Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Proc Natl Acad Sci U S A ; 121(5): e2318904121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38261622

ABSTRACT

Flow patterns exert significant effects on vascular endothelial cells (ECs) to lead to the focal nature of atherosclerosis. Using a step flow chamber to investigate the effects of disturbed shear (DS) and pulsatile shear (PS) on ECs in the same flow channel, we conducted single-cell RNA sequencing analyses to explore the distinct transcriptomic profiles regulated by DS vs. PS. Integrated analysis identified eight cell clusters and demonstrated that DS induces EC transition from atheroprotective to proatherogenic phenotypes. Using an automated cell type annotation algorithm (SingleR), we showed that DS promoted endothelial-to-mesenchymal transition (EndMT) by inducing the transcriptional phenotypes for inflammation, hypoxia responses, transforming growth factor-beta (TGF-ß) signaling, glycolysis, and fatty acid synthesis. Enolase 1 (ENO1), a key gene in glycolysis, was one of the top-ranked genes in the DS-induced EndMT cluster. Pseudotime trajectory analysis revealed that the kinetic expression of ENO1 was significantly associated with EndMT and that ENO1 silencing repressed the DS- and TGF-ß-induced EC inflammation and EndMT. Consistent with these findings, ENO1 was highly expressed in ECs at the inner curvature of the mouse aortic arch (which is exposed to DS) and atherosclerotic lesions, suggesting its proatherogenic role in vivo. In summary, we present a comprehensive single-cell atlas of ECs in response to different flow patterns within the same flow channel. Among the DS-regulated genes, ENO1 plays an important role in DS-induced EC inflammation and EndMT. These results provide insights into how hemodynamic forces regulate vascular endothelium in health and disease.


Subject(s)
Atherosclerosis , Endothelial Cells , Animals , Mice , Gene Expression Profiling , Inflammation , Sequence Analysis, RNA , Transforming Growth Factor beta
2.
Annu Rev Biomed Eng ; 25: 157-184, 2023 06 08.
Article in English | MEDLINE | ID: mdl-36913673

ABSTRACT

The central dogma of gene expression involves DNA transcription to RNA and RNA translation into protein. As key intermediaries and modifiers, RNAs undergo various forms of modifications such as methylation, pseudouridylation, deamination, and hydroxylation. These modifications, termed epitranscriptional regulations, lead to functional changes in RNAs. Recent studies have demonstrated crucial roles for RNA modifications in gene translation, DNA damage response, and cell fate regulation. Epitranscriptional modifications play an essential role in development, mechanosensing, atherogenesis, and regeneration in the cardiovascular (CV) system, and their elucidation is critically important to understanding the molecular mechanisms underlying CV physiology and pathophysiology. This review aims at providing biomedical engineers with an overview of the epitranscriptome landscape, related key concepts, recent findings in epitranscriptional regulations, and tools for epitranscriptome analysis. The potential applications of this important field in biomedical engineering research are discussed.


Subject(s)
Biomedical Engineering , Cardiovascular System , Humans , RNA/genetics , RNA/metabolism , Gene Expression Regulation , Bioengineering
3.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468662

ABSTRACT

The two main blood flow patterns, namely, pulsatile shear (PS) prevalent in straight segments of arteries and oscillatory shear (OS) observed at branch points, are associated with atheroprotective (healthy) and atheroprone (unhealthy) vascular phenotypes, respectively. The effects of blood flow-induced shear stress on endothelial cells (ECs) and vascular health have generally been studied using human umbilical vein endothelial cells (HUVECs). While there are a few studies comparing the differential roles of PS and OS across different types of ECs at a single time point, there is a paucity of studies comparing the temporal responses between different EC types. In the current study, we measured OS and PS transcriptomic responses in human aortic endothelial cells (HAECs) over 24 h and compared these temporal responses of HAECs with our previous findings on HUVECs. The measurements were made at 1, 4, and 24 h in order to capture the responses at early, mid, and late time points after shearing. The results indicate that the responses of HAECs and HUVECs are qualitatively similar for endothelial function-relevant genes and several important pathways with a few exceptions, thus demonstrating that HUVECs can be used as a model to investigate the effects of shear on arterial ECs, with consideration of the differences. Our findings show that HAECs exhibit an earlier response or faster kinetics as compared to HUVECs. The comparative analysis of HAECs and HUVECs presented here offers insights into the mechanisms of common and disparate shear stress responses across these two major endothelial cell types.


