Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Physiol Rev ; 103(2): 1247-1421, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36603156

ABSTRACT

This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.


Subject(s)
Endothelial Cells , Mechanotransduction, Cellular , Humans , Mechanotransduction, Cellular/physiology , Endothelial Cells/metabolism , Epigenesis, Genetic , Signal Transduction/physiology , Myocytes, Smooth Muscle , Stress, Mechanical
2.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Article in English | MEDLINE | ID: mdl-34810252

ABSTRACT

Vascular endothelial cells are exposed to shear stresses with disturbed vs. laminar flow patterns, which lead to proinflammatory vs. antiinflammatory phenotypes, respectively. Effective treatment against endothelial inflammation and the consequent atherogenesis requires the identification of new therapeutic molecules and the development of drugs targeting these molecules. Using Connectivity Map, we have identified vitexin, a natural flavonoid, as a compound that evokes the gene-expression changes caused by pulsatile shear, which mimics laminar flow with a clear direction, vs. oscillatory shear (OS), which mimics disturbed flow without a clear direction. Treatment with vitexin suppressed the endothelial inflammation induced by OS or tumor necrosis factor-α. Administration of vitexin to mice subjected to carotid partial ligation blocked the disturbed flow-induced endothelial inflammation and neointimal formation. In hyperlipidemic mice, treatment with vitexin ameliorated atherosclerosis. Using SuperPred, we predicted that apurinic/apyrimidinic endonuclease1 (APEX1) may directly interact with vitexin, and we experimentally verified their physical interactions. OS induced APEX1 nuclear translocation, which was inhibited by vitexin. OS promoted the binding of acetyltransferase p300 to APEX1, leading to its acetylation and nuclear translocation. Functionally, knocking down APEX1 with siRNA reversed the OS-induced proinflammatory phenotype, suggesting that APEX1 promotes inflammation by orchestrating the NF-κB pathway. Animal experiments with the partial ligation model indicated that overexpression of APEX1 negated the action of vitexin against endothelial inflammation, and that endothelial-specific deletion of APEX1 ameliorated atherogenesis. We thus propose targeting APEX1 with vitexin as a potential therapeutic strategy to alleviate atherosclerosis.


Subject(s)
Apigenin/genetics , Apigenin/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Endothelial Cells/metabolism , Active Transport, Cell Nucleus , Animals , Atherosclerosis , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Mice , Phenotype , Phosphorylation , Protein Binding , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , p300-CBP Transcription Factors/metabolism
3.
Front Cell Dev Biol ; 9: 642150, 2021.
Article in English | MEDLINE | ID: mdl-33898431

ABSTRACT

The transition of flow microenvironments from veins to arteries in vein graft surgery induces "peel-off" of venous endothelial cells (vECs) and results in restenosis. Recently, arterial laminar shear stress (ALS) and oscillatory shear stress (OS) have been shown to affect the cell cycle and inflammation through epigenetic controls such as histone deacetylation by histone deacetylases (HDACs) and trimethylation on lysine 9 of histone 3 (H3K9me3) in arterial ECs. However, the roles of H3K9me3 and HDAC in vEC damage under ALS are not known. We hypothesized that the different responses of HDACs and H3K9me3 might cause vEC damage under the transition of venous flow to arterial flow. We found that arterial ECs showed high expression of H3K9me3 protein and were retained in the G0 phase of the cell cycle after being subjected to ALS. vECs became round under ALS with a decrease in the expression of H3K9me3, HDAC3, and HDAC5, and an increase in the expression of vascular cell adhesion molecule 1 (VCAM-1). Inhibition of HDACs activity by a specific inhibitor, phenylbutyrate, in arterial ECs caused similar ALS-induced inflammation and cell loss as observed in vECs. Activation of HDACs and H3K9me3 by ITSA-1, an HDAC activator, could prevent ALS-induced peel-off and reduced VCAM-1 expression in vECs. Moreover, shear stress modulates EC morphology by the regulation of focal adhesion kinase (FAK) expression. ITSA-1 or EGF could increase phosphorylated (p)-FAK expression in vECs under ALS. We found that perturbation of the activity of p-FAK and increase in p-FAK expression restored ALS-induced H3K9me3 expression in vECs. Hence, the abnormal mechanoresponses of H3K9me3 and HDAC in vECs after being subjected to ALS could be reversed by ITSA-1 or EGF treatment: this offers a strategy to prevent vein graft failure.

