Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
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
2.
Biochem Biophys Res Commun ; 341(4): 1244-51, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16466697

ABSTRACT

Flow patterns in blood vessels contribute to focal distribution of atherosclerosis; the underlying mechanotransduction pathways remain to be investigated. We demonstrate that different flow patterns elicit distinct responses of Krüppel-like factor-2 (KLF2) in endothelial cells (ECs) in vitro and in vivo. While pulsatile flow with a significant forward direction induced sustained expression of KLF2 in cultured ECs, oscillatory flow with little forward direction caused prolonged suppression after a transient induction. The suppressive effect of oscillatory flow was Src-dependent. Immunohistochemical studies on ECs at arterial branch points revealed that KLF2 protein levels were related to local hemodynamics. Such flow-associated expression patterns were also demonstrated in a rat aortic restenosis model. Inhibition of KLF2 with siRNA sensitized ECs to oxidized LDL-induced apoptosis, indicating a protective role of KLF2. In conclusion, differential regulation of KLF2 may mediate the distinct vascular effects induced by various patterns of shear stress.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Kruppel-Like Transcription Factors/biosynthesis , Animals , Aorta, Abdominal/physiology , Aortic Valve Stenosis/physiopathology , Apoptosis/drug effects , Celiac Artery/physiology , Cells, Cultured , Gene Expression Regulation , Hemodynamics , Humans , Kruppel-Like Transcription Factors/deficiency , Male , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Umbilical Veins , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/physiology
3.
Mol Cell Biochem ; 281(1-2): 1-15, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16328952

ABSTRACT

Shear stress, a major hemodynamic force acting on the vessel wall, plays an important role in physiological processes such as cell growth, differentiation, remodelling, metabolism, morphology, and gene expression. We investigated the effect of shear stress on gene expression profiles in co-cultured vascular endothelial cells (ECs) and smooth muscle cells (SMCs). Human aortic ECs were cultured as a confluent monolayer on top of confluent human aortic SMCs, and the EC side of the co-culture was exposed to a laminar shear stress of 12 dyn/cm(2) for 4 or 24 h. After shearing, the ECs and SMCs were separated and RNA was extracted from the cells. The RNA samples were labelled and hybridized with cDNA array slides that contained 8694 genes. Statistical analysis showed that shear stress caused the differential expression (p < or = 0.05) of a total of 1151 genes in ECs and SMCs. In the co-cultured ECs, shear stress caused the up-regulation of 403 genes and down-regulation of 470. In the co-cultured SMCs, shear stress caused the up-regulation of 152 genes and down-regulation of 126 genes. These results provide new information on the gene expression profile and its potential functional consequences in co-cultured ECs and SMCs exposed to a physiological level of laminar shear stress. Although the effects of shear stress on gene expression in monocultured and co-cultured EC are generally similar, the response of some genes to shear stress is opposite between these two types of culture (e.g., ICAM-1 is up-regulated in monoculture and down-regulated in co-culture), which strongly indicates that EC-SMC interactions affect EC responses to shear stress.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Profiling , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , Stress, Mechanical , Cells, Cultured , Coculture Techniques , Endothelium, Vascular/cytology , Humans , Signal Transduction/genetics , Time Factors
4.
J Vasc Res ; 42(1): 77-89, 2005.
Article in English | MEDLINE | ID: mdl-15637443

ABSTRACT

Atherosclerosis occurs preferentially at vascular curvature and branch sites where the vessel walls are exposed to fluctuating shear stress and have high endothelial permeability. Endothelial permeability is modulated by intercellular adhesion molecules such as VE-cadherin. This study was designed to elucidate the effects of different flow patterns on the localization and expression of VE-cadherin in endothelial cells (ECs) both in vivo and in vitro. VE-cadherin staining at EC borders was much stronger in the descending thoracic aorta and abdominal aorta, where the pulsatile flow has a strong net forward component than in the aortic arch and the poststenotic dilatation site beyond an experimental constriction, where the flow near the wall is complex and reciprocating with little net flow. With the use of flow chambers the effects of pulsatile flow (12 +/- 4 dyn/cm2 at 1 Hz) and reciprocating flow (0.5 +/- 4 dyn/cm2 at 1 Hz) on VE-cadherin organization in endothelial monolayers were studied in vitro. VE-cadherin staining was continuous along cell borders in static controls. Following 6 h of either pulsatile or reciprocating flow, the VE-cadherin staining at cell borders became intermittent. When the pulsatile flow was extended to 24, 48 or 72 h the staining around the cell borders became continuous again, but the staining was still intermittent when the reciprocating flow was similarly extended. Exposure to pulsatile or reciprocating flow for 6 and 24 h neither change the expression level of VE-cadherin nor its distribution between membrane and cytosol fractions as determined by Western blot and compared with static controls. These findings suggest that the cell junction remodeling induced by different flow patterns may result from a redistribution of VE-cadherin within the cell membrane. Both the in vivo and in vitro data indicate that pulsatile and reciprocating flow patterns have different effects on cell junction remodeling. The lack of junction reorganization in regions of reciprocating flow in vivo and in vitro may provide a mechanistic basis for the high permeability and the preferential localization of atherosclerosis in regions of the arterial stress with complex flow patterns and fluctuating shear stress.


