Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 560(7720): 655-660, 2018 08.
Article in English | MEDLINE | ID: mdl-30135582

ABSTRACT

Mammalian cells are surrounded by neighbouring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP (also known as YAP1) and TAZ (also known as WWTR1) are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified the Ras-related GTPase RAP2 as a key intracellular signal transducer that relays ECM rigidity signals to control mechanosensitive cellular activities through YAP and TAZ. RAP2 is activated by low ECM stiffness, and deletion of RAP2 blocks the regulation of YAP and TAZ by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate and phosphatidic acid, which activates RAP2 through PDZGEF1 and PDZGEF2 (also known as RAPGEF2 and RAPGEF6). At low stiffness, active RAP2 binds to and stimulates MAP4K4, MAP4K6, MAP4K7 and ARHGAP29, resulting in activation of LATS1 and LATS2 and inhibition of YAP and TAZ. RAP2, YAP and TAZ have pivotal roles in mechanoregulated transcription, as deletion of YAP and TAZ abolishes the ECM stiffness-responsive transcriptome. Our findings show that RAP2 is a molecular switch in mechanotransduction, thereby defining a mechanosignalling pathway from ECM stiffness to the nucleus.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rap GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Transformation, Neoplastic , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , GTPase-Activating Proteins/metabolism , Germinal Center Kinases , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Nerve Tissue Proteins/metabolism , Phospholipase C gamma/metabolism , Phosphoproteins/metabolism , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , YAP-Signaling Proteins , rap GTP-Binding Proteins/genetics
2.
Nature ; 526(7571): 118-21, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26374997

ABSTRACT

Development of functional nanoparticles can be encumbered by unanticipated material properties and biological events, which can affect nanoparticle effectiveness in complex, physiologically relevant systems. Despite the advances in bottom-up nanoengineering and surface chemistry, reductionist functionalization approaches remain inadequate in replicating the complex interfaces present in nature and cannot avoid exposure of foreign materials. Here we report on the preparation of polymeric nanoparticles enclosed in the plasma membrane of human platelets, which are a unique population of cellular fragments that adhere to a variety of disease-relevant substrates. The resulting nanoparticles possess a right-side-out unilamellar membrane coating functionalized with immunomodulatory and adhesion antigens associated with platelets. Compared to uncoated particles, the platelet membrane-cloaked nanoparticles have reduced cellular uptake by macrophage-like cells and lack particle-induced complement activation in autologous human plasma. The cloaked nanoparticles also display platelet-mimicking properties such as selective adhesion to damaged human and rodent vasculatures as well as enhanced binding to platelet-adhering pathogens. In an experimental rat model of coronary restenosis and a mouse model of systemic bacterial infection, docetaxel and vancomycin, respectively, show enhanced therapeutic efficacy when delivered by the platelet-mimetic nanoparticles. The multifaceted biointerfacing enabled by the platelet membrane cloaking method provides a new approach in developing functional nanoparticles for disease-targeted delivery.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Blood Platelets/cytology , Cell Membrane/metabolism , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Platelet Adhesiveness , Animals , Anti-Bacterial Agents/pharmacokinetics , Blood Vessels/cytology , Blood Vessels/metabolism , Blood Vessels/pathology , Collagen/chemistry , Collagen/immunology , Complement Activation/immunology , Coronary Restenosis/blood , Coronary Restenosis/drug therapy , Coronary Restenosis/metabolism , Disease Models, Animal , Docetaxel , Humans , Macrophages/immunology , Male , Mice , Polymers/chemistry , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/blood , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/cytology , Staphylococcus aureus/metabolism , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Unilamellar Liposomes/chemistry , Vancomycin/administration & dosage , Vancomycin/pharmacokinetics
3.
Curr Top Membr ; 87: 255-277, 2021.
Article in English | MEDLINE | ID: mdl-34696887