4.
Sci Adv ; 6(6): eaay0264, 2020 02.
Article in English | MEDLINE | ID: mdl-32076643

ABSTRACT

During endoderm formation, cell identity and tissue morphogenesis are tightly controlled by cell-intrinsic and cell-extrinsic factors such as biochemical and physical inputs. While the effects of biochemical factors are well studied, the physical cues that regulate cell division and differentiation are poorly understood. RNA sequencing analysis demonstrated increases of endoderm-specific gene expression in hPSCs cultured on soft substrate (Young's modulus, 3 ± 0.45 kPa) in comparison with hard substrate (Young's modulus, 165 ± 6.39 kPa). Further analyses revealed that multiple long noncoding RNAs (lncRNAs) were up-regulated on soft substrate; among them, LINC00458 was identified as a stiffness-dependent lncRNA specifically required for hPSC differentiation toward an early endodermal lineage. Gain- and loss-of-function experiments confirmed that LINC00458 is functionally required for hPSC endodermal lineage specification induced by soft substrates. Our study provides evidence that mechanical cues regulate the expression of LINC00458 and induce differentiation of hPSC into hepatic lineage progenitors.


Subject(s)
Endoderm/cytology , Endoderm/metabolism , Gene Expression Regulation, Developmental , RNA, Long Noncoding/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Cells, Cultured , Extracellular Matrix , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Mice , Models, Biological , Organ Specificity/genetics , RNA Interference , Transcriptome
6.
Arterioscler Thromb Vasc Biol ; 39(12): 2492-2504, 2019 12.
Article in English | MEDLINE | ID: mdl-31597449

ABSTRACT

OBJECTIVE: Understanding message delivery among vascular cells is essential for deciphering the intercellular communications in cardiovascular diseases. MicroRNA (miR)-92a is enriched in endothelial cells (ECs) and circulation under atheroprone conditions. Macrophages are the primary immune cells in atherosclerotic lesions that modulate lesion development. Therefore, we hypothesize that, in response to atheroprone stimuli, ECs export miR-92a to macrophages to regulate their functions and enhance atherosclerotic progression. Approach and Results: We investigated the macrophage functions that are regulated by EC miR-92a under atheroprone microenvironments. We first determined the distributions of functional extracellular miR-92a by fractionating the intravesicular and extravesicular compartments from endothelial conditioned media and mice serum. The results indicate that extracellular vesicles are the primary vehicles for EC miR-92a transportation. Overexpression of miR-92a in ECs enhanced the proinflammatory responses and low-density lipoprotein uptake, while impaired the migration, of cocultured macrophage. Opposite effects were found in macrophages cocultured with ECs with miR-92a knockdown. Further analyses demonstrated that intravesicular miR-92a suppressed the expression of target gene KLF4 (Krüppel-like factor 4) in macrophages, suggesting a mechanism by which intravesicular miR-92a regulates recipient cell functions. Indeed, the overexpression of KLF4 rescued the EC miR-92a-induced macrophage atheroprone phenotypes. Furthermore, an inverse correlation of intravesicular miR-92a in blood serum and KLF4 expression in lesions was observed in atherosclerotic animals, indicating the potential function of extracellular miR-92a in regulating vascular diseases. CONCLUSIONS: EC miR-92a can be transported to macrophages through extracellular vesicles to regulate KLF4 levels, thus leading to the atheroprone phenotypes of macrophage and, hence, atherosclerotic lesion formation.