Subject(s)
Cell Cycle/genetics , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Metabolic Networks and Pathways/genetics , Proteome/genetics , Stress, Mechanical , Transcription Factors/genetics , Aorta/cytology , Aorta/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line , Cell Proliferation , Endothelial Cells/cytology , Gene Expression Profiling , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Humans , Models, Biological , Organ Specificity , Phenotype , Proteome/metabolism , Signal Transduction , Systems Biology/methods , Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33579825

ABSTRACT

Atherosclerosis is characterized by the plaque formation that restricts intraarterial blood flow. The disturbed blood flow with the associated oscillatory stress (OS) at the arterial curvatures and branch points can trigger endothelial activation and is one of the risk factors of atherosclerosis. Many studies reported the mechanotransduction related to OS and atherogenesis; however, the transcriptional and posttranscriptional regulatory mechanisms of atherosclerosis remain unclear. Herein, we investigated the role of N6-methyladenosine (m6A) RNA methylation in mechanotransduction in endothelial cells (ECs) because of its important role in epitranscriptome regulation. We have identified m6A methyltransferase METTL3 as a responsive hub to hemodynamic forces and atherogenic stimuli in ECs. OS led to an up-regulation of METTL3 expression, accompanied by m6A RNA hypermethylation, increased NF-κB p65 Ser536 phosphorylation, and enhanced monocyte adhesion. Knockdown of METTL3 abrogated this OS-induced m6A RNA hypermethylation and other manifestations, while METTL3 overexpression led to changes resembling the OS effects. RNA-sequencing and m6A-enhanced cross-linking and immunoprecipitation (eCLIP) experiments revealed NLRP1 and KLF4 as two hemodynamics-related downstream targets of METTL3-mediated hypermethylation. The METTL3-mediated RNA hypermethylation up-regulated NLRP1 transcript and down-regulated KLF4 transcript through YTHDF1 and YTHDF2 m6A reader proteins, respectively. In the in vivo atherosclerosis model, partial ligation of the carotid artery led to plaque formation and up-regulation of METTL3 and NLRP1, with down-regulation of KLF4; knockdown of METTL3 via repetitive shRNA administration prevented the atherogenic process, NLRP3 up-regulation, and KLF4 down-regulation. Collectively, we have demonstrated that METTL3 serves a central role in the atherogenesis induced by OS and disturbed blood flow.


Subject(s)
Adenosine/analogs & derivatives , Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Methyltransferases/metabolism , RNA Processing, Post-Transcriptional , Adenosine/metabolism , Animals , Atherosclerosis/genetics , Endothelium, Vascular/pathology , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , THP-1 Cells , Transcriptome
5.
Proc Natl Acad Sci U S A ; 114(41): 10990-10995, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973892

ABSTRACT

Blood flow and vascular shear stress patterns play a significant role in inducing and modulating physiological responses of endothelial cells (ECs). Pulsatile shear (PS) is associated with an atheroprotective endothelial phenotype, while oscillatory shear (OS) is associated with an atheroprone endothelial phenotype. Although mechanisms of endothelial shear response have been extensively studied, most studies focus on characterization of single molecular pathways, mainly at fixed time points after stress application. Here, we carried out a longitudinal time-series study to measure the transcriptome after the application of PS and OS. We performed systems analyses of transcriptional data of cultured human vascular ECs to elucidate the dynamics of endothelial responses in several functional pathways such as cell cycle, oxidative stress, and inflammation. By combining the temporal data on differentially expressed transcription factors and their targets with existing knowledge on relevant functional pathways, we infer the causal relationships between disparate endothelial functions through common transcriptional regulation mechanisms. Our study presents a comprehensive temporally longitudinal experimental study and mechanistic model of shear stress response. By comparing the relative endothelial expressions of genes between OS and PS, we provide insights and an integrated perspective into EC function in response to differential shear. This study has significant implications for the pathogenesis of vascular diseases.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Pulsatile Flow , Stress, Mechanical , Systems Biology/methods , Transcriptome , Cell Cycle , Cells, Cultured , Epithelial-Mesenchymal Transition , Humans , Inflammation , Oxidative Stress , Transcription Factors/genetics
6.
J Biol Chem ; 288(44): 31853-66, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24045946

ABSTRACT

Histone deacetylase 3 (HDAC3) plays a critical role in the maintenance of endothelial integrity and other physiological processes. In this study, we demonstrated that HDAC3 undergoes unconventional splicing during stem cell differentiation. Four different splicing variants have been identified, designated as HD3α, -ß, -γ, and -δ, respectively. HD3α was confirmed in stem cell differentiation by specific antibody against the sequences from intron 12. Immunofluorescence staining indicated that the HD3α isoform co-localized with CD31-positive or α-smooth muscle actin-positive cells at different developmental stages of mouse embryos. Overexpression of HD3α reprogrammed human aortic endothelial cells into mesenchymal cells featuring an endothelial-to-mesenchymal transition (EndMT) phenotype. HD3α directly interacts with HDAC3 and Akt1 and selectively activates transforming growth factor ß2 (TGFß2) secretion and cleavage. TGFß2 functioned as an autocrine and/or paracrine EndMT factor. The HD3α-induced EndMT was both PI3K/Akt- and TGFß2-dependent. This study provides the first evidence of the role of HDAC3 splicing in the maintenance of endothelial integrity.


