Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Lab Chip ; 16(9): 1625-35, 2016 04 26.
Article in English | MEDLINE | ID: mdl-27025227

ABSTRACT

We present a microfluidic immunoassay platform based on the use of linear microretroreflectors embedded in a transparent polymer layer as an optical sensing surface, and micron-sized magnetic particles as light-blocking labels. Retroreflectors return light directly to its source and are highly detectable using inexpensive optics. The analyte is immuno-magnetically pre-concentrated from a sample and then captured on an antibody-modified microfluidic substrate comprised of embedded microretroreflectors, thereby blocking reflected light. Fluidic force discrimination is used to increase specificity of the assay, following which a difference imaging algorithm that can see single 3 µm magnetic particles without optical calibration is used to detect and quantify signal intensity from each sub-array of retroreflectors. We demonstrate the utility of embedded microretroreflectors as a new sensing modality through a proof-of-concept immunoassay for a small, obligate intracellular bacterial pathogen, Rickettsia conorii, the causative agent of Mediterranean Spotted Fever. The combination of large sensing area, optimized surface chemistry and microfluidic protocols, automated image capture and analysis, and high sensitivity of the difference imaging results in a sensitive immunoassay with a limit of detection of roughly 4000 R. conorii per mL.


Subject(s)
Immunoassay/instrumentation , Lab-On-A-Chip Devices , Rickettsia conorii/isolation & purification , Animals , Antibodies, Immobilized/metabolism , Automation, Laboratory , Cells, Immobilized , Computer-Aided Design , Equipment Design , Image Processing, Computer-Assisted , Immunoassay/methods , Immunomagnetic Separation , Limit of Detection , Magnetic Phenomena , Microscopy , Microscopy, Electron, Scanning , Microspheres , Microtechnology/methods , Polymethyl Methacrylate/chemistry , Proof of Concept Study , Reproducibility of Results , Rickettsia conorii/growth & development , Rickettsia conorii/immunology , Surface Properties
2.
PLoS One ; 10(3): e0118182, 2015.
Article in English | MEDLINE | ID: mdl-25826708

ABSTRACT

Advances in molecular biology, microfluidics, and laboratory automation continue to expand the accessibility and applicability of these methods beyond the confines of conventional, centralized laboratory facilities and into point of use roles in clinical, military, forensic, and field-deployed applications. As a result, there is a growing need to adapt the unit operations of molecular biology (e.g., aliquoting, centrifuging, mixing, and thermal cycling) to compact, portable, low-power, and automation-ready formats. Here we present one such adaptation, the rotary zone thermal cycler (RZTC), a novel wheel-based device capable of cycling up to four different fixed-temperature blocks into contact with a stationary 4-microliter capillary-bound sample to realize 1-3 second transitions with steady state heater power of less than 10 W. We demonstrate the utility of the RZTC for DNA amplification as part of a highly integrated rotary zone PCR (rzPCR) system that uses low-volume valves and syringe-based fluid handling to automate sample loading and unloading, thermal cycling, and between-run cleaning functionalities in a compact, modular form factor. In addition to characterizing the performance of the RZTC and the efficacy of different online cleaning protocols, we present preliminary results for rapid single-plex PCR, multiplex short tandem repeat (STR) amplification, and second strand cDNA synthesis.


Subject(s)
Automation, Laboratory , Polymerase Chain Reaction/methods , Humans , Polymerase Chain Reaction/standards , Reproducibility of Results , Sensitivity and Specificity
3.
Lab Chip ; 15(1): 151-8, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25325619

