundation of several biotechnology research companies addressing this issue and discuss major government-funded initiatives to aid the development of these systems in an effort to fill this existing gap. EXPERT OPINION: Decisions from translational systems that bridge basic drug efficacy and toxicity with clinical outcome must be benchmarked against human-relevant endpoints and clinical data for early meaningful pre-clinical decisions. The use of human primary cells coupled with emerging technologies that allow precise control of the culture environment and analysis of meaningful endpoints paves the way for human organotypic systems as a major initiative in de-risking the drug discovery and development process.


Subject(s)
Cell Culture Techniques , Drug-Related Side Effects and Adverse Reactions , Inactivation, Metabolic , Pharmaceutical Preparations/metabolism , Cell Line , Drug Discovery , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hemodynamics , Hepatocytes/drug effects , Hepatocytes/metabolism , High-Throughput Screening Assays/methods , Humans , Liver/cytology , Liver/drug effects
10.
PLoS One ; 7(5): e37231, 2012.
Article in English | MEDLINE | ID: mdl-22623999

ABSTRACT

BACKGROUND: Tumor cells in vivo encounter diverse types of microenvironments both at the site of the primary tumor and at sites of distant metastases. Understanding how the various mechanical properties of these microenvironments affect the biology of tumor cells during disease progression is critical in identifying molecular targets for cancer therapy. METHODOLOGY/PRINCIPAL FINDINGS: This study uses flexible polyacrylamide gels as substrates for cell growth in conjunction with a novel proteomic approach to identify the properties of rigidity-dependent cancer cell lines that contribute to their differential growth on soft and rigid substrates. Compared to cells growing on more rigid/stiff substrates (>10,000 Pa), cells on soft substrates (150-300 Pa) exhibited a longer cell cycle, due predominantly to an extension of the G1 phase of the cell cycle, and were metabolically less active, showing decreased levels of intracellular ATP and a marked reduction in protein synthesis. Using stable isotope labeling of amino acids in culture (SILAC) and mass spectrometry, we measured the rates of protein synthesis of over 1200 cellular proteins under growth conditions on soft and rigid/stiff substrates. We identified cellular proteins whose syntheses were either preferentially inhibited or preserved on soft matrices. The former category included proteins that regulate cytoskeletal structures (e.g., tubulins) and glycolysis (e.g., phosphofructokinase-1), whereas the latter category included proteins that regulate key metabolic pathways required for survival, e.g., nicotinamide phosphoribosyltransferase, a regulator of the NAD salvage pathway. CONCLUSIONS/SIGNIFICANCE: The cellular properties of rigidity-dependent cancer cells growing on soft matrices are reminiscent of the properties of dormant cancer cells, e.g., slow growth rate and reduced metabolism. We suggest that the use of relatively soft gels as cell culture substrates would allow molecular pathways to be studied under conditions that reflect the different mechanical environments encountered by cancer cells upon metastasis to distant sites.


Subject(s)
Cellular Microenvironment/physiology , Extracellular Matrix/chemistry , Neoplasms/metabolism , Protein Biosynthesis/physiology , Acrylic Resins , Adenosine Triphosphate/metabolism , Biomechanical Phenomena , Bromodeoxyuridine , Cell Line, Tumor , Cyclin D1/metabolism , Extracellular Matrix/metabolism , Humans , Isotope Labeling , Mass Spectrometry , Neoplasms/physiopathology , Proteomics/methods
11.
Magn Reson Med ; 66(5): 1382-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21656547

ABSTRACT

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


Subject(s)
Aorta, Thoracic/anatomy & histology , Magnetic Resonance Imaging, Cine/methods , Animals , Humans , Image Enhancement/methods , Imaging, Three-Dimensional/methods , Mathematics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Shear Strength
12.
Arterioscler Thromb Vasc Biol ; 31(7): 1625-33, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21527747

ABSTRACT

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


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Fibronectins/metabolism , Hemodynamics , Inflammation/metabolism , Signal Transduction , TCF Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Genes, Reporter , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA Interference , Stress, Mechanical , TCF Transcription Factors/genetics , Time Factors , Transcriptional Activation , Transfection , beta Catenin/genetics
13.
PLoS One ; 5(9): e12905, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20886123

ABSTRACT

BACKGROUND: The mechanical properties of the extracellular matrix have an important role in cell growth and differentiation. However, it is unclear as to what extent cancer cells respond to changes in the mechanical properties (rigidity/stiffness) of the microenvironment and how this response varies among cancer cell lines. METHODOLOGY/PRINCIPAL FINDINGS: In this study we used a recently developed 96-well plate system that arrays extracellular matrix-conjugated polyacrylamide gels that increase in stiffness by at least 50-fold across the plate. This plate was used to determine how changes in the rigidity of the extracellular matrix modulate the biological properties of tumor cells. The cell lines tested fall into one of two categories based on their proliferation on substrates of differing stiffness: "rigidity dependent" (those which show an increase in cell growth as extracellular rigidity is increased), and "rigidity independent" (those which grow equally on both soft and stiff substrates). Cells which grew poorly on soft gels also showed decreased spreading and migration under these conditions. More importantly, seeding the cell lines into the lungs of nude mice revealed that the ability of cells to grow on soft gels in vitro correlated with their ability to grow in a soft tissue environment in vivo. The lung carcinoma line A549 responded to culture on soft gels by expressing the differentiated epithelial marker E-cadherin and decreasing the expression of the mesenchymal transcription factor Slug. CONCLUSIONS/SIGNIFICANCE: These observations suggest that the mechanical properties of the matrix environment play a significant role in regulating the proliferation and the morphological properties of cancer cells. Further, the multiwell format of the soft-plate assay is a useful and effective adjunct to established 3-dimensional cell culture models.