Subject(s)
Cadherins/analysis , Endothelial Cells/chemistry , Intercellular Junctions/chemistry , Animals , Antigens, CD , Aorta, Abdominal/chemistry , Cattle , Cells, Cultured , Cytoskeletal Proteins/analysis , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Regional Blood Flow , Shear Strength , Trans-Activators/analysis , beta Catenin
5.
J Vasc Res ; 40(4): 378-88, 2003.
Article in English | MEDLINE | ID: mdl-12891007

ABSTRACT

The interaction of vascular smooth muscle cells (SMCs) and extracellular matrix plays important roles in vascular remodeling. We investigated the signaling pathways involved in SMC-induced matrix contraction and SMC migration in three-dimensional (3D) collagen matrix. Matrix contraction is inhibited by the disruption of actin filaments but not microtubules. Therefore, we investigated the roles of signaling pathways related to actin filaments in matrix contraction. SMC-induced matrix contraction was markedly blocked (-80%) by inhibiting the Rho-p160ROCK pathway and myosin light chain kinase, and was decreased to a lesser extent (30-40%) by a negative mutant of Rac and inhibitors of phosphatidylinositol 3-kinase (PI 3-kinase) or p38 mitogen-activated protein kinase (MAPK), but it was not affected by the inhibition of Ras and Cdc42-Wiskott-Aldrich syndrome protein (WASP) pathways. Inhibition of extracellular-signal-regulated kinase (ERK) decreased SMC-induced matrix contraction by only 15%. The migration speed and persistence of SMCs in the 3D matrix were decreased by the inhibition of p160ROCK, PI 3-kinase, p38 MAPK or WASP to different extents, and p160ROCK inhibitor had the strongest inhibitory effect. Our results suggest that the SMC-induced matrix contraction and the migration of SMCs in 3D matrix share some signaling pathways leading to force generation at cell-matrix adhesions and that various signaling pathways have different relative importance in the regulations of these processes in SMCs.


Subject(s)
Cell Movement/physiology , Extracellular Matrix/metabolism , MAP Kinase Signaling System/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Actin Cytoskeleton/physiology , Animals , Aorta/cytology , Blood Proteins/pharmacology , Cattle , Cells, Cultured , Collagen/physiology , Extracellular Matrix/drug effects , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myosin-Light-Chain Kinase/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal , p38 Mitogen-Activated Protein Kinases , rac GTP-Binding Proteins/metabolism , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
6.
Biochem Biophys Res Commun ; 303(2): 548-55, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12659853

ABSTRACT

We aimed at elucidating the molecular basis of c-fos promoter activation in vascular endothelial cells (ECs) in response to shear stress, with emphases on Rho family GTPases (Rho, Cdc42, and Rac) and intracellular calcium. Dominant-negative and constitutively activated mutants of these GTPases were used to block the action of upstream signals and to activate the downstream pathways, respectively. The role of intracellular calcium was assessed with intracellular calcium chelators. Only Rho, but not Cdc42 or Rac, is involved in the shear stress induction of c-fos. This Rho-mediated shear-induction of c-fos is dependent on intracellular calcium, but not on the Rho effector p160ROCK or actin filaments. While the inhibition of p160ROCK and its ensuing disruption of actin filaments decreased the basal c-fos activity in static ECs (no flow), it did not affect the shear-inductive effect. The calcium chelator BAPTA-AM inhibits the shear-induction, as well as the static level, of c-fos activity.


Subject(s)
Endothelium, Vascular/physiology , Gene Expression Regulation , Genes, fos , Promoter Regions, Genetic , rho GTP-Binding Proteins/metabolism , Animals , Aorta , Cattle , Cells, Cultured , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stress, Mechanical , rho-Associated Kinases
7.
Am J Physiol Cell Physiol ; 283(5): C1540-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12372815

