Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Physiol ; 594(12): 3245-70, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26864374

ABSTRACT

KEY POINTS: Xenopus laevis craniofacial development is a good system for the study of Andersen-Tawil Syndrome (ATS)-associated craniofacial anomalies (CFAs) because (1) Kcnj2 is expressed in the nascent face; (2) molecular-genetic and biophysical techniques are available for the study of ion-dependent signalling during craniofacial morphogenesis; (3) as in humans, expression of variant Kcnj2 forms in embryos causes a muscle phenotype; and (4) variant forms of Kcnj2 found in human patients, when injected into frog embryos, cause CFAs in the same cell lineages. Forced expression of WT or variant Kcnj2 changes the normal pattern of Vmem (resting potential) regionalization found in the ectoderm of neurulating embryos, and changes the normal pattern of expression of ten different genetic regulators of craniofacial development, including markers of cranial neural crest and of placodes. Expression of other potassium channels and two different light-activated channels, all of which have an effect on Vmem , causes CFAs like those induced by injection of Kcnj2 variants. In contrast, expression of Slc9A (NHE3), an electroneutral ion channel, and of GlyR, an inactive Cl(-) channel, do not cause CFAs, demonstrating that correct craniofacial development depends on a pattern of bioelectric states, not on ion- or channel-specific signalling. Using optogenetics to control both the location and the timing of ion flux in developing embryos, we show that affecting Vmem of the ectoderm and no other cell layers is sufficient to cause CFAs, but only during early neurula stages. Changes in Vmem induced late in neurulation do not affect craniofacial development. We interpret these data as strong evidence, consistent with our hypothesis, that ATS-associated CFAs are caused by the effect of variant Kcnj2 on the Vmem of ectodermal cells of the developing face. We predict that the critical time is early during neurulation, and the critical cells are the ectodermal cranial neural crest and placode lineages. This points to the potential utility of extant, ion flux-modifying drugs as treatments to prevent CFAs associated with channelopathies such as ATS. ABSTRACT: Variants in potassium channel KCNJ2 cause Andersen-Tawil Syndrome (ATS); the induced craniofacial anomalies (CFAs) are entirely unexplained. We show that KCNJ2 is expressed in Xenopus and mouse during the earliest stages of craniofacial development. Misexpression in Xenopus of KCNJ2 carrying ATS-associated mutations causes CFAs in the same structures affected in humans, changes the normal pattern of membrane voltage potential regionalization in the developing face and disrupts expression of important craniofacial patterning genes, revealing the endogenous control of craniofacial patterning by bioelectric cell states. By altering cells' resting potentials using other ion translocators, we show that a change in ectodermal voltage, not tied to a specific protein or ion, is sufficient to cause CFAs. By adapting optogenetics for use in non-neural cells in embryos, we show that developmentally patterned K(+) flux is required for correct regionalization of the resting potentials and for establishment of endogenous early gene expression domains in the anterior ectoderm, and that variants in KCNJ2 disrupt this regionalization, leading to the CFAs seen in ATS patients.


Subject(s)
Andersen Syndrome/genetics , Craniofacial Abnormalities/genetics , Potassium Channels, Inwardly Rectifying/genetics , Animals , Embryo, Mammalian , Larva , Mice , Mice, Inbred C57BL , Muscle, Skeletal/abnormalities , Optogenetics , RNA, Messenger/genetics , Xenopus laevis
2.
Cytometry A ; 87(3): 190-4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25483307