ABSTRACT

Extracellular signaling molecules, such as growth factors, cytokines, and hormones, regulate cell behaviors and fate through endocrine, paracrine, and autocrine actions and play essential roles in maintaining tissue homeostasis. MicroRNAs, an important class of posttranscriptional modulators, could stably present in extracellular space and body fluids and participate in intercellular communication in health and diseases. Indeed, recent studies demonstrated that microRNAs could be secreted through vesicular and non-vesicular routes, transported in body fluids, and then transmitted to recipient cells to regulate target gene expression and signaling events. Over the past decade, a great deal of effort has been made to investigate the functional roles of extracellular vesicles and extracellular microRNAs in pathological conditions. Emerging evidence suggests that altered levels of extracellular vesicles and extracellular microRNAs in body fluids, as part of the cellular responses to atherogenic factors, are associated with the development of atherosclerosis. This review article provides a brief overview of extracellular vesicles and perspectives of their applications as therapeutic tools for cardiovascular pathologies. In addition, we highlight the role of extracellular microRNAs in atherogenesis and offer a summary of circulating microRNAs in liquid biopsies associated with atherosclerosis.


Subject(s)
Atherosclerosis , Extracellular Vesicles , MicroRNAs , Atherosclerosis/genetics , Cell Communication , Humans , MicroRNAs/genetics , Signal Transduction
4.
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
5.
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
6.
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
7.
Proc Natl Acad Sci U S A ; 113(52): 15078-15083, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27965388

ABSTRACT

Toll-like receptor-mediated NF-κB activation is a major innate immune reaction of vascular endothelial cells (ECs) in response to prooxidative and proinflammatory stimuli. We identified that TNF-α receptor-associated factor-interacting protein with a forkhead-associated domain (TIFA) is a regulator of priming (signal 1) and activating (signal 2) signals of nucleotide oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome in ECs. Oxidative and inflammatory stresses such as atheroprone flow and hyperlipidemia induce and activate TIFA in vitro and in vivo. For the priming of signal 1, sterol regulatory element-binding protein 2 transactivates TIFA, which in turn induces NF-κB activation and augments the transcription of NLRP3 inflammasome components. For the activation of signal 2, Akt is involved in TIFA Thr9 phosphorylation, which is essential for TIFA-TIFA homophilic oligomerization. Thr9 phosphorylation-dependent TIFA oligomerization facilitates the higher-order assembly of NLRP3 inflammasome, as indicated by the interaction between TIFA and caspase-1 in the activated ECs. Our results suggest that TIFA is a crucial mediator in the endothelial innate immune response by potentiating and amplifying NLRP3 inflammasome via augmenting signals 1 and 2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Apolipoproteins E/genetics , Endothelium/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunity, Innate , Inflammation , Lung/metabolism , Male , Mice , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 2/metabolism , Transcription, Genetic
8.
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
9.
Circ Res ; 113(1): 40-51, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23603512