Subject(s)
Atherosclerosis/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Macrophages/metabolism , MicroRNAs/genetics , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Communication , Cells, Cultured , Extracellular Fluid/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/ultrastructure , Mice , MicroRNAs/biosynthesis , Microscopy, Electron, Transmission
7.
Proc Natl Acad Sci U S A ; 116(26): 12974-12979, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31182601

ABSTRACT

Pulsatile shear (PS) and oscillatory shear (OS) elicit distinct mechanotransduction signals that maintain endothelial homeostasis or induce endothelial dysfunction, respectively. A subset of microRNAs (miRs) in vascular endothelial cells (ECs) are differentially regulated by PS and OS, but the regulation of the miR processing and its implications in EC biology by shear stress are poorly understood. From a systematic in silico analysis for RNA binding proteins that regulate miR processing, we found that nucleolin (NCL) is a major regulator of miR processing in response to OS and essential for the maturation of miR-93 and miR-484 that target mRNAs encoding Krüppel-like factor 2 (KLF2) and endothelial nitric oxide synthase (eNOS). Additionally, anti-miR-93 and anti-miR-484 restore KLF2 and eNOS expression and NO bioavailability in ECs under OS. Analysis of posttranslational modifications of NCL identified that serine 328 (S328) phosphorylation by AMP-activated protein kinase (AMPK) was a major PS-activated event. AMPK phosphorylation of NCL sequesters it in the nucleus, thereby inhibiting miR-93 and miR-484 processing and their subsequent targeting of KLF2 and eNOS mRNA. Elevated levels of miR-93 and miR-484 were found in sera collected from individuals afflicted with coronary artery disease in two cohorts. These findings provide translational relevance of the AMPK-NCL-miR-93/miR-484 axis in miRNA processing in EC health and coronary artery disease.


Subject(s)
Coronary Artery Disease/genetics , Mechanotransduction, Cellular/genetics , MicroRNAs/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adult , Aged , Animals , Case-Control Studies , Cells, Cultured , Computational Biology , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Gene Knockdown Techniques , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , Middle Aged , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Protein Processing, Post-Translational , RNA Processing, Post-Transcriptional , Serine/metabolism , Stress, Mechanical , Nucleolin
8.
Biomaterials ; 204: 59-69, 2019 06.
Article in English | MEDLINE | ID: mdl-30884320

ABSTRACT

The use of biochemical signaling to derive smooth muscle cells (SMCs) from mesenchymal stem cells (MSCs) has been explored, but the induction of a fully functional SMC phenotype remains to be a major challenge. Cell morphology has been shown to regulate MSC differentiation into various lineages, including SMCs. We engineered substrates with microgrooves to induce cell elongation to study the mechanism underlying the MSC shape modulation in SMC differentiation. In comparison to those on flat substrates, MSCs cultured on engineered substrates were elongated with increased aspect ratios for both cell body and nucleus, as well as augmented cytoskeletal tensions. Biochemical studies indicated that the microgroove-elongated cells expressed significantly higher levels of SMC markers. MicroRNA analyses showed that up-regulation of miR-145 and the consequent repression of KLF4 in these elongated cells promoted MSC-to-SMC differentiation. Rho/ROCK inhibitions, which impair cytoskeletal tension, attenuated cell and nuclear elongations and disrupted the miR-145/KLF4 regulation for SMC differentiation. Furthermore, cell traction force measurements showed that miR-145 is essential for the functional contractility in the microgroove-induced SMC differentiation. Collectively, our findings demonstrate that, through a Rho-ROCK/miR-145/KLF4 pathway, the elongated cell shape serves as a decisive geometric cue to direct MSC differentiation into functional SMCs.


Subject(s)
Cell Differentiation , Cell Shape , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , Myocytes, Smooth Muscle/cytology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape/drug effects , Cell Shape/genetics , Dimethylpolysiloxanes/pharmacology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Proc Natl Acad Sci U S A ; 115(50): E11681-E11690, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478057

ABSTRACT

The dramatic reorganization of chromatin during mitosis is perhaps one of the most fundamental of all cell processes. It remains unclear how epigenetic histone modifications, despite their crucial roles in regulating chromatin architectures, are dynamically coordinated with chromatin reorganization in controlling this process. We have developed and characterized biosensors with high sensitivity and specificity based on fluorescence resonance energy transfer (FRET). These biosensors were incorporated into nucleosomes to visualize histone H3 Lys-9 trimethylation (H3K9me3) and histone H3 Ser-10 phosphorylation (H3S10p) simultaneously in the same live cell. We observed an anticorrelated coupling in time between H3K9me3 and H3S10p in a single live cell during mitosis. A transient increase of H3S10p during mitosis is accompanied by a decrease of H3K9me3 that recovers before the restoration of H3S10p upon mitotic exit. We further showed that H3S10p is causatively critical for the decrease of H3K9me3 and the consequent reduction of heterochromatin structure, leading to the subsequent global chromatin reorganization and nuclear envelope dissolution as a cell enters mitosis. These results suggest a tight coupling of H3S10p and H3K9me3 dynamics in the regulation of heterochromatin dissolution before a global chromatin reorganization during mitosis.


Subject(s)
Biosensing Techniques/methods , Chromatin Assembly and Disassembly , Histone Code , Bacterial Proteins , Chromatin Assembly and Disassembly/genetics , Fluorescence Resonance Energy Transfer/methods , Green Fluorescent Proteins , HEK293 Cells , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Code/genetics , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Luminescent Proteins , Mitosis , Models, Biological , Single-Cell Analysis/methods
10.
Proc Natl Acad Sci U S A ; 115(1): 133-138, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29255056

ABSTRACT

Leukocyte transmigration across vessel walls is a critical step in the innate immune response. Upon their activation and firm adhesion to vascular endothelial cells (VECs), leukocytes preferentially extravasate across junctional gaps in the endothelial monolayer (paracellular diapedesis). It has been hypothesized that VECs facilitate paracellular diapedesis by opening their cell-cell junctions in response to the presence of an adhering leukocyte. However, it is unclear how leukocytes interact mechanically with VECs to open the VEC junctions and migrate across the endothelium. In this study, we measured the spatial and temporal evolution of the 3D traction stresses generated by the leukocytes and VECs to elucidate the sequence of mechanical events involved in paracellular diapedesis. Our measurements suggest that the contractile stresses exerted by the leukocytes and the VECs can separately perturb the junctional tensions of VECs to result in the opening of gaps before the initiation of leukocyte transmigration. Decoupling the stresses exerted by the transmigrating leukocytes and the VECs reveals that the leukocytes actively contract the VECs to open a junctional gap and then push themselves across the gap by generating strong stresses that push into the matrix. In addition, we found that diapedesis is facilitated when the tension fluctuations in the VEC monolayer were increased by proinflammatory thrombin treatment. Our findings demonstrate that diapedesis can be mechanically regulated by the transmigrating leukocytes and by proinflammatory signals that increase VEC contractility.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Junctions/metabolism , Leukocytes/metabolism , Models, Biological , Transendothelial and Transepithelial Migration/physiology , HL-60 Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Leukocytes/cytology
11.
Sci Rep ; 7(1): 14996, 2017 11 08.
Article in English | MEDLINE | ID: mdl-29118325

ABSTRACT

The earliest atherosclerotic lesions preferentially develop in arterial regions experienced disturbed blood flow, which induces endothelial expression of pro-atherogenic genes and the subsequent endothelial dysfunction. Our previous study has demonstrated an up-regulation of DNA methyltransferase 1 (DNMT1) and a global hypermethylation in vascular endothelium subjected to disturbed flow. Here, we determined that DNMT1-specific inhibition in arterial wall ameliorates the disturbed flow-induced atherosclerosis through, at least in part, targeting cell cycle regulator cyclin A and connective tissue growth factor (CTGF). We identified the signaling pathways mediating the flow-induction of DNMT1. Inhibition of the mammalian target of rapamycin (mTOR) suppressed the DNMT1 up-regulation both in vitro and in vivo. Together, our results demonstrate that disturbed flow influences endothelial function and induces atherosclerosis in an mTOR/DNMT1-dependent manner. The conclusions obtained from this study might facilitate further evaluation of the epigenetic regulation of endothelial function during the pathological development of atherosclerosis and offer novel prevention and therapeutic targets of this disease.


Subject(s)
Atherosclerosis/pathology , Endothelium, Vascular/pathology , Epigenesis, Genetic/physiology , Hemorheology/physiology , Animals , Arteries/pathology , Arteries/physiopathology , Atherosclerosis/genetics , Atherosclerosis/physiopathology , Cattle , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Cyclin A/genetics , Cyclin A/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/physiology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Promoter Regions, Genetic/genetics , TOR Serine-Threonine Kinases/metabolism
12.
Proc Natl Acad Sci U S A ; 114(31): 8271-8276, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716920

ABSTRACT

Vascular endothelial cells (ECs) at arterial branches and curvatures experience disturbed blood flow and induce a quiescent-to-activated phenotypic transition of the adjacent smooth muscle cells (SMCs) and a subsequent smooth muscle hyperplasia. However, the mechanism underlying the flow pattern-specific initiation of EC-to-SMC signaling remains elusive. Our previous study demonstrated that endothelial microRNA-126-3p (miR-126-3p) acts as a key intercellular molecule to increase turnover of the recipient SMCs, and that its release is reduced by atheroprotective laminar shear (12 dynes/cm2) to ECs. Here we provide evidence that atherogenic oscillatory shear (0.5 ± 4 dynes/cm2), but not atheroprotective pulsatile shear (12 ± 4 dynes/cm2), increases the endothelial secretion of nonmembrane-bound miR-126-3p and other microRNAs (miRNAs) via the activation of SNAREs, vesicle-associated membrane protein 3 (VAMP3) and synaptosomal-associated protein 23 (SNAP23). Knockdown of VAMP3 and SNAP23 reduces endothelial secretion of miR-126-3p and miR-200a-3p, as well as the proliferation, migration, and suppression of contractile markers in SMCs caused by EC-coculture. Pharmacological intervention of mammalian target of rapamycin complex 1 in ECs blocks endothelial secretion and EC-to-SMC transfer of miR-126-3p through transcriptional inhibition of VAMP3 and SNAP23. Systemic inhibition of VAMP3 and SNAP23 by rapamycin or periadventitial application of the endocytosis inhibitor dynasore ameliorates the disturbed flow-induced neointimal formation, whereas intraluminal overexpression of SNAP23 aggravates it. Our findings demonstrate the flow-pattern-specificity of SNARE activation and its contribution to the miRNA-mediated EC-SMC communication.


Subject(s)
Hyperplasia/pathology , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/metabolism , Animals , Endothelial Cells/physiology , Humans , Mice , Mice, Knockout , MicroRNAs/genetics , Myocytes, Smooth Muscle/physiology , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , SNARE Proteins/metabolism , Vesicle-Associated Membrane Protein 3/genetics
13.
Physiol Genomics ; 49(7): 339-345, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28500253

ABSTRACT

The long noncoding RNAs (lncRNAs), which constitute a large portion of the transcriptome, have gained intense research interest because of their roles in regulating physiological and pathophysiological functions in the cell. We identified from RNA-Seq profiling a set of lncRNAs in cultured human umbilical vein endothelial cells (HUVECs) that are differentially regulated by atheroprotective vs. atheroprone shear flows. Among the comprehensively annotated lncRNAs, including both known and novel transcripts, LINC00341 is one of the most abundant lncRNAs in endothelial cells. Moreover, its expression level is enhanced by atheroprotective pulsatile shear flow and atorvastatin. Overexpression of LINC00341 suppresses the expression of vascular cell adhesion molecule 1 (VCAM1) and the adhesion of monocytes induced by atheroprone flow and tumor necrosis factor-alpha. Underlying this anti-inflammatory role, LINC00341 guides enhancer of zest homolog 2, a core histone methyltransferase of polycomb repressive complex 2, to the promoter region of the VCAM1 gene to suppress VCAM1. Network analysis reveals that the key signaling pathways (e.g., Rho and PI3K/AKT) are co-regulated with LINC00341 in endothelial cells in response to pulsatile shear. Together, these findings suggest that LINC00341, as an example of lncRNAs, plays important roles in modulating endothelial function in health and disease.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Inflammation/genetics , RNA, Long Noncoding/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Atorvastatin/pharmacology , Cell Adhesion , Gene Expression Regulation , Gene Regulatory Networks , Humans , Inflammation/pathology , Monocytes/pathology , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Proc Natl Acad Sci U S A ; 113(41): 11525-11530, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27671657