ABSTRACT

Small vertebrate model organisms have recently gained popularity as attractive experimental models that enhance our understanding of human tissue and organ development. Despite a large body of evidence using optical spectroscopy for the characterization of small model organism on chip-based devices, no attempts have been so far made to interface microfabricated technologies with environmental scanning electron microscopy (ESEM). Conventional scanning electron microscopy requires high vacuum environments and biological samples must be, therefore, submitted to many preparative procedures to dehydrate, fix, and subsequently stain the sample with gold-palladium deposition. This process is inherently low-throughput and can introduce many analytical artifacts. This work describes a proof-of-concept microfluidic chip-based system for immobilizing zebrafish larvae for ESEM imaging that is performed in a gaseous atmosphere, under low vacuum mode and without any need for sample staining protocols. The microfabricated technology provides a user-friendly and simple interface to perform ESEM imaging on zebrafish larvae. Presented lab-on-a-chip device was fabricated using a high-speed infrared laser micromachining in a biocompatible poly(methyl methacrylate) thermoplastic. It consisted of a reservoir with multiple semispherical microwells designed to hold the yolk of dechorionated zebrafish larvae. Immobilization of the larvae was achieved by a gentle suction generated during blotting of the medium. Trapping region allowed for multiple specimens to be conveniently positioned on the chip-based device within few minutes for ESEM imaging.


Subject(s)
Cells, Immobilized/ultrastructure , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Zebrafish , Animals , Larva , Microscopy, Electron, Scanning/methods , Time Factors
3.
Cytometry A ; 85(11): 921-32, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25287981

ABSTRACT

Biotests performed on small vertebrate model organisms provide significant investigative advantages as compared with bioassays that employ cell lines, isolated primary cells, or tissue samples. The main advantage offered by whole-organism approaches is that the effects under study occur in the context of intact physiological milieu, with all its intercellular and multisystem interactions. The gap between the high-throughput cell-based in vitro assays and low-throughput, disproportionally expensive and ethically controversial mammal in vivo tests can be closed by small model organisms such as zebrafish or Xenopus. The optical transparency of their tissues, the ease of genetic manipulation and straightforward husbandry, explain the growing popularity of these model organisms. Nevertheless, despite the potential for miniaturization, automation and subsequent increase in throughput of experimental setups, the manipulation, dispensing and analysis of living fish and frog embryos remain labor-intensive. Recently, a new generation of miniaturized chip-based devices have been developed for zebrafish and Xenopus embryo on-chip culture and experimentation. In this work, we review the critical developments in the field of Lab-on-a-Chip devices designed to alleviate the limits of traditional platforms for studies on zebrafish and clawed frog embryo and larvae. © 2014 International Society for Advancement of Cytometry.


Subject(s)
Microfluidic Analytical Techniques/methods , Zebrafish/embryology , Animals , Automation, Laboratory/methods , Biological Assay/methods , Embryo Culture Techniques , Xenopus/embryology
4.
Cytometry A ; 85(6): 537-47, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24664821

ABSTRACT

Transgenic zebrafish (Danio rerio) models of human diseases have recently emerged as innovative experimental systems in drug discovery and molecular pathology. None of the currently available technologies, however, allow for automated immobilization and treatment of large numbers of spatially encoded transgenic embryos during real-time developmental analysis. This work describes the proof-of-concept design and validation of an integrated 3D microfluidic chip-based system fabricated directly in the poly(methyl methacrylate) transparent thermoplastic using infrared laser micromachining. At its core, the device utilizes an array of 3D micromechanical traps to actively capture and immobilize single embryos using a low-pressure suction. It also features built-in piezoelectric microdiaphragm pumps, embryo-trapping suction manifold, drug delivery manifold, and optically transparent indium tin oxide heating element to provide optimal temperature during embryo development. Furthermore, we present design of the proof-of-concept off-chip electronic interface equipped with robotic servo actuator driven stage, innovative servomotor-actuated pinch valves, and embedded miniaturized fluorescent USB microscope. Our results showed that the innovative device has 100% embryo-trapping efficiency while supporting normal embryo development for up to 72 hr in a confined microfluidic environment. We also showed data that this microfluidic system can be readily applied to kinetic analysis of a panel of investigational antiangiogenic agents in transgenic zebrafish lines. The optical transparency and embryo immobilization allow for convenient visualization of developing vasculature patterns in response to drug treatment without the need for specimen re-positioning. The integrated electronic interfaces bring the lab-on-a-chip systems a step closer to realization of complete analytical automation.