ABSTRACT

RATIONALE: Endothelial microRNA-126 (miR-126) modulates vascular development and angiogenesis. However, its role in the regulation of smooth muscle cell (SMC) function is unknown. OBJECTIVE: To elucidate the role of miR-126 secreted by endothelial cells (ECs) in regulating SMC turnover in vitro and in vivo, as well as the effects of shear stress on the regulation. METHODS AND RESULTS: Coculture of SMCs with ECs or treatment of SMCs with conditioned media from static EC monoculture (EC-CM) increased SMC miR-126 level and SMC turnover; these effects were abolished by inhibition of endothelial miR-126 and by the application of laminar shear stress to ECs. SMC miR-126 did not increase when treated with EC-CM from ECs subjected to inhibition of miR biogenesis, or with CM from sheared ECs. Depletion of extracellular/secreted vesicles in EC-CM did not affect the increase of SMC miR-126 by EC-CM. Biotinylated miR-126 or FLAG (DYKDDDDK epitope)-tagged Argonaute2 transfected into ECs was detected in the cocultured or EC-CM-treated SMCs, indicating a direct EC-to-SMC transmission of miR-126 and Argonaute2. Endothelial miR-126 represses forkhead box O3, B-cell lymphoma 2, and insulin receptor substrate 1 mRNAs in the cocultured SMCs, suggesting the functional roles of the transmitted miR-126. Systemic depletion of miR-126 in mice inhibited neointimal lesion formation of carotid arteries induced by cessation of blood flow. Administration of EC-CM or miR-126 mitigated the inhibitory effect. CONCLUSIONS: Endothelial miR-126 acts as a key intercellular mediator to increase SMC turnover, and its release is reduced by atheroprotective laminar shear stress.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/physiology , Myocytes, Smooth Muscle/cytology , Animals , Argonaute Proteins/genetics , Argonaute Proteins/physiology , Carotid Artery, Common/pathology , Cell Culture Techniques/instrumentation , Cell Line , Coculture Techniques , Culture Media, Conditioned/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Silencing , Genes, bcl-2 , Hemorheology , Human Umbilical Vein Endothelial Cells , Humans , Insulin Receptor Substrate Proteins/biosynthesis , Insulin Receptor Substrate Proteins/genetics , Ligation , Mice , MicroRNAs/genetics , MicroRNAs/therapeutic use , Muscle, Smooth, Vascular/cytology , Neointima , Paracrine Communication , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Recombinant Fusion Proteins/physiology , Umbilical Arteries/cytology
10.
Arterioscler Thromb Vasc Biol ; 34(7): 1437-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24855060

ABSTRACT

OBJECTIVE: The site-specificity of endothelial phenotype is attributable to the local hemodynamic forces. The flow regulation of microRNAs in endothelial cells (ECs) plays a significant role in vascular homeostasis and diseases. The objective of this study was to elucidate the molecular mechanism by which the pulsatile shear flow-induced microRNA-23b (miR-23b) exerts antiproliferative effects on ECs. APPROACH AND RESULTS: We used a combination of a cell perfusion system and experimental animals to examine the flow regulation of miR-23b in modulating EC proliferation. Our results demonstrated that pulsatile shear flow induces the transcription factor Krüppel-like factor 2 to promote miR-23b biosynthesis; the increase in miR-23b then represses cyclin H to impair the activity and integrity of cyclin-dependent kinase-activating kinase (CAK) complex. The inhibitory effect of miR-23b on CAK exerts dual actions to suppress cell cycle progression, and reduce basal transcription by deactivating RNA polymerase II. Whereas pulsatile shear flow regulates the miR-23b/CAK pathway to exert antiproliferative effects on ECs, oscillatory shear flow has little effect on the miR-23b/CAK pathway and hence does not cause EC growth arrest. Such flow pattern-dependent phenomena are validated with an in vivo model on rat carotid artery: the flow disturbance induced by partial carotid ligation led to a lower expression of miR-23b and a higher EC proliferation in comparison with the pulsatile flow regions of the unligated vessels. Local delivery of miR-23b mitigated the proliferative EC phenotype in partially ligated vessels. CONCLUSIONS: Our findings unveil a novel mechanism by which hemodynamic forces modulate EC proliferative phenotype through the miR-23b/CAK pathway.


Subject(s)
Carotid Artery Diseases/enzymology , Cell Proliferation , Cyclin H/metabolism , Cyclin-Dependent Kinases/metabolism , Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , MicroRNAs/metabolism , Transcription, Genetic , Animals , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Cell Cycle Checkpoints , Cells, Cultured , Cyclin H/genetics , Cyclin-Dependent Kinases/genetics , Disease Models, Animal , Humans , Kruppel-Like Transcription Factors/metabolism , Male , Mechanotransduction, Cellular , MicroRNAs/genetics , Perfusion , Phenotype , Pulsatile Flow , RNA Interference , RNA Polymerase II/metabolism , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection , Cyclin-Dependent Kinase-Activating Kinase
11.
Proc Natl Acad Sci U S A ; 109(28): 11110-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22665785