ABSTRACT

Blood flow can modulate vascular cell functions. We studied interactions between integrins and Flk-1 in transducing the mechanical shear stress due to flow. This application of a step shear stress caused Flk-1. Casitas B-lineage lymphoma (Cbl) activation (Flk-1. Cbl association, tyrosine phosphorylation of the Cbl-bound Flk-1, and tyrosine phosphorylation of Cbl) in bovine aortic endothelial cells (BAECs). The activation of integrins by plating BAECs on vitronectin or fibronectin also induced this Flk-1. Cbl activation. The shear-induced Flk-1. Cbl activation was blocked by inhibitory antibodies for alphavbeta3- or beta1-integrin, suggesting that it is mediated by integrins. Inhibition of Flk-1 by SU1498 also abolished this shear-induced Flk-1. Cbl activation. In contrast to the requirement of integrins for Flk-1. Cbl activation, the Flk-1 blocker SU1498 had no detectable effect on the shear-induced integrin activation, suggesting that integrins and Flk-1 play sequential roles in the signal transduction hierarchy induced by shear stress. Integrins are essential for the mechanical activation of Flk-1 by shear stress but not for the chemical activation of Flk-1 by VEGF.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Endothelium, Vascular/metabolism , Integrin beta1/metabolism , Receptors, Vitronectin/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Aorta/cytology , Cattle , Cells, Cultured , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Intercellular Signaling Peptides and Proteins/pharmacology , Ligands , Lymphokines/pharmacology , Lymphoma, B-Cell , Oncogene Protein v-cbl , Phosphorylation , Proteins/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction/drug effects , Stress, Mechanical , Tyrosine/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Physiol Genomics ; 12(1): 1-11, 2002 Dec 26.
Article in English | MEDLINE | ID: mdl-12399447

ABSTRACT

Statistical methods for identifying differentially expressed genes from microarray data are evolving. We developed a test for the statistical significance of differential expression as a function of time. When applied to microarray data obtained from endothelial cells exposed to shearing for different durations, the new multi-group test (G-test) identified three times as many genes as the one-way ANOVA at the same significance level. Using simulated data, we showed that this increase in sensitivity was achieved without sacrificing specificity. Several genes known to respond to shear stress by Northern blotting were identified by the G-test at P < or = 0.01 (but not by ANOVA), with similar temporal patterns. The validity and utility of the G-test were further supported by the examination of a few more example genes in relation to the present knowledge of their regulatory mechanisms. This new significance test may have broad application for the analysis of gene-expression studies and, in fact, to other biological studies in general.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Profiling/statistics & numerical data , Immediate-Early Proteins , Models, Statistical , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Humans , Kinetics , Reproducibility of Results , Sensitivity and Specificity , Stress, Mechanical , Transcription Factors/biosynthesis , Transcription Factors/genetics
9.
J Cell Sci ; 115(Pt 10): 2199-206, 2002 May 15.
Article in English | MEDLINE | ID: mdl-11973360

ABSTRACT

Integrins mediate cell adhesion and signal transduction at focal adhesions. Here we investigate the roles of integrin beta subunits in the regulation of actin cytoskeletal structure and the activities of Rho and Rac. The overexpression of beta3 integrin in Chinese hamster ovary cells enhances Rho activity and stress fiber formation, whereas the overexpression of beta1 integrin increases Rac activity and lamellipodia formation. The overexpression of a mutant beta1-3-1 integrin, in which the extracellular I-domain-like sequence of beta1 integrin has been replaced with the corresponding sequence of beta3 integrin, also enhances Rho activity and the formation of stress fibers. Our results demonstrate that beta1 and beta3 integrins differentially regulate the activities of Rho family GTPases and that the extracellular domains of integrin beta subunits play a critical role in transducing the extracellular ligand-binding information into specific intracellular signaling events.


Subject(s)
Integrin beta1/chemistry , Integrin beta1/metabolism , Integrin beta3/chemistry , Integrin beta3/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , CHO Cells , Cell Adhesion , Cell Size , Cricetinae , Fibrinogen/metabolism , Fibronectins/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Time Factors , Transfection , rac GTP-Binding Proteins/metabolism
10.
Biochem Biophys Res Commun ; 292(4): 892-9, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11944898

ABSTRACT

Fluid shear stress can activate PI-3 kinase and JNK in vascular endothelial cells. This study was designed to establish the role of Cbl as an upstream molecule in the shear stress activation of PI-3 kinase and JNK. Confluent monolayers of bovine aortic endothelial cells (BAECs) were subjected to a shear stress of 12 dyn/cm(2) over intervals ranging from 0.5 to 30 min. Shear stress increased Cbl phosphorylation to 2.9-fold of control and Cbl association with the regulatory PI-3 kinase subunit p85 to 5.4-fold. The PI-3 kinase activity measured in Cbl-immunoprecipitated complexes increased to 11.7-fold in response to shear, suggesting that the shear stress activation of PI-3 kinase involves its association with Cbl. Furthermore, the shear stress induction of JNK was attenuated by a negative mutant of Cbl. Finally, shear stress caused an activation of PI 3-kinase only in BAECs seeded onto fibronectin, vitronectin, or laminin, but not poly-l-lysine. Our results suggest that Cbl plays a critical role in the shear stress induction of PI 3-kinase and JNK activities, and that this shear-induced activation requires the interaction of endothelial integrins with extracellular matrix proteins.


Subject(s)
Endothelium, Vascular/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases , Animals , Cattle , Cell Membrane/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Enzyme Induction/physiology , Extracellular Matrix/metabolism , Integrins/metabolism , JNK Mitogen-Activated Protein Kinases , Mutation , Phosphorylation , Precipitin Tests , Protein Transport/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-cbl , Signal Transduction/physiology , Stress, Mechanical , Transfection , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...