ABSTRACT

The focal nature of atherosclerotic lesions suggests an important role of local hemodynamic environment. Recent studies have demonstrated significant roles of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in mediating mechanotransduction and vascular homeostasis. The objective of this study is to investigate the functional role of YAP/TAZ in the flow regulation of atheroprone endothelial phenotypes and the consequential development of atherosclerotic lesions. We found that exposure of cultured endothelial cells (ECs) to the atheroprone disturbed flow resulted in YAP/TAZ activation and translocation into EC nucleus to up-regulate the target genes, including cysteine-rich angiogenic inducer 61 (CYR61), connective tissue growth factor (CTGF), and ankyrin repeat domain 1 (ANKRD1). In contrast, the athero-protective laminar flow suppressed YAP/TAZ activities. En face analysis of mouse arteries demonstrated an increased nuclear localization of YAP/TAZ and elevated levels of the target genes in the endothelium in atheroprone areas compared with athero-protective areas. YAP/TAZ knockdown significantly attenuated the disturbed flow induction of EC proliferative and proinflammatory phenotypes, whereas overexpression of constitutively active YAP was sufficient to promote EC proliferation and inflammation. In addition, treatment with statin, an antiatherosclerotic drug, inhibited YAP/TAZ activities to diminish the disturbed flow-induced proliferation and inflammation. In vivo blockade of YAP/TAZ translation by morpholino oligos significantly reduced endothelial inflammation and the size of atherosclerotic lesions. Our results demonstrate a critical role of the activation of YAP/TAZ by disturbed flow in promoting atheroprone phenotypes and atherosclerotic lesion development. Therefore, inhibition of YAP/TAZ activation is a promising athero-protective therapeutic strategy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Rheology , Animals , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Carotid Artery, Common/pathology , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Connective Tissue Growth Factor/metabolism , Cysteine-Rich Protein 61/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/pathology , Mice , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Phenotype , Repressor Proteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
16.
Sci Rep ; 6: 22147, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26915560

ABSTRACT

Little is known regarding the interplays between the mechanical and molecular bases for vein graft restenosis. We elucidated the stenosis initiation using a high-frequency ultrasonic (HFU) echogenicity platform and estimated the endothelium yield stress from von-Mises stress computation to predict the damage locations in living rats over time. The venous-arterial transition induced the molecular cascades for autophagy and apoptosis in venous endothelial cells (ECs) to cause neointimal hyperplasia, which correlated with the high echogenicity in HFU images and the large mechanical stress that exceeded the yield strength. The ex vivo perfusion of arterial laminar shear stress to isolated veins further confirmed the correlation. EC damage can be rescued by inhibiting autophagy formation using 3-methyladenine (3-MA). Pretreatment of veins with 3-MA prior to grafting reduced the pathological increases of echogenicity and neointima formation in rats. Therefore, this platform provides non-invasive temporal spatial measurement and prediction of restenosis after venous-arterial transition as well as monitoring the progression of the treatments.


Subject(s)
Adenine/analogs & derivatives , Apoptosis/physiology , Autophagy/physiology , Graft Occlusion, Vascular/prevention & control , Neointima/pathology , Neointima/prevention & control , Adenine/therapeutic use , Animals , Arteries/surgery , Endothelial Cells/pathology , Graft Occlusion, Vascular/pathology , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Veins/pathology , Veins/surgery
17.
Proc Natl Acad Sci U S A ; 113(8): 2206-11, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858399

ABSTRACT

The functional maturation and preservation of hepatic cells derived from human induced pluripotent stem cells (hiPSCs) are essential to personalized in vitro drug screening and disease study. Major liver functions are tightly linked to the 3D assembly of hepatocytes, with the supporting cell types from both endodermal and mesodermal origins in a hexagonal lobule unit. Although there are many reports on functional 2D cell differentiation, few studies have demonstrated the in vitro maturation of hiPSC-derived hepatic progenitor cells (hiPSC-HPCs) in a 3D environment that depicts the physiologically relevant cell combination and microarchitecture. The application of rapid, digital 3D bioprinting to tissue engineering has allowed 3D patterning of multiple cell types in a predefined biomimetic manner. Here we present a 3D hydrogel-based triculture model that embeds hiPSC-HPCs with human umbilical vein endothelial cells and adipose-derived stem cells in a microscale hexagonal architecture. In comparison with 2D monolayer culture and a 3D HPC-only model, our 3D triculture model shows both phenotypic and functional enhancements in the hiPSC-HPCs over weeks of in vitro culture. Specifically, we find improved morphological organization, higher liver-specific gene expression levels, increased metabolic product secretion, and enhanced cytochrome P450 induction. The application of bioprinting technology in tissue engineering enables the development of a 3D biomimetic liver model that recapitulates the native liver module architecture and could be used for various applications such as early drug screening and disease modeling.