Subject(s)
Cell Proliferation , Extracellular Matrix/chemistry , Neoplasms/physiopathology , Animals , Biomechanical Phenomena , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism
14.
Circ Res ; 106(11): 1703-11, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20378855

ABSTRACT

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


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Fibronectins/metabolism , Hemodynamics , Inflammation/metabolism , Mechanotransduction, Cellular , Animals , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/physiopathology , Feedback, Physiological , Fibronectins/genetics , Humans , Inflammation/genetics , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pulsatile Flow , RNA Interference , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection , Up-Regulation
15.
J Vasc Res ; 47(2): 168-80, 2010.
Article in English | MEDLINE | ID: mdl-19851078

ABSTRACT

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


Subject(s)
Atherosclerosis/immunology , Inflammation/immunology , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cell Adhesion Molecules/metabolism , Cell Proliferation , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Hemodynamics , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/physiopathology , Inflammation Mediators/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , Phenotype , Protein Processing, Post-Translational , Signal Transduction , Stress, Mechanical , Transcription, Genetic
16.
J Muscle Res Cell Motil ; 30(1-2): 41-55, 2009.
Article in English | MEDLINE | ID: mdl-19205907

ABSTRACT

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


Subject(s)
Atherosclerosis/metabolism , Cytoskeletal Proteins/biosynthesis , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Phosphoproteins/biosynthesis , Shear Strength , Animals , Aorta/injuries , Aorta/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Cell Adhesion , Cell Movement , Collagen/metabolism , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , LIM Domain Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Stress Fibers/metabolism , Tenascin/metabolism
17.
Arterioscler Thromb Vasc Biol ; 29(5): 725-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19229069

ABSTRACT

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


Subject(s)
Atherosclerosis/physiopathology , Endothelial Cells/metabolism , Interleukin-8/physiology , Myocytes, Smooth Muscle/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Coculture Techniques , Coronary Vessels/cytology , Coronary Vessels/metabolism , Humans , Tunica Intima/metabolism , Umbilical Veins/cytology
18.
Am J Physiol Heart Circ Physiol ; 296(2): H442-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19028801

ABSTRACT

Platelet-derived growth factor (PDGF)-BB is a well-known smooth muscle (SM) cell (SMC) phenotypic modulator that signals by binding to PDGF alphaalpha-, alphabeta-, and betabeta-membrane receptors. PDGF-DD is a recently identified PDGF family member, and its role in SMC phenotypic modulation is unknown. Here we demonstrate that PDGF-DD inhibited expression of multiple SMC genes, including SM alpha-actin and SM myosin heavy chain, and upregulated expression of the potent SMC differentiation repressor gene Kruppel-like factor-4 at the mRNA and protein levels. On the basis of the results of promoter-reporter assays, changes in SMC gene expression were mediated, at least in part, at the level of transcription. Attenuation of the SMC phenotypic modulatory activity of PDGF-DD by pharmacological inhibitors of ERK phosphorylation and by a small interfering RNA to Kruppel-like factor-4 highlight the role of these two pathways in this process. PDGF-DD failed to repress SM alpha-actin and SM myosin heavy chain in mouse SMCs lacking a functional PDGF beta-receptor. Importantly, PDGF-DD expression was increased in neointimal lesions in the aortic arch region of apolipoprotein C-deficient (ApoE(-/-)) mice. Furthermore, human endothelial cells exposed to an atherosclerosis-prone flow pattern, as in vascular regions susceptible to the development of atherosclerosis, exhibited a significant increase in PDGF-DD expression. These findings demonstrate a novel activity for PDGF-DD in SMC biology and highlight the potential contribution of this molecule to SMC phenotypic modulation in the setting of disturbed blood flow.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Lymphokines/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/metabolism , Actins/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/physiopathology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, Reporter , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myosin Heavy Chains/metabolism , Phenotype , Phosphorylation , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Recombinant Proteins/metabolism , Regional Blood Flow , Stress, Mechanical , Time Factors , Up-Regulation , ets-Domain Protein Elk-1/metabolism , Calponins
19.
Am J Pathol ; 173(4): 1220-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18772338

ABSTRACT

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


Subject(s)
CD36 Antigens/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Signal Transduction , Thrombospondin 1/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Endothelial Cells/drug effects , Humans , Inflammation , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phenotype , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stress, Mechanical , Thrombospondin 1/pharmacology , Time Factors , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/pharmacology
20.
Circ Res ; 103(6): 671-9, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18669917

ABSTRACT

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


Subject(s)
NF-kappa B/metabolism , Oxidants/physiology , Signal Transduction/physiology , p21-Activated Kinases/physiology , Animals , Blood Flow Velocity/physiology , Cattle , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Humans , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Shear Strength
SELECTION OF CITATIONS
SEARCH DETAIL
...