ABSTRACT

We use a novel 3D inter-/intracellular force microscopy technique based on 3D traction force microscopy to measure the cell-cell junctional and intracellular tensions in subconfluent and confluent vascular endothelial cell (EC) monolayers under static and shear flow conditions. We found that z-direction cell-cell junctional tensions are higher in confluent EC monolayers than those in subconfluent ECs, which cannot be revealed in the previous 2D methods. Under static conditions, subconfluent cells are under spatially non-uniform tensions, whereas cells in confluent monolayers are under uniform tensions. The shear modulations of EC cytoskeletal remodeling, extracellular matrix (ECM) adhesions, and cell-cell junctions lead to significant changes in intracellular tensions. When a confluent monolayer is subjected to flow shear stresses with a high forward component comparable to that seen in the straight part of the arterial system, the intracellular and junction tensions preferentially increase along the flow direction over time, which may be related to the relocation of adherens junction proteins. The increases in intracellular tensions are shown to be a result of chemo-mechanical responses of the ECs under flow shear rather than a direct result of mechanical loading. In contrast, the intracellular tensions do not show a preferential orientation under oscillatory flow with a very low mean shear. These differences in the directionality and magnitude of intracellular tensions may modulate translation and transcription of ECs under different flow patterns, thus affecting their susceptibility for atherogenesis.


Subject(s)
Endothelial Cells/cytology , Animals , Atherosclerosis , Cell Communication , Cells, Cultured/cytology , Extracellular Matrix/metabolism , Finite Element Analysis , Fluorescence Resonance Energy Transfer , Humans , Imaging, Three-Dimensional , Microscopy, Atomic Force/methods , Microscopy, Confocal/methods , Models, Biological , Models, Statistical , Oscillometry/methods , Shear Strength
12.
Proc Natl Acad Sci U S A ; 108(25): 10355-60, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21636785

ABSTRACT

Adhesion of circulating monocytes to vascular endothelial cells (ECs) is a critical event leading to vascular inflammation and, hence, development of atherosclerosis. MicroRNAs (miRs) are a class of endogenous, highly conserved, noncoding small RNAs that play important roles in regulating gene expression and cellular function, as well as pathogenesis of atherosclerosis. Here, we showed that oscillatory shear stress (OSS) induces the expression of miR-21 at the transcriptional level in cultured human umbilical vein ECs via an increased binding of c-Jun, which is a component of transcription factor activator protein-1 (AP-1), to the promoter region of miR-21. OSS induction of miR-21 inhibited the translation, but not transcription, of peroxisome proliferators-activated receptor-α (PPARα) by 3'-UTR targeting. Overexpression of miR-21 up-regulated AP-1 activation, which was attenuated by exogenous expression of PPARα. OSS and overexpression of miR-21 enhanced the expression of adhesion molecules vascular cell adhesion molecule-1 and monocyte chemotactic protein-1 and the consequential adhesion of monocytes to ECs. Overexpression of PPARα significantly attenuated the AP-1-mediated miR-21 expression. These results demonstrate a unique mechanism by which OSS induces AP-1-dependent miR-21 expression, which directly targets PPARα to inhibit its expression, thereby allowing activation of AP-1 and the promotion of monocyte adhesion. Our findings suggest the presence of a positive feedback loop that enables the sustained induction of miR-21, thus contributing to the proinflammatory responses of vascular endothelium under OSS.