Subject(s)
Alternative Splicing/physiology , Autocrine Communication/physiology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Enzymologic/physiology , Histone Deacetylases/biosynthesis , Paracrine Communication/physiology , Transforming Growth Factor beta2/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endothelial Cells/cytology , Histone Deacetylases/genetics , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta2/genetics
7.
Circulation ; 127(16): 1712-22, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23529610

ABSTRACT

BACKGROUND: Vascular endothelial cell growth factor plays a pivotal role in angiogenesis via regulating endothelial cell proliferation. The X-box binding protein 1 (XBP1) is believed to be a signal transducer in the endoplasmic reticulum stress response. It is unknown whether there is crosstalk between vascular endothelial cell growth factor signaling and XBP1 pathway. METHODS AND RESULTS: We found that vascular endothelial cell growth factor induced the kinase insert domain receptor internalization and interaction through C-terminal domain with the unspliced XBP1 and the inositol requiring enzyme 1 α in the endoplasmic reticulum, leading to inositol requiring enzyme 1 α phosphorylation and XBP1 mRNA splicing, which was abolished by siRNA-mediated knockdown of kinase insert domain receptor. Spliced XBP1 regulated endothelial cell proliferation in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-dependent manner and modulated the cell size increase in a PI3K/Akt/GSK3ß/ß-catenin/E2F2-independent manner. Knockdown of XBP1 or inositol requiring enzyme 1 α decreased endothelial cell proliferation via suppression of Akt/GSK3ß phosphorylation, ß-catenin nuclear translocation, and E2F2 expression. Endothelial cell-specific knockout of XBP1 (XBP1ecko) in mice retarded the retinal vasculogenesis in the first 2 postnatal weeks and impaired the angiogenesis triggered by ischemia. Reconstitution of XBP1 by Ad-XBP1s gene transfer significantly improved angiogenesis in ischemic tissue in XBP1ecko mice. Transplantation of bone marrow from wild-type o XBP1ecko mice could also slightly improve the foot blood reperfusion in ischemic XBP1ecko mice. CONCLUSIONS: These results suggest that XBP1 can function via growth factor signaling pathways to regulate endothelial proliferation and angiogenesis.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/agonists , Animals , Aorta/cytology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryo, Mammalian/blood supply , Endoplasmic Reticulum Stress/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/physiopathology , Ischemia/therapy , Leg/blood supply , Mice , Mice, Knockout , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA Splicing/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Chimera , Regulatory Factor X Transcription Factors , Retinal Vessels/drug effects , Retinal Vessels/growth & development , Transcription Factors/deficiency , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/physiology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/physiology , X-Box Binding Protein 1
8.
FASEB J ; 16(2): 216-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11744620

ABSTRACT

Vascular endothelium transduces the temporal gradients in shear stress (tau) originating from unsteady blood flow into functional responses. We measured the effects of step-tau and ramp-tau (i.e., t with different temporal shear gradients) on the lipid lateral diffusion coefficient (D) in the apical membranes of confluent cultured bovine aortic endothelial cells by using fluorescence recovery after photobleaching. A step-tau of 10 dynes/cm2 elicited a rapid (5 s) increase of D in the portion of the cell upstream of the nucleus and a concomitant decrease in the downstream portion. A ramp-tau with a rate of 20 dynes/cm2 per min elicited a rapid (5 s) decrease of D in both the upstream and the downstream portions. The mitogen-activated protein kinases (MAPKs) ERK and JNK were activated by step-tau but not by ramping to the same tau level. Benzyl alcohol, which increases D, enhanced the activities of both MAPKs; cholesterol, which reduces D, diminished these activities. We conclude that the lipid bilayer can sense the temporal features of the applied tau with spatial discrimination and that the tau-induced membrane perturbations can be transduced into MAPK activation. These results have implications for understanding the role of t in modulating vascular functions in health and disease.