Subject(s)
Ecotoxicology , Pharmaceutical Preparations/administration & dosage , Zebrafish , Animals , Animals, Genetically Modified , Drug Discovery , Ecotoxicology/instrumentation , Ecotoxicology/methods , Embryo, Nonmammalian/drug effects , Humans , Kinetics , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
5.
Curr Protoc Cytom ; 67: 9.44.1-9.44.16, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24510773

ABSTRACT

Zebrafish (Danio rerio) embryo assays have recently come into the spotlight as convenient experimental models in both biomedicine and ecotoxicology. As a small aquatic model organism, zebrafish embryo assays allow for rapid physiological, embryo-, and genotoxic tests of drugs and environmental toxins that can be simply dissolved in water. This protocol describes prototyping and application of an innovative, miniaturized, and polymeric chip-based device capable of immobilizing a large number of living fish embryos for real-time and/or time-lapse microscopic examination. The device provides a physical address designation to each embryo during analysis, continuous perfusion of medium, and post-analysis specimen recovery. Miniaturized embryo array is a new concept of immobilization and real-time drug perfusion of multiple individual and developing zebrafish embryos inside the mesofluidic device. The OpenSource device presented in this protocol is particularly suitable to perform accelerated fish embryo biotests in ecotoxicology and phenotype-based pharmaceutical screening.


Subject(s)
Biological Assay/instrumentation , Biological Assay/methods , Embryo, Nonmammalian/metabolism , Lab-On-A-Chip Devices , Zebrafish/embryology , Animals , Dimethylpolysiloxanes/chemistry , Embryo Culture Techniques , Hydrodynamics
6.
Curr Protoc Cytom ; Chapter 9: 9.41.1-9.41.8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23835805

ABSTRACT

Cell death within cell populations is a stochastic process where cell-to-cell variation in temporal progression through the various stages of cell death arises from asynchrony of subtle fluctuations in the signaling pathways. Most cell death assays rely on detection of the specific marker of cell demise at the end-point of cell culturing. Such an approach cannot account for the asynchrony and the stochastic nature of cell response to the death-inducing signal. There is a need therefore for rapid and high-throughput bioassays capable of continuously tracking viability of individual cells from the time of encountering a stress signal up to final stages of their demise. In this context, a new anthracycline derivative, DRAQ7, is gaining increasing interest as an easy-to-use marker capable of long-term monitoring of cell death in real-time. This novel probe neither penetrates the plasma membrane of living cells nor does it affect the cells' susceptibility to the death-inducing agents. However, when the membrane integrity is compromised, DRAQ7 enters cells undergoing demise and binds readily to nuclear DNA to report cell death. Here, we provide three sets of protocols for viability assays using DRAQ7 probe. The first protocol describes the innovative use of single-color DRAQ7 real-time assay to dynamically track cell viability. The second protocol outlines a simplified end-point DRAQ7 staining approach. The final protocol highlights the real-time and multiparametric apoptosis assay utilizing DRAQ7 dye concurrently with tetramethylrhodamine methyl ester (TMRM), the mitochondrial trans-membrane electrochemical potential (ΔΨm) sensing probe.


Subject(s)
Anthracyclines/metabolism , Biological Assay/methods , Cell Death , Cell Line , Cell Survival , Humans , Kinetics , Membrane Potential, Mitochondrial , Rhodamines , Staining and Labeling , Time Factors
7.
Biosens Bioelectron ; 48: 188-96, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23685315

ABSTRACT

Lab-on-a-Chip (LOC) biomicrofluidic technologies are rapidly emerging bioanalytical tools that can miniaturize and revolutionize in situ research on embryos of small vertebrate model organisms such as zebrafish (Danio rerio) and clawed African frog (Xenopus laevis). Despite considerable progress being made in fabrication techniques of chip-based devices, they usually still require excessive and manual actuation and data acquisition that significantly reduce throughput and introduce operator-related analytical bias. This work describes the development of a proof-of-concept embedded platform that integrates an innovative LOC zebrafish embryo array technology with an electronic interface to provide higher levels of laboratory automation for in situ biotests. The integrated platform was designed to perform automatic immobilization, culture and treatment of developing zebrafish embryos during fish embryo toxicity (FET) biotests. The system was equipped with a stepper motor driven stage, solenoid-actuated pinch valves, miniaturized peristaltic pumps as well as Peltier heating module. Furthermore, a Field Programmable Gate Array (FPGA) was used to implement an embedded hardware/software solution and interface to enable real-time control over embryo loading and immobilization; accurate microfluidic flow control; temperature stabilization and also automatic time-resolved image acquisition of developing zebrafish embryos. This work presents evidence that integration of embedded electronic interfaces with microfluidic chip-based technologies can bring the Lab-on-a-Chip a step closer to fully automated analytical systems.


Subject(s)
Lab-On-A-Chip Devices , Tissue Array Analysis/instrumentation , Toxicity Tests/instrumentation , Zebrafish/embryology , Animals , Equipment Design , Image Processing, Computer-Assisted , Tissue Culture Techniques/instrumentation
8.
Cytometry A ; 83(2): 227-34, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23165976

ABSTRACT

The exclusion of charged fluorescent dyes by intact cells has become a well-established assay for determining viability of cells. In search for a noninvasive fluorescent probe capable of long-term monitoring of cell death in real-time, we evaluated a new anthracycline derivative DRAQ7. The novel probe does not penetrate the plasma membrane of living cells but when the membrane integrity is compromised, it enters and binds readily to nuclear DNA to report cell death. It proved to be nontoxic to a panel of cancer cell lines grown continuously for up to 72 h and did not induce any detectable DNA damage signaling when analyzed using laser scanning microscopy and flow cytometry. The DRAQ7 provided a sensitive, real-time readout of cell death induced by a variety of stressors such as hypoxia, starvation, and drug-induced cytotoxicity. The overall responses to anticancer agents and resulting pharmacological dose-response profiles were not affected by the growth of tumor cells in the presence DRAQ7. Moreover, we for the first time introduced a near real-time microflow cytometric assay based on combination of DRAQ7 and mitochondrial inner membrane potential (ΔΨ(m) ) sensitive probe TMRM. We provide evidence that this low-dosage, real-time labeling procedure provides multiparameter and kinetic fingerprint of anticancer drug action.


Subject(s)
Anthracyclines/chemistry , Apoptosis , Fluorescent Dyes/chemistry , Anthracyclines/pharmacology , Benzamides/pharmacology , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Dactinomycin/pharmacology , Etoposide/pharmacology , Flow Cytometry , Fluorescent Dyes/pharmacology , Histones/metabolism , Humans , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects , Nitrophenols/pharmacology , Piperazines/pharmacology , Rhodamines/chemistry , Staurosporine/pharmacology , Sulfonamides/pharmacology , Sulfones/pharmacology
9.
Biosens Bioelectron ; 42: 586-91, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23261693

ABSTRACT

Multiparameter analysis of apoptosis in relation to cell cycle position is helpful in exploring mechanism of action of anticancer drugs that target specific molecular cogs of the cell cycle. This work demonstrates a new rationale for using microfluidic Lab-on-a-Chip flow cytometry (µFCM) with a simple 2D hydrodynamic focusing for the multiparameter analysis of apoptosis and DNA ploidy analysis in human hematopoietic cancer cells. The microfluidic system employs disposable microfluidic cartridges fabricated using injection moulding in optically transparent poly(methylmethacrylate). The dedicated and miniaturized electronic hardware interface enables up to six parameter detections using a combination of spatially separated solid-state 473 nm (10 mW) and 640 nm (20 mW) lasers and x-y stage for rapid laser alignment adjustment. We provide evidence that the simple 2D flow focusing on a chip-based device is sufficient to measure cellular DNA content in both fixed and living tumor cells. The feasibility of using the µFCM system for multiparameter analysis of caspase activation and dissipation of mitochondrial inner membrane potential (ΔΨ(m) loss) in relation to DNA content is also demonstrated. The data shows that straightforward microfluidic chip designs are sufficient to acquire high quality biological data when combined with sophisticated electronic interfaces. They can be a viable alternative to conventional FCM for multiparameter detection of programmed cell death.