Subject(s)
Endothelium, Vascular/pathology , Feedback, Physiological/physiology , Inflammation/metabolism , MicroRNAs/metabolism , PPAR alpha/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Inflammation/pathology , MicroRNAs/genetics , PPAR alpha/genetics , Stress, Mechanical , Transcription Factor AP-1/metabolism , Transcriptional Activation
13.
Biomaterials ; 305: 122428, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38147743

ABSTRACT

Cancer thrives in a complex environment where interactions between cellular and acellular components, surrounding the tumor, play a crucial role in disease development and progression. Despite significant progress in cancer research, the mechanism driving tumor growth and therapeutic outcomes remains elusive. Two-dimensional (2D) cell culture assays and in vivo animal models are commonly used in cancer research and therapeutic testing. However, these models suffer from numerous shortcomings including lack of key features of the tumor microenvironment (TME) & cellular composition, cost, and ethical clearance. To that end, there is an increased interest in incorporating and elucidating the influence of TME on cancer progression. Advancements in 3D-engineered ex vivo models, leveraging biomaterials and microengineering technologies, have provided an unprecedented ability to reconstruct native-like bioengineered cancer models to study the heterotypic interactions of TME with a spatiotemporal organization. These bioengineered cancer models have shown excellent capabilities to bridge the gap between oversimplified 2D systems and animal models. In this review article, we primarily provide an overview of the immune and stromal cellular components of the TME and then discuss the latest state-of-the-art 3D-engineered ex vivo platforms aiming to recapitulate the complex TME features. The engineered TME model, discussed herein, are categorized into three main sections according to the cellular interactions within TME: (i) Tumor-Stromal interactions, (ii) Tumor-Immune interactions, and (iii) Complex TME interactions. Finally, we will conclude the article with a perspective on how these models can be instrumental for cancer translational studies and therapeutic testing.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Neoplasms/pathology , Cell Culture Techniques/methods , Biocompatible Materials , Cell Communication
14.
Biomaterials ; 306: 122505, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38359507

ABSTRACT

Atherosclerosis, a chronic inflammatory disease, is the primary cause of myocardial infarction and ischemic stroke. Recent studies have demonstrated that dysregulation of yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding domain (TAZ) contributes to plaque development, making YAP/TAZ potential therapeutic targets. However, systemic modulation of YAP/TAZ expression or activities risks serious off-target effects, limiting clinical applicability. To address the challenge, this study develops monocyte membrane-coated nanoparticles (MoNP) as a targeted delivery system for activated and inflamed endothelium lining the plaque surface. The MoNP system is used to deliver verteporfin (VP), aimed at inhibiting YAP/TAZ specifically within arterial regions prone to atherosclerosis. The results reveal that MoNP significantly enhance payload delivery to inflamed endothelial cells (EC) while avoiding phagocytic cells. When administered in mice, MoNP predominantly accumulate in intima of the atheroprone artery. MoNP-mediated delivery of VP substantially reduces YAP/TAZ expression, thereby suppressing inflammatory gene expression and macrophage infiltration in cultured EC and mouse arteries exposed to atherogenic stimuli. Importantly, this targeted VP nanodrug effectively decreases plaque development in mice without causing noticeable histopathological changes in major organs. Collectively, these findings demonstrate a lesion-targeted and pathway-specific biomimetic nanodrug, potentially leading to safer and more effective treatments for atherosclerosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Trans-Activators/metabolism , YAP-Signaling Proteins , Endothelial Cells/metabolism , Biomimetics , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Inflammation/drug therapy
15.
Proc Natl Acad Sci U S A ; 107(7): 3234-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133741

ABSTRACT

MicroRNAs (miRs) can regulate many cellular functions, but their roles in regulating responses of vascular endothelial cells (ECs) to mechanical stimuli remain unexplored. We hypothesize that the physiological responses of ECs are regulated by not only mRNA and protein signaling networks, but also expression of the corresponding miRs. EC growth arrest induced by pulsatile shear (PS) flow is an important feature for flow regulation of ECs. miR profiling showed that 21 miRs are differentially expressed (8 up- and 13 downregulated) in response to 24-h PS as compared to static condition (ST). The mRNA expression profile indicates EC growth arrest under 24-h PS. Analysis of differentially expressed miRs yielded 68 predicted mRNA targets that overlapped with results of microarray mRNA profiling. Functional analysis of miR profile indicates that the cell cycle network is highly regulated. The upregulation of miR-23b and miR-27b was found to correlate with the PS-induced EC growth arrest. Inhibition of miR-23b using antagomir-23b oligonucleotide (AM23b) reversed the PS-induced E2F1 reduction and retinoblastoma (Rb) hypophosphorylation and attenuated the PS-induced G1/G0 arrest. Antagomir AM27b regulated E2F1 expression, but did not affect Rb and growth arrest. Our findings indicate that PS suppresses EC proliferation through the regulation of miR-23b and provide insights into the role of miRs in mechanotransduction.