ABSTRACT

Digital microfluidics (DMF) is a powerful technique for sample preparation and analysis for a broad range of biological and chemical applications. In many cases, it is desirable to carry out DMF on an open surface, such that the matrix surrounding the droplets is ambient air. However, the utility of the air-matrix DMF format has been severely limited by problems with droplet evaporation, especially when the droplet-based biochemical reactions require high temperatures for long periods of time. We present a simple solution for managing evaporation in air-matrix DMF: just-in-time replenishment of the reaction volume using droplets of solvent. We demonstrate that this solution enables DMF-mediated execution of several different biochemical reactions (RNA fragmentation, first-strand cDNA synthesis, and PCR) over a range of temperatures (4-95 °C) and incubation times (up to 1 h or more) without use of oil, humidifying chambers, or off-chip heating modules. Reaction volumes and temperatures were maintained roughly constant over the course of each experiment, such that the reaction kinetics and products generated by the air-matrix DMF device were comparable to those of conventional benchscale reactions. This simple yet effective solution for evaporation management is an important advance in developing air-matrix DMF for a wide variety of new, high-impact applications, particularly in the biomedical sciences.


Subject(s)
Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Solutions/analysis , Solvents/analysis , Air , DNA, Complementary , Equipment Design , Humans , Leukocytes, Mononuclear/chemistry , Models, Chemical , Particle Size , Polymerase Chain Reaction , RNA/analysis , RNA/isolation & purification , Solutions/chemistry , Solvents/chemistry
4.
Anal Chem ; 86(8): 3856-62, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24479881

ABSTRACT

Digital microfluidics (DMF) is a powerful technique for simple and precise manipulation of microscale droplets of fluid. This technique enables processing and analysis of a wide variety of samples and reagents and has proven useful in a broad range of chemical, biological, and medical applications. Handling of "real-world" samples has been a challenge, however, because typically their volumes are greater than those easily accommodated by DMF devices and contain analytes of interest at low concentration. To address this challenge, we have developed a novel "world-to-DMF" interface in which an integrated companion module drives the large-volume sample through a 10 µL droplet region on the DMF device, enabling magnet-mediated recovery of bead-bound analytes onto the device as they pass through the region. To demonstrate its utility, we use this system for extraction of RNA from human whole blood lysates (110-380 µL) and further purification in microscale volumes (5-15 µL) on the DMF device itself. Processing by the system was >2-fold faster and consumed 12-fold less reagents, yet produced RNA yields and quality fully comparable to conventional preparations and supporting qRT-PCR and RNA-Seq analyses. The world-to-DMF system is designed for flexibility in accommodating different sample types and volumes, as well as for facile integration of additional modules to enable execution of more complex protocols for sample processing and analysis. As the first technology of its kind, this innovation represents an important step forward for DMF, further enhancing its utility for a wide range of applications.


Subject(s)
Microfluidic Analytical Techniques/methods , Microfluidics/methods , RNA/blood , Equipment Design , Humans , Indicators and Reagents , RNA/isolation & purification , Reproducibility of Results
5.
PLoS One ; 8(7): e68988, 2013.
Article in English | MEDLINE | ID: mdl-23894387

ABSTRACT

Next-generation sequencing (NGS) is emerging as a powerful tool for elucidating genetic information for a wide range of applications. Unfortunately, the surging popularity of NGS has not yet been accompanied by an improvement in automated techniques for preparing formatted sequencing libraries. To address this challenge, we have developed a prototype microfluidic system for preparing sequencer-ready DNA libraries for analysis by Illumina sequencing. Our system combines droplet-based digital microfluidic (DMF) sample handling with peripheral modules to create a fully-integrated, sample-in library-out platform. In this report, we use our automated system to prepare NGS libraries from samples of human and bacterial genomic DNA. E. coli libraries prepared on-device from 5 ng of total DNA yielded excellent sequence coverage over the entire bacterial genome, with >99% alignment to the reference genome, even genome coverage, and good quality scores. Furthermore, we produced a de novo assembly on a previously unsequenced multi-drug resistant Klebsiella pneumoniae strain BAA-2146 (KpnNDM). The new method described here is fast, robust, scalable, and automated. Our device for library preparation will assist in the integration of NGS technology into a wide variety of laboratories, including small research laboratories and clinical laboratories.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/instrumentation , Microfluidic Analytical Techniques/instrumentation , Sequence Analysis, DNA/instrumentation , DNA, Bacterial/genetics , Genome, Bacterial/genetics , Genome, Human/genetics , Humans , Systems Integration
6.
Electrophoresis ; 33(23): 3506-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23135807