Subject(s)
Apoptosis/genetics , Cell Cycle Proteins/isolation & purification , Flow Cytometry/methods , Lab-On-A-Chip Devices , DNA/chemistry , HL-60 Cells , Humans , Microfluidics , Mitochondrial Membranes/chemistry , Ploidies
10.
Curr Protoc Cytom ; 66: 9.42.1-9.42.15, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24510726

ABSTRACT

The age of microfluidic flow cytometry (µFCM) is fast becoming a reality. One of the most exciting applications of miniaturized chip-based cytometers is multivariate analysis using sampling volumes as small as 10 µl while matching the multiparameter data collection of conventional flow cytometers. We outline several innovative protocols for analyzing caspase-dependent cell death and cell cycle (DNA-content) profile using a fully integrated microfluidic flow cytometry system, Fishman-R. The first protocol describes the use of a new plasma membrane-permeability marker, DRAQ7, and the fluorogenic caspase substrate PhiPhiLux to track caspase activation during programmed cell death. Also outlined is the use of DRAQ7 fluorochrome in conjunction with the mitochondrial membrane potential-sensitive probe TMRM to track dissipation of inner mitochondrial cross-membrane potential. Another protocol adds the ability to measure dissipation of mitochondrial inner membrane potential (using TMRM probe) in relation to the cell cycle profile (using DRAQ5 probe) in living leukemic cells. Finally, we describe the combined use of fluorogenic caspases substrate PhiPhiLux with DRAQ5 probe to measure caspase activation in relation to the cell cycle profile in living tumor cells.


Subject(s)
Apoptosis , Flow Cytometry/methods , Lab-On-A-Chip Devices , Anthracyclines/metabolism , Anthraquinones/metabolism , Caspases/metabolism , Cell Cycle , Cell Line, Tumor , Enzyme Activation , Humans , Membrane Potential, Mitochondrial , Rhodamines/metabolism
11.
Proc SPIE Int Soc Opt Eng ; 86152013 Mar 09.
Article in English | MEDLINE | ID: mdl-24386542

ABSTRACT

Measurement of apoptotic markers in tumors can be directly correlated with the cell cycle phase using flow cytometry (FCM). The conventional DNA content analysis requires cell permeabilization to stain nuclei with fluorescent probes such as propidium iodide or use of a costly UV-excitation line for Hoechst 33342 probe. The access to FCM is also still limited to centralized core facilities due to its inherent high costs and complex operation. This work describes development and proof-of-concept validation of a portable and user-friendly microfluidic flow cytometer (µFCM) that can perform multivariate real time analysis on live cells using sampling volumes as small as 10 microliters. The µFCM system employs disposable microfluidic cartridges fabricated using injection molding in poly(methylmethacrylate) transparent thermoplastic. Furthermore, the dedicated and miniaturized electronic hardware interface enables up to six parameter detection using a combination of spatially separated solid-state 473 (10 mW) and 640 nm (20 mW) lasers and x-y stage for rapid laser alignment adjustment. We provide new evidence that a simple 2D flow focusing on a chip is sufficient to measure cellular DNA content in live tumor cells using a far-red DNA probe DRAQ5. The feasibility of using the µFCM system for a dose-response profiling of investigational anti-cancer agents on human hematopoietic cancer cells is also demonstrated. The data show that µFCM can provide a viable novel alternative to conventional FCM for multiparameter detection of caspase activation and dissipation of mitochondrial inner membrane potential (ΔΨm) in relation to DNA content (cell cycle phase) in live tumor cells.