Subject(s)
Endothelial Cells/physiology , Gene Expression Regulation/physiology , MicroRNAs/physiology , Pulsatile Flow/physiology , Retinoblastoma Protein/metabolism , Analysis of Variance , Bromodeoxyuridine , Cell Proliferation , Flow Cytometry , Humans , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
16.
bioRxiv ; 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37163067

ABSTRACT

Atherosclerosis, characterized by the buildup of lipid-rich plaque on the vessel wall, is the primary cause of myocardial infarction and ischemic stroke. Recent studies have demonstrated that dysregulation of yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding domain (TAZ) contributes to plaque development, making YAP/TAZ potential therapeutic targets. However, systemic modulation of YAP/TAZ expression or activities risks serious off-target effects, limiting clinical applicability. To address the challenge, this study develops monocyte membrane-coated nanoparticles (MoNP) as a drug delivery vehicle targeting activated endothelium lining the plaque surface and utilizes MoNP to deliver verteporfin (VP), a potent YAP/TAZ inhibitor, for lesion-specific treatment of atherosclerosis. The results reveal that MoNP significantly enhance payload delivery to inflamed endothelial cells (EC) while avoiding phagocytic cells, and preferentially accumulate in atherosclerotic regions. MoNP-mediated delivery of VP substantially reduces YAP/TAZ expression, suppressing inflammatory gene expression and macrophage infiltration in cultured EC and mouse arteries exposed to atherogenic stimuli. Importantly, this lesion-targeted VP nanodrug effectively decreases plaque development in mice without causing noticeable histopathological changes in major organs. Collectively, these findings demonstrate a plaque-targeted and pathway-specific biomimetic nanodrug, potentially leading to safer and more effective treatments for atherosclerosis.

17.
Circulation ; 124(5): 633-41, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21768538

ABSTRACT

BACKGROUND: Upregulated by atheroprotective flow, the transcription factor Krüppel-like factor 2 (KLF2) is crucial for maintaining endothelial function. MicroRNAs (miRNAs) are noncoding small RNAs that regulate gene expression at the posttranscriptional level. We examined the role of miRNAs, particularly miR-92a, in the atheroprotective flow-regulated KLF2. METHODS AND RESULTS: Dicer knockdown increased the level of KLF2 mRNA in human umbilical vein endothelial cells, suggesting that KLF2 is regulated by miRNA. In silico analysis predicted that miR-92a could bind to the 3' untranslated region of KLF2 mRNA. Overexpression of miR-92a decreased the expression of KLF2 and the KLF2-regulated endothelial nitric oxide synthase and thrombomodulin at mRNA and protein levels. A complementary finding is that miR-92a inhibitor increased the mRNA and protein expression of KLF2, endothelial nitric oxide synthase, and thrombomodulin. Subsequent studies revealed that atheroprotective laminar flow downregulated the level of miR-92a precursor to induce KLF2, and the level of this flow-induced KLF2 was reduced by miR-92a precursor. Furthermore, miR-92a level was lower in human umbilical vein endothelial cells exposed to the atheroprotective pulsatile shear flow than under atheroprone oscillatory shear flow. Anti-Ago1/2 immunoprecipitation coupled with real-time polymerase chain reaction revealed that pulsatile shear flow decreased the functional targeting of miR-92a precursor/KLF2 mRNA in human umbilical vein endothelial cells. Consistent with these findings, mouse carotid arteries receiving miR-92a precursor exhibited impaired vasodilatory response to flow. CONCLUSIONS: Atheroprotective flow patterns decrease the level of miR-92a, which in turn increases KLF2 expression to maintain endothelial homeostasis.


Subject(s)
Atherosclerosis/physiopathology , Endothelial Cells/physiology , Kruppel-Like Transcription Factors/genetics , MicroRNAs/physiology , Pulsatile Flow/physiology , 3' Untranslated Regions/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Endothelial Cells/cytology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Homeostasis/physiology , Humans , Kruppel-Like Transcription Factors/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , Stress, Mechanical , Thrombomodulin/genetics , Thrombomodulin/metabolism , Umbilical Veins/cytology , Vasodilation/physiology
18.
Front Cell Dev Biol ; 9: 673599, 2021.
Article in English | MEDLINE | ID: mdl-34109179

ABSTRACT

Biophysical cues, such as mechanical properties, play a critical role in tissue growth and homeostasis. During organ development and tissue injury repair, compressive and tensional forces generated by cell-extracellular matrix or cell-cell interaction are key factors for cell fate determination. In the vascular system, hemodynamic forces, shear stress, and cyclic stretch modulate vascular cell phenotypes and susceptibility to atherosclerosis. Despite that emerging efforts have been made to investigate how mechanotransduction is involved in tuning cell and tissue functions in various contexts, the regulatory mechanisms remain largely unknown. One of the challenges is to understand the signaling cascades that transmit mechanical cues from the plasma membrane to the cytoplasm and then to the nuclei to generate mechanoresponsive transcriptomes. YAP and its homolog TAZ, the Hippo pathway effectors, have been identified as key mechanotransducers that sense mechanical stimuli and relay the signals to control transcriptional programs for cell proliferation, differentiation, and transformation. However, the upstream mechanosensors for YAP/TAZ signaling and downstream transcriptome responses following YAP/TAZ activation or repression have not been well characterized. Moreover, the mechanoregulation of YAP/TAZ in literature is highly context-dependent. In this review, we summarize the biomechanical cues in the tissue microenvironment and provide an update on the roles of YAP/TAZ in mechanotransduction in various physiological and pathological conditions.

19.
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
20.
World J Gastroenterol ; 12(10): 1551-7, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16570347

ABSTRACT

AIM: To study the baculovirus/mammalian cell system for efficient expression of functional large hepatitis delta antigen (L-HDAg). METHODS: A recombinant baculovirus expressing histidine-tagged L-HDAg (L-HDAgH) was constructed to transduce baby hamster kidney (BHK) cells by a simplified transduction protocol. RESULTS: The recombinant baculovirus transduced BHK cells with efficiencies higher than 90% as determined by flow cytometry. The expression level was significantly higher than that obtained by plasmid transfection and was further enhanced 3-fold to around 19 pg/cell by the addition of 10 mmol/L sodium butyrate. Importantly, the expressed L-HDAgH was localized to the cell nucleus and correctly isoprenylated as determined by immunofluorescence labeling and confocal microscopy. Moreover, L-HDAgH interacted with hepatitis B surface antigen to form virus-like particles. CONCLUSION: The fusion with histidine tags as well as overexpression of L-HDAgH in the baculovirus-transduced BHK cells does not impair the biological functions. Taken together, the baculovirus/mammalian cell system offers an attractive alternative for high level expression of L-HDAgH or other proteins that require extensive post-translational modifications.


Subject(s)
Baculoviridae/genetics , Hepatitis delta Antigens/genetics , Protein Processing, Post-Translational , Transduction, Genetic , Animals , Cells, Cultured , Cricetinae , DNA, Recombinant , DNA, Viral/analysis , Hepatitis Viruses/genetics , Hepatitis Viruses/immunology , Histidine , Kidney , Plasmids , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL