Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38741699

ABSTRACT

The stiffness of the extracellular matrix induces differential tension within integrin-based adhesions, triggering differential mechanoresponses. However, it has been unclear if the stiffness-dependent differential tension is induced solely by myosin activity. Here, we report that in the absence of myosin contractility, 3T3 fibroblasts still transmit stiffness-dependent differential levels of traction. This myosin-independent differential traction is regulated by polymerizing actin assisted by actin nucleators Arp2/3 and formin where formin has a stronger contribution than Arp2/3 to both traction and actin flow. Intriguingly, despite only slight changes in F-actin flow speed observed in cells with the combined inhibition of Arp2/3 and myosin compared to cells with sole myosin inhibition, they show a 4-times reduction in traction than cells with myosin-only inhibition. Our analyses indicate that traditional models based on rigid F-actin are inadequate for capturing such dramatic force reduction with similar actin flow. Instead, incorporating the F-actin network's viscoelastic properties is crucial. Our new model including the F-actin viscoelasticity reveals that Arp2/3 and formin enhance stiffness sensitivity by mechanically reinforcing the F-actin network, thereby facilitating more effective transmission of flow-induced forces. This model is validated by cell stiffness measurement with atomic force microscopy and experimental observation of model-predicted stiffness-dependent actin flow fluctuation.

2.
Am J Physiol Heart Circ Physiol ; 326(5): H1180-H1192, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38457352

ABSTRACT

Endothelial cells (ECs) within the vascular system encounter fluid shear stress (FSS). High, laminar FSS promotes vasodilation and anti-inflammatory responses, whereas low or disturbed FSS induces dysfunction and inflammation. However, the adaptation of endothelial cells (ECs) to dynamically changing FSS patterns remains underexplored. Here, by combining traction force microscopy with a custom flow chamber, we examined human umbilical vein endothelial cells adapting their traction during transitions from short-term low shear to long-term high shear stress. We discovered that the initial low FSS elevates the traction by only half of the amount in response to direct high FSS even after flow changes to high FSS. However, in the long term under high FSS, the flow started with low FSS triggers a substantial second rise in traction for over 10 h. In contrast, the flow started directly with high FSS results in a quick traction surge followed by a huge reduction below the baseline traction in <30 min. Importantly, we find that the orientation of traction vectors is steered by initial shear exposure. Using Granger causality analysis, we show that the traction that aligns in the flow direction under direct high FSS functionally causes cell alignment toward the flow direction. However, EC traction that orients perpendicular to the flow that starts with temporary low FSS functionally causes cell orientation perpendicular to the flow. Taken together, our findings elucidate the significant influence of initial short-term low FSS on lasting changes in endothelial traction that induces EC alignment.NEW & NOTEWORTHY In our study, we uncover that preconditioning with low shear stress yields enduring impacts on endothelial cell traction and orientation, persisting even after transitioning to high-shear conditions. Using Granger causality analysis, we demonstrate a functional link between the direction of cell traction and subsequent cellular alignment across varying shear environments.


Subject(s)
Human Umbilical Vein Endothelial Cells , Stress, Mechanical , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Cells, Cultured , Mechanotransduction, Cellular , Time Factors , Cell Adhesion
3.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37790318

ABSTRACT

Within the vascular system, endothelial cells (ECs) are exposed to fluid shear stress (FSS), a mechanical force exerted by blood flow that is critical for regulating cellular tension and maintaining vascular homeostasis. The way ECs react to FSS varies significantly; while high, laminar FSS supports vasodilation and suppresses inflammation, low or disturbed FSS can lead to endothelial dysfunction and increase the risk of cardiovascular diseases. Yet, the adaptation of ECs to dynamically varying FSS remains poorly understood. This study focuses on the dynamic responses of ECs to brief periods of low FSS, examining its impact on endothelial traction-a measure of cellular tension that plays a crucial role in how endothelial cells respond to mechanical stimuli. By integrating traction force microscopy (TFM) with a custom-built flow chamber, we analyzed how human umbilical vein endothelial cells (HUVECs) adjust their traction in response to shifts from low to high shear stress. We discovered that initial exposure to low FSS prompts a marked increase in traction force, which continues to rise over 10 hours before slowly decreasing. In contrast, immediate exposure to high FSS causes a quick spike in traction followed by a swift reduction, revealing distinct patterns of traction behavior under different shear conditions. Importantly, the direction of traction forces and the resulting cellular alignment under these conditions indicate that the initial shear experience dictates long-term endothelial behavior. Our findings shed light on the critical influence of short-lived low-shear stress experiences in shaping endothelial function, indicating that early exposure to low FSS results in enduring changes in endothelial contractility and alignment, with significant consequences for vascular health and the development of cardiovascular diseases.