Subject(s)
Endothelium, Vascular/chemistry , JNK Mitogen-Activated Protein Kinases , Membrane Lipids/chemistry , Benzyl Alcohol/pharmacology , Cholesterol/pharmacology , Diffusion , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Activation/drug effects , Lipid Bilayers/chemistry , MAP Kinase Kinase 4 , Membrane Fluidity/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Sensitivity and Specificity , Stress, Mechanical
9.
J Orthop Res ; 31(9): 1360-5, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23606500

ABSTRACT

Human mesenchymal stem cells (hMSCs) can differentiate into various cell types, including osteogenic and chondrogenic cells. The matrix elasticity and cell seeding density are important factors in hMSCs differentiation. We cultured hMSCs at different seeding densities on polyacrylamide hydrogels with different stiffness corresponding to Young's moduli of 1.6 ± 0.3 and 40 ± 3.6 kPa. The promotion of osteogenic marker expression by hard gel is overridden by a high seeding density. Cell seeding density, however, did not influence the chondrogenic marker expressions induced by soft gel. These findings suggest that interplays between cell-matrix and cell-cell interactions contribute to hMSCs differentiation. The promotion of osteogenic differentiation on hard matrix was shown to be mediated through the Ras pathway. Inhibition of Ras (RasN17) significantly decreased ERK, Smad1/5/8 and AKT activation, and osteogenic markers expression. However, constitutively active Ras (RasV12) had little effect on osteogenic marker expression, suggesting that the Ras pathways are necessary but not sufficient for osteogenesis. Taken together, our results indicate that matrix elasticity and cell density are important microenvironmental cues driving hMSCs proliferation and differentiation.


Subject(s)
Elasticity/physiology , Extracellular Matrix/physiology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Biomarkers/metabolism , Cell Count , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Elastic Modulus/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Genes, ras , Humans , Mesenchymal Stem Cells/cytology , Proto-Oncogene Proteins c-akt/metabolism , Smad Proteins/metabolism
10.
Biomaterials ; 34(38): 9812-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24060419

ABSTRACT

ADSCs are a great cell source for tissue engineering and regenerative medicine. However, the development of methods to appropriately manipulate these cells in vitro remains a challenge. Here the proliferation and differentiation of ADSCs on microfabricated surfaces with varying geometries were investigated. To create the patterned substrates, a maskless biofabrication method was developed based on dynamic optical projection stereolithography. Proliferation and early differentiation of ADSCs were compared across three distinct multicellular patterns, namely stripes (ST), symmetric fork (SF), and asymmetric fork (AF). The ST pattern was designed for uniaxial cell alignment while the SF and AF pattern were designed with altered cell directionality to different extents. The SF and AF patterns generated similar levels of regional peak stress, which were both significantly higher than those within the ST pattern. No significant difference in ADSC proliferation was observed among the three patterns. In comparison to the ST pattern, higher peak stress levels of the SF and AF patterns were associated with up-regulation of the chondrogenic and osteogenic markers SOX9 and RUNX2. Interestingly, uniaxial cell alignment in the ST pattern seemed to increase the expression of SM22α and smooth muscle α-actin, suggesting an early smooth muscle lineage progression. These results indicate that geometric cues that promote uniaxial alignment might be more potent for myogenesis than those with increased peak stress. Overall, the use of these patterned geometric cues for modulating cell alignment and form-induced stress can serve as a powerful and versatile technique towards controlling differentiation in ADSCs.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/physiology , Stem Cells/cytology , Actins/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Tissue Scaffolds
11.
Proc Natl Acad Sci U S A ; 104(21): 8875-9, 2007 May 22.
Article in English | MEDLINE | ID: mdl-17496149

ABSTRACT

Vascular endothelial cells are continuously exposed to mechanical and chemical stimuli, such as shear stress and VEGF, respectively. It is still not clear how cells perceive these stimuli and orchestrate their responses. Studying the molecular mechanism by which shear stress and VEGF regulate the signaling pathways in bovine endothelial aortic cells, we found that VEGF induced a rapid association of VEGF receptor 2 (Flk-1) with Nck beta, but shear stress did not have such an effect. SU1498 (a specific inhibitor of Flk-1) and Nck beta(nm) (a negative mutant of Nck beta) blocked the VEGF-induced ERK and JNK activities. Only SU1498, but not Nck beta(nm), inhibited the shear-induced ERK activity. Furthermore, neither SU1498 nor Nck beta(nm) had significant effects on the shear-induced JNK activity, which can be blocked by inhibitors of Src family kinase and ROCK kinase. Therefore, mechanical (shear stress) and chemical (VEGF) stimuli diverge at the receptor Flk-1 in terms of the recruitment of the adapter protein Nck beta, and they employ different components of the complex signaling network in regulating downstream molecules, e.g., ERK and JNK.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cells, Cultured , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Protein Binding , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL