Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Anal Chem ; 96(10): 4111-4119, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38417100

ABSTRACT

Multiplexed analysis in medical diagnostics is widely accepted as a more thorough and complete method compared to single-analyte detection. While analytical methods like polymerase chain reaction and enzyme-linked immunosorbent assay (ELISA) exist for multiplexed detection of biomarkers, they remain time-consuming and expensive. Lateral flow assays (LFAs) are an attractive option for point-of-care testing, and examples of multiplexed LFAs exist. However, these devices are limited by spatial resolution of test lines, large sample volume requirements, cross-reactivity, and poor sensitivity. Recent work has developed capillary-flow microfluidic ELISA platforms as a more sensitive alternative to LFAs; however, multiplexed detection on these types of devices has yet to be demonstrated. In the aftermath of the initial SARS-CoV-2 pandemic, the need for rapid, sensitive point-of-care devices has become ever clearer. Moving forward, devices that can distinguish between diseases with similar presenting symptoms would be the ideal home diagnostic. Here, the first example of a multiplexed capillary-flow immunoassay device for the simultaneous detection of multiple biomarkers is reported. From a single sample addition step, the reagents and washing steps required for two simultaneous ELISAs are delivered to spatially separated test strips. Visual results can be obtained in <15 min, and images captured with a smartphone can be analyzed for quantitative data. This device was used to distinguish between and quantify H1N1 hemagglutinin (HA) and SARS-CoV-2 nucleocapsid protein (N-protein). Using this device, analytical detection limits of 840 and 133 pg/mL were obtained for hemagglutinin and nucleocapsid protein, respectively. The presence of one target in the device did not increase the signal on the other test line, indicating no cross-reactivity between the assays. Additionally, simultaneous detection of both N-protein and HA was performed as well as simultaneous detection of N-protein and human C-reactive protein (CRP). Elevated levels of CRP in a patient infected with SARS-CoV-2 have been shown to correlate with more severe outcomes and a greater risk of death as well. To further expand on the simultaneous detection of two biomarkers, CRP and N-protein were detected simultaneously, and the presence of SARS-CoV-2 N-protein did not interfere with the detection of CRP when both targets were present in the sample.


Subject(s)
Hemagglutinins , Influenza A Virus, H1N1 Subtype , Humans , Immunoassay/methods , SARS-CoV-2 , C-Reactive Protein/analysis , Biomarkers/analysis , Nucleocapsid Proteins
2.
Anal Chim Acta ; 1277: 341634, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37604607

ABSTRACT

Over the last few years, the SARS-CoV-2 pandemic has made the need for rapid, affordable diagnostics more compelling than ever. While traditional laboratory diagnostics like PCR and well-plate ELISA are sensitive and specific, they can be costly and take hours to complete. Diagnostic tests that can be used at the point-of-care or at home, like lateral flow assays (LFAs) are a simple, rapid alternative, but many commercially available LFAs have been criticized for their lack of sensitivity compared to laboratory methods like well-plate ELISAs. The Capillary-Driven Immunoassay (CaDI) device described in this work uses microfluidic channels and capillary action to passively automate the steps of a traditional well-plate ELISA for visual read out. This work builds on prior capillary-flow devices by further simplifying operation and use of colorimetric detection. Upon adding sample, an enzyme-conjugated secondary antibody, wash steps, and substrate are sequentially delivered to test and control lines on a nitrocellulose strip generating a colorimetric response. The end user can visually detect SARS-CoV-2 antigen in 15-20 min by naked eye, or results can be quantified using a smartphone and software such as ImageJ. An analytical detection limit of 83 PFU/mL for SARS-CoV-2 was determined for virus in buffer, and 222 PFU/mL for virus spiked into nasal swabs using image analysis, similar to the LODs determined by traditional well-plate ELISA. Additionally, a visual detection limit of 100 PFU/mL was determined in contrived nasal swab samples by polling 20 untrained end-users. While the CaDI device was used for detecting clinically relevant levels of SARS-CoV-2 in this study, the CaDI device can be easily adapted to other immunoassay applications by changing the reagents and antibodies.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2 , Immunoassay , Enzyme-Linked Immunosorbent Assay , Antibodies , COVID-19 Testing
3.
Anal Methods ; 15(22): 2721-2728, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37099406