4.
Mol Biol Cell ; 34(12): ar115, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37672339

ABSTRACT

Directional cell migration is driven by the conversion of oscillating edge motion into lasting periods of leading edge protrusion. Actin polymerization against the membrane and adhesions control edge motion, but the exact mechanisms that determine protrusion period remain elusive. We addressed this by developing a computational model in which polymerization of actin filaments against a deformable membrane and variable adhesion dynamics support edge motion. Consistent with previous reports, our model showed that actin polymerization and adhesion lifetime power protrusion velocity. However, increasing adhesion lifetime decreased the protrusion period. Measurements of adhesion lifetime and edge motion in migrating cells confirmed that adhesion lifetime is associated with and promotes protrusion velocity, but decreased duration. Our model showed that adhesions' control of protrusion persistence originates from the Brownian ratchet mechanism for actin filament polymerization. With longer adhesion lifetime or increased-adhesion density, the proportion of actin filaments tethered to the substrate increased, maintaining filaments against the cell membrane. The reduced filament-membrane distance generated pushing force for high edge velocity, but limited further polymerization needed for protrusion duration. We propose a mechanism for cell edge protrusion in which adhesion strength regulates actin filament polymerization to control the periods of leading edge protrusion.


Subject(s)
Actins , Models, Biological , Actins/metabolism , Cell Movement/physiology , Actin Cytoskeleton/metabolism , Pseudopodia/metabolism
5.
STAR Protoc ; 3(2): 101387, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35578733

ABSTRACT

Real-time cell analysis (RTCA) enables high-throughput, quantitative kinetic measurements of cytopathic effect (CPE) in virus-infected cells. Here, we detail a RTCA approach for assessing antibody neutralization. We describe how to evaluate the neutralizing potency of monoclonal antibodies (mAbs) and identify viral escape mutants to antibody neutralization for severe respiratory syndrome coronavirus 2 (SARS-CoV-2). For complete details on the use and execution of this protocol, please refer to Zost et al. (2020) and Suryadevara et al. (2021).


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Monoclonal , Antibodies, Viral , COVID-19/diagnosis , Humans , Spike Glycoprotein, Coronavirus
6.
Cancer Lett ; 536: 215641, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35339615

ABSTRACT

As a partner of tetraspanins, EWI2 suppresses glioblastoma, melanoma, and prostate cancer; but its role in lung cancer has not been investigated. Bioinformatics analysis reveals that EWI2 gene expression is up regulated in lung adenocarcinoma and higher expression of EWI2 mRNA may predict poorer overall survival. However, experimental analysis shows that EWI2 protein is actually downregulated constantly in the tissues of lung adenocarcinoma and lung squamous cell carcinoma. Forced expression of EWI2 in human lung adenocarcinoma cells reduces total cellular and cell surface levels of various integrins and growth factor receptors, which initiates the outside-in motogenic and mitogenic signaling. These reductions result in the decreases in 1) cell-matrix adhesion, cell movement, and cell transformation in vitro and 2) tumor growth, burden, and metastasis in vivo, and result from the increases in lysosomal trafficking and proteolytic degradation of theses membrane receptors. EWI2 elevates lysosome formation by promoting nuclear retention of TFEB, the master transcription factor driving lysosomogenesis. In conclusion, EWI2 as a lung cancer suppressor attenuates lung cancer cells in a comprehensive fashion by inhibiting both tumor growth and tumor metastasis; EWI2 as an endolysosome regulator promotes lysosome activity to enhance lysosomal degradation of growth factor receptors and integrins and then reduce their levels and functions; and EWI2 can become a promising therapeutic candidate given its accessibility at the cell surface, dual inhibition on growth factor receptors and integrins, and broad-spectrum anti-cancer activity. More importantly, our observations also provide a novel therapeutic strategy to bypass the resistance to EGFR inhibitors.


Subject(s)
Adenocarcinoma of Lung , Antigens, CD/metabolism , Lung Neoplasms , Membrane Proteins/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Humans , Integrins/genetics , Integrins/metabolism , Lung Neoplasms/metabolism , Lysosomes/metabolism , Male , Receptors, Growth Factor/metabolism
7.
Curr Protoc ; 1(9): e233, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34491632

ABSTRACT

Accurate measurement of cellular traction force is critical for understanding physical interaction between cells and the extracellular matrix. Traction force microscopy (TFM) has become the most widely used tool for this purpose. While TFM has made continual progress in terms of resolution and accuracy, there have been challenges regarding obtaining user-friendly software and choosing the right values for parameters and sub-processes associated with the software. Here we provide step-by-step instructions for a MATLAB-based TFM software application equipped with multiple methods for image deformation quantification and force reconstruction, along with clarification on the computational meaning of the parameters within the software. We outline how to choose the optimal sub-methods and values for parameters for each process, depending on the characteristics of images and purpose of the analyses. The software's runtime is 20, 4, and 0.05 min by Fast BEM L1 (Boundary Element Method L1-regularization), Fast BEM L2 (L2-regularization), and FTTC (Fourier Transform Traction Cytometry), respectively, in addition to 7 min of particle-tracking velocimetry-based deformation tracking, for a single image (1280 × 960 pixel) on a standard workstation. Finally, the colocalization accuracies, in reference to a paxillin-GFP image, are compared between the three force reconstruction methods. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Setting up the TFM package in MATLAB Basic Protocol 2: Running the TFM package Alternate Protocol 1: Stage drift correction: Efficient subpixel registration Alternate Protocol 2: Force field calculation: FastBEM.


Subject(s)
Algorithms , Traction , Computer Simulation , Microscopy, Atomic Force , Software
8.
Biosens Bioelectron ; 171: 112709, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33075724

ABSTRACT

Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was classified as a pandemic by the World Health Organization and has caused over 550,000 deaths worldwide as of July 2020. Accurate and scalable point-of-care devices would increase screening, diagnosis, and monitoring of COVID-19 patients. Here, we demonstrate rapid label-free electrochemical detection of SARS-CoV-2 antibodies using a commercially available impedance sensing platform. A 16-well plate containing sensing electrodes was pre-coated with receptor binding domain (RBD) of SARS-CoV-2 spike protein, and subsequently tested with samples of anti-SARS-CoV-2 monoclonal antibody CR3022 (0.1 µg/ml, 1.0 µg/ml, 10 µg/ml). Subsequent blinded testing was performed on six serum specimens taken from COVID-19 and non-COVID-19 patients (1:100 dilution factor). The platform was able to differentiate spikes in impedance measurements from a negative control (1% milk solution) for all CR3022 samples. Further, successful differentiation and detection of all positive clinical samples from negative control was achieved. Measured impedance values were consistent when compared to standard ELISA test results showing a strong correlation between them (R2=0.9). Detection occurs in less than five minutes and the well-based platform provides a simplified and familiar testing interface that can be readily adaptable for use in clinical settings.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/immunology , Biosensing Techniques/instrumentation , Clinical Laboratory Techniques , Coronavirus Infections/blood , Dielectric Spectroscopy/instrumentation , Pneumonia, Viral/blood , Antibodies, Viral/immunology , Biosensing Techniques/economics , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques/economics , Coronavirus Infections/diagnosis , Coronavirus Infections/economics , Coronavirus Infections/immunology , Dielectric Spectroscopy/economics , Electric Impedance , Equipment Design , Humans , Immobilized Proteins/immunology , Pandemics , Pneumonia, Viral/diagnosis , Pneumonia, Viral/immunology , SARS-CoV-2 , Sensitivity and Specificity , Spike Glycoprotein, Coronavirus/immunology , Time Factors
9.
iScience ; 23(3): 100959, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32179481

ABSTRACT

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

10.
Toxicol In Vitro ; 50: 47-53, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29366910

ABSTRACT

Co-culture of hepatocyte and fibroblasts has shown distinct advantages in enhancing certain liver specific functions and maintaining hepatic polarity. However, the utility of hepatocyte co-culture models for studies, such as drug-drug interaction studies, has not been completely elucidated. In this study the induction of Cyp1a2, Cyp2b1/2, and Cyp3a2, the three major cytochrome P450 (CYP) isoforms in the rat liver, was evaluated in randomly mixed co-cultures and micropatterned co-cultures. We found that in both co-culture configurations, the drug-induced Cyp1a2, Cyp2b1/2, Cyp3a2 mRNA and activity were suppressed relative to those in monocultured hepatocytes. Further, we observed a significant increase in TGFß1 production in the co-cultures. Addition of 100 pg/ml TGFß1 to hepatocyte monocultures resulted in the suppression of Cyp1a2, Cyp2b1/2, and Cyp3a2 induction. These findings implicate TGFß1 as one of the important factors impairing drug induced CYP induction in co-cultures and suggests that caution needs to be exercised in the use of hepatocyte-fibroblast co-cultures for CYP induction studies.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Enzyme Induction , Fibroblasts/metabolism , Hepatocytes/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Coculture Techniques , Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme System/genetics , Male , Mice , NIH 3T3 Cells , Rats, Wistar
11.
Adv Healthc Mater ; 6(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-27774763

ABSTRACT

In this essay the authors argue that chamber pressure dominates the biomechanics of the contraction cycle of the heart, while tissue stiffness dominates the relaxation cycle. This appears to be an under-recognized challenge in cardiac tissue engineering. Optimal approaches will involve constructing chambers or modulating the stiffness of the scaffold/substrate in synchrony with the beating cycle.


Subject(s)
Myocardium , Tissue Engineering/methods , Tissue Scaffolds , Animals , Humans
12.
Mol Pharm ; 13(6): 1947-57, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27157693

ABSTRACT

Pluripotent stem cell derived hepatocyte-like cells (hPSC-HLCs) are an attractive alternative to primary human hepatocytes (PHHs) used in applications ranging from therapeutics to drug safety testing studies. It would be critical to improve and maintain mature hepatocyte functions of the hPSC-HLCs, especially for long-term studies. If 3D culture systems were to be used for such purposes, it would be important that the system can support formation and maintenance of optimal-sized spheroids for long periods of time, and can also be directly deployed in liver drug testing assays. We report the use of 3-dimensional (3D) cellulosic scaffold system for the culture of hPSC-HLCs. The scaffold has a macroporous network which helps to control the formation and maintenance of the spheroids for weeks. Our results show that culturing hPSC-HLCs in 3D cellulosic scaffolds increases functionality, as demonstrated by improved urea production and hepatic marker expression. In addition, hPSC-HLCs in the scaffolds exhibit a more mature phenotype, as shown by enhanced cytochrome P450 activity and induction. This enables the system to show a higher sensitivity to hepatotoxicants and a higher degree of similarity to PHHs when compared to conventional 2D systems. These results suggest that 3D cellulosic scaffolds are ideal for the long-term cultures needed to mature hPSC-HLCs. The mature hPSC-HLCs with improved cellular function can be continually maintained in the scaffolds and directly used for hepatotoxicity assays, making this system highly attractive for drug testing applications.


Subject(s)
Cellulose/metabolism , Hepatocytes/physiology , Pluripotent Stem Cells/physiology , Animals , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Liver/physiology , Pluripotent Stem Cells/metabolism
13.
ACS Biomater Sci Eng ; 2(9): 1649-1657, 2016 Sep 12.
Article in English | MEDLINE | ID: mdl-33440598

ABSTRACT

Obtaining functional hepatocytes from human pluripotent stem cells (hPSCs) holds great potential for applications in drug safety testing, as well in the field of regenerative medicine. However, developing functionally mature hPSC-derived hepatocytes (hPSC-Heps) remains a challenge. We hypothesized that the cellular microenvironment plays a vital role in the maturation of immature hepatocytes. In this study, we examined the role of mechanical stiffness, a key component of the cellular microenvironment, in the maturation of hPSC-Heps. We cultured hPSC-Heps on collagen-coated polyacrylamide hydrogels with varying elastic moduli. On softer substrates the hPSC-Heps formed compact colonies while on stiffer substrates they formed a diffuse monolayer. We observed an inverse correlation between albumin production and substrate stiffness. The expression of key cytochrome enzymes, which are expressed at higher levels in the adult liver compared to the fetal liver, also correlated inversely with substrate stiffness, whereas fetal markers such as Cyp3A7 and AFP showed no correlation with stiffness. Culture of hPSC-Heps on soft substrates for 12 days led to 10-30 fold increases in the expression of drug-metabolizing enzymes. These results demonstrate that substrate stiffness similar to that of the liver enables aspects of the maturation of hPSC-Heps.

14.
Stem Cell Reports ; 5(6): 1226-1238, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26626178

ABSTRACT

The quantitative analysis of cardiomyocyte function is essential for stem cell-based approaches for the in vitro study of human cardiac physiology and pathophysiology. We present a method to comprehensively assess the function of single human pluripotent stem cell-derived cardiomyocyte (hPSC-CMs) through simultaneous quantitative analysis of contraction kinetics, force generation, and electrical activity. We demonstrate that statistical analysis of movies of contracting hPSC-CMs can be used to quantify changes in cellular morphology over time and compute contractile kinetics. Using a biomechanical model that incorporates substrate stiffness, we calculate cardiomyocyte force generation at single-cell resolution and validate this approach with conventional traction force microscopy. The addition of fluorescent calcium indicators or membrane potential dyes allows the simultaneous analysis of contractility and calcium handling or action potential morphology. Accordingly, our approach has the potential for broad application in the study of cardiac disease, drug discovery, and cardiotoxicity screening.


Subject(s)
Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Action Potentials , Biomechanical Phenomena , Calcium/analysis , Cell Differentiation , Cell Line , Humans , Kinetics , Myocardial Contraction , Single-Cell Analysis/methods
16.
Gene ; 552(2): 209-18, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25241644

ABSTRACT

Microsatellites or Simple Sequence Repeats (SSRs) are tandem iterations of one to six base pairs, non-randomly distributed throughout prokaryotic and eukaryotic genomes. Limited knowledge is available about distribution of microsatellites in single stranded DNA (ssDNA) viruses, particularly vertebrate infecting viruses. We studied microsatellite distribution in 118 ssDNA virus genomes belonging to three families of vertebrate infecting viruses namely Circoviridae, Parvoviridae, and Anelloviridae, and found that microsatellites constitute an important component of these virus genomes. Mononucleotide repeats were predominant followed by dinucleotide and trinucleotide repeats. A strong positive relationship existed between number of mononucleotide repeats and genome size among all the three virus families. A similar relationship existed for the occurrence of DTTPH (di-, tri-, tetra-, penta- and hexa-nucleotide) repeats in the families Anelloviridae and Parvoviridae only. Relative abundance and relative density of mononucleotide repeats showed a strong positive relationship with genome size in Circoviridae and Parvoviridae. However, in the case of DTTPH repeats, these features showed a strong relationship with genome size in Circoviridae only. On the other hand, relative microsatellite abundance and relative density of mononucleotide repeats were negatively correlated with GC content (%) in Parvoviridae genomes. On the basis of available annotations, our analysis revealed maximum occurrence of mononucleotide as well as DTTPH repeats in the coding regions of these virus genomes. Interestingly, after normalizing the length of the coding and non-coding regions of each virus genome, we found relative density of microsatellites much higher in the non-coding regions. We understand that the present study will help in the better characterization of the stability, genome organization and evolution of these virus classes and may provide useful leads to decipher the etiopathogenesis of these viruses.


Subject(s)
Anelloviridae/genetics , Circoviridae/genetics , Genome, Viral , Microsatellite Repeats , Parvoviridae/genetics , DNA, Single-Stranded/genetics , Genome Size
17.
Development ; 140(20): 4165-76, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24026118

ABSTRACT

In mammals, cardiac development proceeds from the formation of the linear heart tube, through complex looping and septation, all the while increasing in mass to provide the oxygen delivery demands of embryonic growth. The developing heart must orchestrate regional differences in cardiomyocyte proliferation to control cardiac morphogenesis. During ventricular wall formation, the compact myocardium proliferates more vigorously than the trabecular myocardium, but the mechanisms controlling such regional differences among cardiomyocyte populations are not understood. Control of definitive cardiomyocyte proliferation is of great importance for application to regenerative cell-based therapies. We have used murine and human pluripotent stem cell systems to demonstrate that, during in vitro cellular differentiation, early ventricular cardiac myocytes display a robust proliferative response to ß-catenin-mediated signaling and conversely accelerate differentiation in response to inhibition of this pathway. Using gain- and loss-of-function murine genetic models, we show that ß-catenin controls ventricular myocyte proliferation during development and the perinatal period. We further demonstrate that the differential activation of the Wnt/ß-catenin signaling pathway accounts for the observed differences in the proliferation rates of the compact versus the trabecular myocardium during normal cardiac development. Collectively, these results provide a mechanistic explanation for the differences in localized proliferation rates of cardiac myocytes and point to a practical method for the generation of the large numbers of stem cell-derived cardiac myocytes necessary for clinical applications.


Subject(s)
Heart Ventricles/cytology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Developmental , Heart Ventricles/embryology , Humans , Mice , Morphogenesis , Myocytes, Cardiac/metabolism
18.
PLoS One ; 7(12): e51796, 2012.
Article in English | MEDLINE | ID: mdl-23284769

ABSTRACT

A common assumption made when performing in vitro cellular assays is that the concentration of substances in the culture system is uniform. However, since the cells that internalize and secrete substances reside at the bottom of the well, it is conceivable that a concentration gradient could arise across the fluid layer. Importantly, the concentration of a substance in the vicinity of a cell, which is the concentration of interest, cannot be measured via existing methods. In this work a simple strategy for estimating the concentration of a chemical species at the surface of a cell is presented. Finally, this result is used to outline a method for determining the appropriate concentration ranges for testing in vitro autocrine loops and small molecules.


Subject(s)
Autocrine Communication , Cell Physiological Phenomena , Embryonic Stem Cells/metabolism , Erythrocytes/metabolism , Fibroblasts/metabolism , Pharmaceutical Preparations/metabolism , Receptors, Cell Surface/metabolism , Animals , Biological Transport , Cell Culture Techniques , Cell Membrane/metabolism , Computer Simulation , Cytokines/metabolism , Embryonic Stem Cells/cytology , Enzyme-Linked Immunosorbent Assay , Erythrocytes/cytology , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Humans , Mice , Models, Biological
19.
Stem Cell Res ; 6(2): 168-76, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21112823

ABSTRACT

An improved understanding of the role of extracellular factors in controlling the embryonic stem cell (ESC) phenotype will aid the development of cell-based therapies. While the role of extracellular factors in controlling the pluripotency and differentiation of embryonic stem cells (ESCs) has been the subject of much investigation, the identity and role of extrinsic factors in modulating ESC growth under conditions supporting self-renewal remain largely unknown. We demonstrate that mouse ESC (mESC) growth is density dependent and that one of the mechanisms underlying this phenomenon is the action of survival-enhancing autocrine factors. Proteomic analysis of proteins secreted by mouse ESCs demonstrates significant levels of cyclophilin A which increases the growth rate of mouse ESCs in a dose-dependent manner. Additionally, inhibition of the cyclophilin A receptor CD147 decreases the growth rate of mESCs. These findings identify cyclophilin A as a novel survival-enhancing autocrine factor in mouse ESC cultures.


Subject(s)
Autocrine Communication , Cyclophilin A/metabolism , Embryonic Stem Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Cell Culture Techniques , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Culture Media/chemistry , Embryonic Stem Cells/cytology , Mice
20.
Lab Chip ; 7(9): 1146-53, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17713613

ABSTRACT

We present a novel technique to accurately position single cells on a substrate using negative dielectrophoresis and cell-substrate adhesion. The cells are suspended in physiological media throughout the patterning process. We also verify the biocompatibility of this method by demonstrating that the patterned cells proliferate and show normal morphology. We calculate the temperatures and transmembrane potential that cells in the device experience and compare them to physiologically acceptable levels described in previous studies.


Subject(s)
Cell Separation/instrumentation , Electrophoresis/instrumentation , Flow Cytometry/instrumentation , Microelectrodes , Microfluidic Analytical Techniques/instrumentation , Stem Cells/cytology , Stem Cells/physiology , Cell Separation/methods , Cells, Cultured , Electrophoresis/methods , Equipment Design , Equipment Failure Analysis , Flow Cytometry/methods , HL-60 Cells , Humans , Microfluidic Analytical Techniques/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...