12.
Biomicrofluidics ; 6(2): 24102-2410214, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22655014

ABSTRACT

The lack of technologies that combine automated manipulation, sorting, as well as immobilization of single metazoan embryos remains the key obstacle to high-throughput organism-based ecotoxicological analysis and drug screening routines. Noticeably, the major obstacle hampering the automated trapping and arraying of millimetre-sized embryos on chip-based devices is their substantial size and mass, which lead to rapid gravitational-induced sedimentation and strong inertial forces. In this work, we present a comprehensive mechanistic and design rationale for manipulation and passive trapping of individual zebrafish embryos using only hydrodynamic forces. We provide evidence that by employing innovative design features, highly efficient hydrodynamic positioning of large embryos on a chip can be achieved. We also show how computational fluid dynamics-guided design and the Lagrangian particle tracking modeling can be used to optimize the chip performance. Importantly, we show that rapid prototyping and medium scale fabrication of miniaturized devices can be greatly accelerated by combining high-speed laser prototyping with replica moulding in poly(dimethylsiloxane) instead of conventional photolithography techniques. Our work establishes a new paradigm for chip-based manipulation of large multicellular organisms with diameters well above 1 mm and masses often exceeding 1 mg. Passive docking of large embryos is an attractive alternative to provide high level of automation while alleviating potentially deleterious effects associated with the use of active chip actuation. This greatly expands the capabilities of bioanalyses performed on small model organisms and offers numerous and currently inaccessible laboratory automation advantages.

13.
PLoS One ; 7(5): e36630, 2012.
Article in English | MEDLINE | ID: mdl-22606275

ABSTRACT

Zebrafish (Danio rerio) has recently emerged as a powerful experimental model in drug discovery and environmental toxicology. Drug discovery screens performed on zebrafish embryos mirror with a high level of accuracy the tests usually performed on mammalian animal models, and fish embryo toxicity assay (FET) is one of the most promising alternative approaches to acute ecotoxicity testing with adult fish. Notwithstanding this, automated in-situ analysis of zebrafish embryos is still deeply in its infancy. This is mostly due to the inherent limitations of conventional techniques and the fact that metazoan organisms are not easily susceptible to laboratory automation. In this work, we describe the development of an innovative miniaturized chip-based device for the in-situ analysis of zebrafish embryos. We present evidence that automatic, hydrodynamic positioning, trapping and long-term immobilization of single embryos inside the microfluidic chips can be combined with time-lapse imaging to provide real-time developmental analysis. Our platform, fabricated using biocompatible polymer molding technology, enables rapid trapping of embryos in low shear stress zones, uniform drug microperfusion and high-resolution imaging without the need of manual embryo handling at various developmental stages. The device provides a highly controllable fluidic microenvironment and post-analysis eleuthero-embryo stage recovery. Throughout the incubation, the position of individual embryos is registered. Importantly, we also for first time show that microfluidic embryo array technology can be effectively used for the analysis of anti-angiogenic compounds using transgenic zebrafish line (fli1a:EGFP). The work provides a new rationale for rapid and automated manipulation and analysis of developing zebrafish embryos at a large scale.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Zebrafish/embryology , Animals , Animals, Genetically Modified , Computer Systems , Drug Delivery Systems , Embryo Culture Techniques , Equipment Design , Hydrodynamics , Microfluidic Analytical Techniques/methods , Miniaturization , Models, Animal , Neovascularization, Physiologic , Perfusion , Time-Lapse Imaging , Zebrafish/genetics
14.
Cytometry A ; 79(10): 799-813, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21548078