ABSTRACT

We have developed an automated quality control (QC) platform for next-generation sequencing (NGS) library characterization by integrating a droplet-based digital microfluidic (DMF) system with a capillary-based reagent delivery unit and a quantitative CE module. Using an in-plane capillary-DMF interface, a prepared sample droplet was actuated into position between the ground electrode and the inlet of the separation capillary to complete the circuit for an electrokinetic injection. Using a DNA ladder as an internal standard, the CE module with a compact LIF detector was capable of detecting dsDNA in the range of 5-100 pg/µL, suitable for the amount of DNA required by the Illumina Genome Analyzer sequencing platform. This DMF-CE platform consumes tenfold less sample volume than the current Agilent BioAnalyzer QC technique, preserving precious sample while providing necessary sensitivity and accuracy for optimal sequencing performance. The ability of this microfluidic system to validate NGS library preparation was demonstrated by examining the effects of limited-cycle PCR amplification on the size distribution and the yield of Illumina-compatible libraries, demonstrating that as few as ten cycles of PCR bias the size distribution of the library toward undesirable larger fragments.


Subject(s)
Electrophoresis, Microchip/instrumentation , Gene Library , Sequence Analysis, DNA/instrumentation , DNA/analysis , DNA/chemistry , Electrophoresis, Microchip/methods , Equipment Design , Humans , Leukocytes, Mononuclear/chemistry , Limit of Detection , Reproducibility of Results , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
7.
Appl Opt ; 51(14): 2532-40, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22614470

ABSTRACT

An integrated fiber-optic sensor is described that uses incoherent broadband cavity enhanced absorption spectroscopy for sensitive detection of aqueous samples in nanoliter volumes. Absorption was measured in a 100 µm gap between the ends of two short segments of multimode graded-index fiber that were integrated into a capillary using a precision machined V-grooved fixture that allowed for passive fiber alignment. The other ends of the fibers were coated with dielectric mirrors to form a 9.5 cm optical resonator. Light from a fiber-coupled superluminescent diode was directly coupled into one end of the cavity, and transmission was measured using a fiber-coupled silicon photodiode. Dilute aqueous solutions of near infrared dye were used to determine the minimum detectable absorption change of 2.4×10(-4) under experimental conditions in which pressure fluctuations limited performance. We also determined that the absolute minimum detectable absorption change would be 1.6×10(-5) for conditions of constant pressure in which absorption measurement is limited by electronic and optical noise. Tolerance requirements for alignment are also presented.


Subject(s)
Chemistry Techniques, Analytical/instrumentation , Fiber Optic Technology/instrumentation , Spectroscopy, Near-Infrared/instrumentation , Limit of Detection , Microchemistry/instrumentation , Optical Fibers , Pharmaceutical Preparations/analysis
8.
J Lab Autom ; 16(6): 405-14, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22093297

ABSTRACT

Next-generation sequencing (NGS) technology is a promising tool for identifying and characterizing unknown pathogens, but its usefulness in time-critical biodefense and public health applications is currently limited by the lack of fast, efficient, and reliable automated DNA sample preparation methods. To address this limitation, we are developing a digital microfluidic (DMF) platform to function as a fluid distribution hub, enabling the integration of multiple subsystem modules into an automated NGS library sample preparation system. A novel capillary interface enables highly repeatable transfer of liquid between the DMF device and the external fluidic modules, allowing both continuous-flow and droplet-based sample manipulations to be performed in one integrated system. Here, we highlight the utility of the DMF hub platform and capillary interface for automating two key operations in the NGS sample preparation workflow. Using an in-line contactless conductivity detector in conjunction with the capillary interface, we demonstrate closed-loop automated fraction collection of target analytes from a continuous-flow sample stream into droplets on the DMF device. Buffer exchange and sample cleanup, the most repeated steps in NGS library preparation, are also demonstrated on the DMF platform using a magnetic bead assay and achieving an average DNA recovery efficiency of 80%±4.8%.