Subject(s)
Bioprinting/methods , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Liver/anatomy & histology , Printing, Three-Dimensional , Albumins/biosynthesis , Biomimetics/methods , Cell Culture Techniques , Cell Differentiation , Gene Expression , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Liver/cytology , Tissue Engineering/methods
18.
Stroke ; 46(4): 1099-106, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25712942

ABSTRACT

BACKGROUND AND PURPOSE: Endovascular thrombectomy has shown promise for the treatment of acute strokes resulting from large-vessel occlusion. Reperfusion-related injury may contribute to the observed decoupling of angiographic and clinical outcomes. Iatrogenic disruption of the endothelium during thrombectomy is potentially a key mediator of this process that requires further study. METHODS: An in vitro live-cell platform was developed to study the effect of various commercially available endovascular devices on the endothelium. In vivo validation was performed using porcine subjects. RESULTS: This novel in vitro platform permitted high-resolution quantification and characterization of the pattern and timing of endothelial-cell injury among endovascular thrombectomy devices and vessel diameters. Thrombectomy devices displayed heterogeneous effects on the endothelium; the device performance assessed in vitro was substantiated by in vivo findings. CONCLUSIONS: In vitro live-cell artificial vessel modeling enables a detailed study of the endothelium after thrombectomy and may contribute to future device design. Large animal studies confirm the relevance of this in vitro system to investigate endothelial physiology. This artificial vessel model may represent a practical, scalable, and physiologically relevant system to assess new endovascular technologies.


Subject(s)
Endothelium, Vascular/injuries , Mechanical Thrombolysis , Stroke/therapy , Animals , Disease Models, Animal , In Vitro Techniques , Mechanical Thrombolysis/adverse effects , Mechanical Thrombolysis/instrumentation , Mechanical Thrombolysis/standards , Swine
19.
J Virol Methods ; 212: 8-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25446515

ABSTRACT

Human papillomavirus (HPV) types 16 and 18 are known to be high-risk viruses that cause cervical cancer. An HPV rapid testing kit that could help physicians to make early and more informed decisions regarding patient care is needed urgently but not yet available. This study aimed to develop a multiplex nested polymerase chain reaction-immunochromatographic test (PCR-ICT) for the rapid identification of HPV 16 and 18. A multiplex nested PCR was constructed to amplify the HPV 16 and 18 genotype-specific L1 gene fragments and followed by ICT which coated with antibodies to identify rapidly the different PCR products. The type-specific gene regions of high-risk HPV 16 and 18 could be amplified successfully by multiplex nested PCR at molecular sizes of approximately 99 and 101bp, respectively. The capture antibodies raised specifically against the moleculars labeled on the PCR products could be detected simultaneously both HPV 16 and 18 in one strip. Under optimal conditions, this PCR-ICT assay had the capability to detect HPV in a sample with as low as 100 copies of HPV viral DNA. The PCR-ICT system has the advantage of direct and simultaneous detection of two high-risk HPV 16 and 18 DNA targets in one sample, which suggested a significant potential of this assay for clinical application.


Subject(s)
Chromatography, Affinity/methods , Human papillomavirus 16/isolation & purification , Human papillomavirus 18/isolation & purification , Multiplex Polymerase Chain Reaction/methods , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , Polymerase Chain Reaction/methods , Humans , Sensitivity and Specificity
20.
Proc Natl Acad Sci U S A ; 111(28): 10347-52, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24958852

ABSTRACT

Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Subject(s)
Cardiovascular System/embryology , Embryonic Development/physiology , Endothelial Cells/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Animals , Cardiovascular System/cytology , Endothelial Cells/cytology , Ion Channels/genetics , Mice , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...