ABSTRACT

Small multicellular organisms such as nematodes, fruit flies, clawed frogs, and zebrafish are emerging models for an increasing number of biomedical and environmental studies. They offer substantial advantages over cell lines and isolated tissues, providing analysis under normal physiological milieu of the whole organism. Many bioassays performed on these alternative animal models mirror with a high level of accuracy those performed on inherently low-throughput, costly, and ethically controversial mammalian models of human disease. Analysis of small model organisms in a high-throughput and high-content manner is, however, still a challenging task not easily susceptible to laboratory automation. In this context, recent advances in photonics, electronics, as well as material sciences have facilitated the emergence of miniaturized bioanalytical systems collectively known as Lab-on-a-Chip (LOC). These technologies combine micro- and nanoscale sciences, allowing the application of laminar fluid flow at ultralow volumes in spatially confined chip-based circuitry. LOC technologies are particularly advantageous for the development of a wide array of automated functionalities. The present work outlines the development of innovative miniaturized chip-based devices for the in situ analysis of small model organisms. We also introduce a new term "wormometry" to collectively distinguish these up-and-coming chip-based technologies that go far beyond the conventional meaning of the term "cytometry."


Subject(s)
Caenorhabditis elegans/drug effects , Drosophila melanogaster/drug effects , Flow Cytometry/methods , High-Throughput Screening Assays , Image Cytometry/methods , Microfluidic Analytical Techniques/methods , Xenopus laevis/physiology , Zebrafish/physiology , Animals , Automation, Laboratory , Biological Products/pharmacology , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/physiology , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/physiology , Flow Cytometry/instrumentation , Humans , Image Cytometry/instrumentation , Microfluidic Analytical Techniques/instrumentation , Models, Animal , Nanotechnology/instrumentation , Nanotechnology/methods , Oligonucleotide Array Sequence Analysis , Xenopus laevis/anatomy & histology , Zebrafish/anatomy & histology
15.
Anal Chem ; 83(8): 3217-21, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21443166

ABSTRACT

Development of the dielectrophoretic (DEP) live cell trapping technology and its interfacing with the environmental scanning electron microscopy (ESEM) is described. DEP microelectrode arrays were fabricated on glass substrate using photolithography and lift-off. Chip-based arrays were applied for ESEM analysis of DEP-trapped human leukemic cells. This work provides proof-of-concept interfacing of the DEP cell retention and trapping technology with ESEM to provide a high-resolution analysis of individual nonadherent cells.


Subject(s)
Leukemia/pathology , Electrophoresis , Humans , Microelectrodes , Microscopy, Electron, Scanning/instrumentation , Surface Properties , Tumor Cells, Cultured , U937 Cells
16.
Anal Chem ; 83(6): 2133-44, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21344868

ABSTRACT

Quantification of programmed and accidental cell death provides useful end-points for the anticancer drug efficacy assessment. Cell death is, however, a stochastic process. Therefore, the opportunity to dynamically quantify individual cellular states is advantageous over the commonly employed static, end-point assays. In this work, we describe the development and application of a microfabricated, dielectrophoretic (DEP) cell immobilization platform for the real-time analysis of cancer drug-induced cytotoxicity. Microelectrode arrays were designed to generate weak electro-thermal vortices that support efficient drug mixing and rapid cell immobilization at the delta-shape regions of strong electric field formed between the opposite microelectrodes. We applied this technology to the dynamic analysis of hematopoietic tumor cells that represent a particular challenge for real-time imaging due to their dislodgement during image acquisition. The present study was designed to provide a comprehensive mechanistic rationale for accelerated cell-based assays on DEP chips using real-time labeling with cell permeability markers. In this context, we provide data on the complex behavior of viable vs dying cells in the DEP fields and probe the effects of DEP fields upon cell responses to anticancer drugs and overall bioassay performance. Results indicate that simple DEP cell immobilization technology can be readily applied for the dynamic analysis of investigational drugs in hematopoietic cancer cells. This ability is of particular importance in studying the outcome of patient derived cancer cells, when exposed to therapeutic drugs, as these cells are often rare and difficult to collect, purify and immobilize.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/instrumentation , Electrophoresis/instrumentation , Lab-On-A-Chip Devices , Cell Death/drug effects , Cell Line, Tumor , Cells, Immobilized/drug effects , Computer Simulation , Cycloheximide/pharmacology , Electric Impedance , Hematopoiesis/drug effects , Humans , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...