Subject(s)
DNA/analysis , Infections/genetics , Automation, Laboratory , Conductometry , High-Throughput Nucleotide Sequencing/instrumentation , High-Throughput Nucleotide Sequencing/methods , Humans , Infections/diagnosis , Microfluidic Analytical Techniques , Reproducibility of Results
9.
Electrophoresis ; 31(16): 2804-12, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20737447

ABSTRACT

Rapid and specific characterization of bacterial endospores is dependent on the ability to rupture the cell wall to enable analysis of the intracellular components. In particular, bacterial spores from the bacillus genus are inherently robust and very difficult to lyze or solubilize. Standard protocols for spore inactivation include chemical treatment, sonication, pressure, and thermal lysis. Although these protocols are effective for the inactivation of these agents, they are less well suited for sample preparation for analysis using proteomic and genomic approaches. To overcome this difficulty, we have designed a simple capillary device to perform thermal lysis of bacterial spores. Using this device, we were able to super heat (195 degrees C) an ethylene glycol lysis buffer to perform rapid flow-through rupture and solubilization of bacterial endospores. We demonstrated that the lysates from this preparation method are compatible with CGE as well as DNA amplification analysis. We further demonstrated the flow-through lysing device could be directly coupled to a miniaturized electrophoresis instrument for integrated sample preparation and analysis. In this arrangement, we were enabled to perform sample lysis, fluorescent dye labeling, and protein electrophoresis analysis of bacterial spores in less than 10 min. The described sample preparation device is rapid, simple, inexpensive, and easily integratable with various microfluidic devices.


Subject(s)
Bacteriolysis/physiology , Spores, Bacterial/physiology , Bacillus/genetics , Bacillus/growth & development , Bacillus/isolation & purification , Bacillus/physiology , Bacillus anthracis/genetics , Bacillus anthracis/physiology , Bacillus cereus/genetics , Bacillus cereus/physiology , Bacillus subtilis/genetics , Bacillus subtilis/physiology , Capillary Action , Cell Division , DNA Primers , DNA, Bacterial/genetics , Fluorescent Dyes , Nucleic Acid Amplification Techniques/methods , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Solubility , Spores, Bacterial/genetics , Spores, Bacterial/isolation & purification , Thermodynamics
10.
Electrophoresis ; 31(15): 2632-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20665921

ABSTRACT

Field-deployable detection technologies in the nation's water supplies have become a high priority in recent years. The unattended water sensor is presented which employs microfluidic chip-based gel electrophoresis for monitoring proteinaceous analytes in a small integrated sensor platform. The instrument collects samples directly from a domestic water flow. The sample is then processed in an automated microfluidic module using in-house designed fittings, microfluidic pumps and valves prior to analysis via Sandia's microChemLab module, which couples chip-based electrophoresis separations with sensitive LIF detection. The system is controlled using LabVIEW software to analyze water samples about every 12 min. The sample preparation, detection and data analysis has all been fully automated. Pressure transducers and a positive control verify correct operation of the system, remotely. A two-color LIF detector with internal standards allows corrections to migration time to account for ambient temperature changes. The initial unattended water sensor prototype is configured to detect protein biotoxins such as ricin as a first step toward a total bioanalysis capability based on protein profiling. The system has undergone significant testing at two water utilities. The design and optimization of the sample preparation train is presented with results from both laboratory and field testing.


Subject(s)
Electrophoresis, Microchip/instrumentation , Proteins/isolation & purification , Toxins, Biological/isolation & purification , Water/analysis , Animals , Chemical Warfare Agents/isolation & purification , Electrophoresis, Microchip/methods , Equipment Design , Ricin/isolation & purification
11.
Anal Chem ; 81(9): 3261-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19323537

ABSTRACT

The ability to monitor cell signaling events is crucial to the understanding of immune defense against invading pathogens. Conventional analytical techniques such as flow cytometry, microscopy, and Western blot are powerful tools for signaling studies. Nevertheless, each approach is currently stand-alone and limited by multiple time-consuming and labor-intensive steps. In addition, these techniques do not provide correlated signaling information on total intracellular protein abundance and subcellular protein localization. We report on a novel phosphoFlow Chip (pFC) that relies on monolithic microfluidic technology to rapidly conduct signaling studies. The pFC platform integrates cell stimulation and preparation, microscopy, and subsequent flow cytometry. pFC allows host-pathogen phosphoprofiling in 30 min with an order of magnitude reduction in the consumption of reagents. For pFC validation, we monitor the mitogen-activated protein kinases ERK1/2 and p38 in response to Escherichia coli lipopolysaccharide (LPS) stimulation of murine macrophage cells (RAW 264.7). pFC permits ERK1/2 phosphorylation monitoring starting at 5 s after LPS stimulation, with phosphorylation observed at 5 min. In addition, ERK1/2 phosphorylation is correlated with subsequent recruitment into the nucleus, as observed from fluorescence microscopy performed on cells upstream of flow cytometric analysis. The fully integrated cell handling has the added advantage of reduced cell aggregation and cell loss, with no detectable cell activation. The pFC approach is a step toward unified, automated infrastructure for high-throughput systems biology.


Subject(s)
Macrophages/metabolism , Microfluidic Analytical Techniques/methods , Systems Integration , Animals , Automation , Cell Adhesion , Cell Line , Cell Membrane Permeability , Flow Cytometry , Lipopolysaccharides/immunology , Macrophage Activation , Macrophages/cytology , Macrophages/immunology , Mice , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Pressure , Signal Transduction , Systems Biology , Time Factors , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Lab Chip ; 9(4): 507-15, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19190785

ABSTRACT

To enable several on-chip cell handling operations in a fused-silica substrate, small shallow micropores are radially embedded in larger deeper microchannels using an adaptation of single-level isotropic wet etching. By varying the distance between features on the photolithographic mask (mask distance), we can precisely control the overlap between two etch fronts and create a zero-thickness semi-elliptical micropore (e.g. 20 microm wide, 6 microm deep). Geometrical models derived from a hemispherical etch front show that micropore width and depth can be expressed as a function of mask distance and etch depth. These models are experimentally validated at different etch depths (25.03 and 29.78 microm) and for different configurations (point-to-point and point-to-edge). Good reproducibility confirms the validity of this approach to fabricate micropores with a desired size. To illustrate the wide range of cell handling operations enabled by micropores, we present three on-chip functionalities: continuous-flow particle concentration, immobilization of single cells, and picoliter droplet generation. (1) Using pressure differentials, particles are concentrated by removing the carrier fluid successively through a series of 44 shunts terminated by 31 microm wide, 5 microm deep micropores. Theoretical values for the concentration factor determined by a flow circuit model in conjunction with finite volume modeling are experimentally validated. (2) Flowing macrophages are individually trapped in 20 microm wide, 6 microm deep micropores by hydrodynamic confinement. The translocation of transcription factor NF-kappaB into the nucleus upon lipopolysaccharide stimulation is imaged by fluorescence microscopy. (3) Picoliter-sized droplets are generated at a 20 microm wide, 7 microm deep micropore T-junction in an oil stream for the encapsulation of individual E. coli bacteria cells.


Subject(s)
Cytological Techniques , Microfluidic Analytical Techniques/methods , Animals , Cell Line , Equipment Design , Escherichia coli/cytology , Macrophages/cytology , Mice , Microfluidic Analytical Techniques/instrumentation , Porosity , Reproducibility of Results
13.
Lab Chip ; 8(12): 2046-53, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19023467

ABSTRACT

Towards designing a portable diagnostic device for detecting biological toxins in bodily fluids, we have developed microfluidic chip-based immunoassays that are rapid (< 20 minutes), require minimal sample volume (<10 microL) and have appreciable sensitivity and dynamic range (microM-pM). The microfluidic chip is being integrated with miniaturized electronics, optical elements, fluid-handling components, and data acquisition software to develop a portable, self-contained device. The device is intended for rapid, point-of-care (and, in future, point-of-incident) testing in case of an accidental or intentional exposure/intoxication to biotoxins. Detection of toxins and potential host-response markers is performed using microfluidic electrophoretic immunoassays integrated with sample preconcentration and mixing of analytes with fluorescently labeled antibodies. Preconcentration is enabled by photopolymerizing a thin, nanoporous membrane with a MW cut-off of approximately 10 kDa in the sample loading region of the chip. Polymeric gels with larger pores are located adjacent to the size exclusion membrane to perform electrophoretic separation of antibody-analyte complex and excess antibody. Measurement of the ratio of bound and unbound immune-complex using sensitive laser-induced fluorescence detection provides quantitation of analyte in the sample. We have demonstrated electrophoretic immunoassays for the biotoxins ricin, Shiga toxin I, and Staphylococcal enterotoxin B (SEB). With off-chip mixing and no sample preconcentration, the limits of detection (LOD) were 300 pM for SEB, 500 pM for Shiga toxin I, and 20 nM for ricin. With a 10 min on-chip preconcentration, the LOD for SEB is <10 pM. The portable device being developed is readily applicable to detection of proteinaceous biomarkers of many other diseases and is intended to represent the next-generation diagnostic devices capable of rapid and quantitative measurements of multiple analytes simultaneously.


Subject(s)
Microfluidics/methods , Reproducibility of Results , Toxins, Biological/chemistry , Dose-Response Relationship, Drug , Enterotoxins/analysis , Enterotoxins/chemistry , Immunoassay/instrumentation , Immunoassay/methods , Microfluidics/instrumentation , Ricin/analysis , Ricin/chemistry , Shiga Toxin 1/analysis , Shiga Toxin 1/chemistry , Time Factors , Toxins, Biological/analysis
14.
Anal Chem ; 80(22): 8416-23, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18847280

ABSTRACT

We are developing an automated system for the simultaneous, rapid detection of a group of select agents and toxins in the environment. To detect toxins, we modified and automated an antibody-based approach previously developed for manual medical diagnostics that uses fluorescent eTag reporter molecules and is suitable for highly multiplexed assays. Detection is based on two antibodies binding simultaneously to a single antigen, one of which is labeled with biotin while the other is conjugated to a fluorescent eTag through a cleavable linkage. Aqueous samples are incubated with the mixture of antibodies along with streptavidin-coated magnetic beads and a photoactive porphyrin complex. In the presence of antigen, a molecular complex is formed where the cleavable linkage is held in proximity to the photoactive group. Upon excitation at 680 nm, free radicals are generated, which diffuse and cleave the linkage, releasing the eTags. Released eTags are analyzed using capillary gel electrophoresis with laser-induced fluorescence detection. Limits of detection for ovalbumin and botulinum toxoid individually were 4 (or 80 pg) and 16 ng/mL (or 320 pg), respectively, using the manual assay. In addition, we demonstrated the use of pairs of antibodies from different sources in a single assay to decrease the rate of false positives. Automation of the assay was demonstrated in a flow-through format with higher LODs of 32 ng/mL (or 640 ng) each of a mixture of ovalbumin and botulinum toxoid. This versatile assay can be easily modified with the appropriate antibodies to detect a wide range of toxins and other proteins.


Subject(s)
Botulinum Toxins/analysis , Clostridium botulinum/chemistry , Immunoassay/instrumentation , Immunoassay/methods , Magnetics , Microspheres , Animals , Automation , Botulinum Toxins/immunology , Computers , Ovalbumin/analysis , Ovalbumin/immunology , Safety , Sensitivity and Specificity , Time Factors , Toxoids/analysis , Toxoids/immunology
15.
Anal Chem ; 80(23): 9005-12, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19551975

ABSTRACT

We present a rapid method for the identification of viruses using microfluidic chip gel electrophoresis (CGE) of high-copy number proteins to generate unique protein profiles. Viral proteins are solubilized by heating at 95 degrees C in borate buffer containing detergent (5 min), then labeled with fluorescamine dye (10 s), and analyzed using the microChemLab CGE system (5 min). Analyses of closely related T2 and T4 bacteriophage demonstrate sufficient assay sensitivity and peak resolution to distinguish the two phage. CGE analyses of four additional viruses--MS2 bacteriophage, Epstein-Barr, respiratory syncytial, and vaccinia viruses--demonstrate reproducible and visually distinct protein profiles. To evaluate the suitability of the method for unique identification of viruses, we employed a Bayesian classification approach. Using a subset of 126 replicate electropherograms of the six viruses and phage for training purposes, successful classification with non-training data was 66/69 or 95% with no false positives. The classification method is based on a single attribute (elution time), although other attributes such as peak width, peak amplitude, or peak shape could be incorporated and may improve performance further. The encouraging results suggest a rapid and simple way to identify viruses without requiring specialty reagents such as PCR probes and antibodies.


Subject(s)
Electrophoresis, Microchip/instrumentation , Electrophoresis, Microchip/methods , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Viral Proteins/analysis , Viruses/chemistry , Bacteriophages/chemistry , Calibration , Electrophoresis, Microchip/economics , Electrophoresis, Polyacrylamide Gel , Equipment Design , Microfluidic Analytical Techniques/economics , Sensitivity and Specificity , Time Factors
16.
Anal Chem ; 79(15): 5763-70, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17591754

ABSTRACT

For domestic and military security, an autonomous system capable of continuously monitoring for airborne biothreat agents is necessary. At present, no system meets the requirements for size, speed, sensitivity, and selectivity to warn against and lead to the prevention of infection in field settings. We present a fully automated system for the detection of aerosolized bacterial biothreat agents such as Bacillus subtilis (surrogate for Bacillus anthracis) based on protein profiling by chip gel electrophoresis coupled with a microfluidic sample preparation system. Protein profiling has previously been demonstrated to differentiate between bacterial organisms. With the goal of reducing response time, multiple microfluidic component modules, including aerosol collection via a commercially available collector, concentration, thermochemical lysis, size exclusion chromatography, fluorescent labeling, and chip gel electrophoresis were integrated together to create an autonomous collection/sample preparation/analysis system. The cycle time for sample preparation was approximately 5 min, while total cycle time, including chip gel electrophoresis, was approximately 10 min. Sensitivity of the coupled system for the detection of B. subtilis spores was 16 agent-containing particles per liter of air, based on samples that were prepared to simulate those collected by wetted cyclone aerosol collector of approximately 80% efficiency operating for 7 min.


Subject(s)
Aerosols/chemistry , Biosensing Techniques/methods , Microbiological Techniques/methods , Microfluidics/methods , Proteins/chemistry , Spores, Bacterial/isolation & purification , Bacillus anthracis/cytology , Bacillus anthracis/immunology , Bacillus anthracis/isolation & purification , Bacillus subtilis/cytology , Bacillus subtilis/immunology , Bacillus subtilis/isolation & purification , Biosensing Techniques/instrumentation , Chromatography, Gel , Electrophoresis , Fluorescent Dyes/chemistry , Microbiological Techniques/instrumentation , Microfluidics/instrumentation , Reproducibility of Results , Sensitivity and Specificity , Spores, Bacterial/cytology , Spores, Bacterial/immunology
17.
Electrophoresis ; 26(6): 1144-54, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15704246

ABSTRACT

We report the development of a hand-held instrument capable of performing two simultaneous microchip separations (gel and zone electrophoresis), and demonstrate this instrument for the detection of protein biotoxins. Two orthogonal analysis methods are chosen over a single method in order to improve the probability of positive identification of the biotoxin in an unknown mixture. Separations are performed on a single fused-silica wafer containing two separation channels. The chip is housed in a microfluidic manifold that utilizes o-ring sealed fittings to enable facile and reproducible fluidic connection to the chip. Sample is introduced by syringe injection into a septum-sealed port on the device exterior that connects to a sample loop etched onto the chip. Detection of low nanomolar concentrations of fluorescamine-labeled proteins is achieved using a miniaturized laser-induced fluorescence detection module employing two diode lasers, one per separation channel. Independently controlled miniature high-voltage power supplies enable fully programmable electrokinetic sample injection and analysis. As a demonstration of the portability of this instrument, we evaluated its performance in a laboratory field test at the Defence Science and Technology Laboratory with a series of biotoxin variants. The two separation methods cleanly distinguish between members of a biotoxin test set. Analysis of naturally occurring variants of ricin and two closely related staphylococcal enterotoxins indicates the two methods can be used to readily identify ricin in its different forms and can discriminate between two enterotoxin isoforms.


Subject(s)
Electrophoresis, Microchip/methods , Microfluidic Analytical Techniques/methods , Toxins, Biological/isolation & purification , Electrophoresis, Microchip/instrumentation , Enterotoxins/isolation & purification , Equipment Reuse , Miniaturization , Ricin/isolation & purification , Ricinus/chemistry , Sensitivity and Specificity , Staphylococcus aureus
18.
Lab Chip ; 5(2): 184-90, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15672133

ABSTRACT

This paper presents the first systematic engineering study of the impact of chemical formulation and surface functionalization on the performace of free-standing microfluidic polymer elements used for high-pressure fluid control in glass microsystems. System design, chemical wet-etch processes, and laser-induced polymerization techniques are described, and parametric studies illustrate the effects of polymer formulation, glass surface modification, and geometric constraints on system performance parameters. In particular, this study shows that highly crosslinked and fluorinated polymers can overcome deficiencies in previously-reported microvalve architectures, particularly limited solvent compatibility. Substrate surface modification is shown effective in reducing the friction of the polymer-glass interface and thereby facilitating valve actuation. A microchip one-way valve constructed using this architecture shows a 2 x 10(8) ratio of forward and backward flow rates at 7 MPa. This valve architecture is integrated on chip with minimal dead volumes (70 pl), and should be applicable to systems (including chromatography and chemical synthesis devices) requiring high pressures and solvents of varying polarity.

19.
Anal Chem ; 77(2): 435-41, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15649038

ABSTRACT

The design, fabrication, and demonstration of a hand-held microchip-based analytical instrument for detection and identification of proteins and other biomolecules are reported. The overall system, referred to as muChemLab, has a modular design that provides for reliability and flexibility and that facilitates rapid assembly, fluid and microchip replacement, troubleshooting, and sample analysis. Components include two independent separation modules that incorporate interchangeable fluid cartridges, a 2-cm-square fused-silica microfluidic chip, and a miniature laser-induced fluorescence detection module. A custom O-ring sealed manifold plate connects chip access ports to a fluids cartridge and a syringe injection port and provides sample introduction and world-to-chip interface. Other novel microfluidic connectors include capillary needle fittings for fluidic connection between septum-sealed fluid reservoirs and the manifold housing the chip, enabling rapid chip priming and fluids replacement. Programmable high-voltage power supplies provide bidirectional currents up to 100 microAlpha at 5000 V, enabling real-time current and voltage monitoring and facilitating troubleshooting and methods development. Laser-induced fluorescence detection allows picomolar (10(-11) M) detection sensitivity of fluorescent dyes and nanomolar sensitivity (10(-9) M) for fluorescamine-labeled proteins. Migration time reproducibility was significantly improved when separations were performed under constant current control (0.5-1%) as compared to constant voltage control (2-8%).


Subject(s)
Electrophoresis, Microchip/instrumentation , Proteins/isolation & purification , Equipment Design
SELECTION OF CITATIONS
SEARCH DETAIL
...