ABSTRACT

A capillary-driven microfluidic sequential flow device, designed for eventual at-home or doctor's office use, was developed to perform an enzyme-linked immunosorbent assay (ELISA) for serology assays. Serology assays that detect SARS-CoV-2 antibodies can be used to determine prior infection, immunity status, and/or individual vaccination status and are typically run using well-plate ELISAs in centralized laboratories, but in this format SARs-CoV-2 serology tests are too expensive and/or slow for most situations. Instead, a point-of-need device that can be used at home or in doctor's offices for COVID-19 serology testing would provide critical information for managing infections and determining immune status. Lateral flow assays are common and easy to use, but lack the sensitivity needed to reliably detect SARS-CoV-2 antibodies in clinical samples. This work describes a microfluidic sequential flow device that is as simple to use as a lateral flow assay, but as sensitive as a well-plate ELISA through sequential delivery of reagents to the detection area using only capillary flow. The device utilizes a network of microfluidic channels made of transparency film and double-sided adhesive combined with paper pumps to drive flow. The geometry of the channels and storage pads enables automated sequential washing and reagent addition steps with two simple end-user steps. An enzyme label and colorimetric substrate produce an amplified, visible signal for increased sensitivity, while the integrated washing steps decrease false positives and increase reproducibility. Naked-eye detection can be used for qualitative results or a smartphone camera for quantitative analysis. The device detected antibodies at 2.8 ng mL-1 from whole blood, while a well-plate ELISA using the same capture and detection antibodies could detect 1.2 ng mL-1. The performance of the capillary-driven immunoassay (CaDI) system developed here was confirmed by demonstrating SARS-CoV-2 antibody detection, and we believe that the device represents a fundamental step forward in equipment-free point-of-care technology.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/diagnosis , Microfluidics , Reproducibility of Results , Enzyme-Linked Immunosorbent Assay/methods , Antibodies, Viral
4.
ACS Meas Sci Au ; 2(6): 584-594, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36570470

ABSTRACT

The COVID-19 pandemic focused attention on a pressing need for fast, accurate, and low-cost diagnostic tests. This work presents an electrochemical capillary driven immunoassay (eCaDI) developed to detect SARS-CoV-2 nucleocapsid (N) protein. The low-cost flow device is made of polyethylene terephthalate (PET) and adhesive films. Upon addition of a sample, reagents and washes are sequentially delivered to an integrated screen-printed carbon electrode for detection, thus automating a full sandwich immunoassay with a single end-user step. The modified electrodes are sensitive and selective for SARS-CoV-2 N protein and stable for over 7 weeks. The eCaDI was tested with influenza A and Sindbis virus and proved to be selective. The eCaDI was also successfully applied to detect nine different SARS-CoV-2 variants, including Omicron.

5.
Anal Chem ; 94(21): 7545-7550, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35588209

ABSTRACT

Urinary tract infections (UTIs) are one of the most common infections across the world and can lead to serious complications such as sepsis if not treated in a timely manner. Uropathogenic Escherichia coli account for 75% of all UTIs. Early diagnosis is crucial to help control UTIs, but current culturing methods are expensive and time-consuming and lack sensitivity. The existing point-of-care methods fall short because they rely on indirect detection from elevated nitrates in urine rather than detecting the actual bacteria causing the infection. Magnetophoresis is a powerful method used to separate and/or isolate cells of interest from complex matrices for analysis. However, magnetophoresis typically requires complex and expensive instrumentation to control flow in microfluidic devices. Coupling magnetophoresis with microfluidic paper-based analytical devices (µPADs) enables pump-free flow control and simple and low-cost operation. Early magnetophoresis µPADs showed detection limits competitive with traditional methods but higher than targets for clinical use. Here, we demonstrate magnetophoresis using hybrid µPADs that rely on capillary action in hydrophilic polyethylene terephthalate channels combined with paper pumps. We were able to detect E. coli with a calculated limit of detection of 2.40 × 102 colony-forming units per mL.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Escherichia coli Infections/diagnosis , Escherichia coli Infections/microbiology , Humans , Lab-On-A-Chip Devices , Point-of-Care Systems , Urinary Tract Infections/diagnosis , Urinary Tract Infections/microbiology
6.
Anal Chem ; 94(11): 4712-4719, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35263100

ABSTRACT

Point-of-care (POC) methods currently available for detecting SARS-CoV-2 infections still lack accuracy. Here, we report the development of a highly sensitive electrochemical immunoassay capable of quantitatively detecting the presence of the SARS-CoV-2 virus in patient nasopharyngeal samples using stencil-printed carbon electrodes (SPCEs) functionalized with capture antibodies targeting the SARS-CoV-2 nucleocapsid protein (N protein). Samples are added to the electrode surface, followed by horseradish peroxidase (HRP)-conjugated detection antibodies also targeting the SARS-CoV-2 N protein. The concentration of the virus in samples is quantified using chronoamperometry in the presence of 3,3'5,5'-tetramethylbenzidine. Limits of detection equivalent to less than 50 plaque forming units/mL (PFU/mL) were determined with virus sample volumes of 20 µL. No cross-reactivity was detected with the influenza virus and other coronavirus N proteins. Patient nasopharyngeal samples were tested as part of a proof-of-concept clinical study where samples were also tested using the gold-standard real-time quantitative polymerase chain reaction (RT-qPCR) method. Preliminary results from a data set of 22 samples demonstrated a clinical specificity of 100% (n = 9 negative samples according to RT-qPCR) and a clinical sensitivity of 70% for samples with RT-PCR cycle threshold (Ct) values under 30 (n = 10) and 100% for samples with Ct values under 25 (n = 5), which complies with the World Health Organization (WHO) criteria for POC COVID-19 diagnostic tests. Our functionalized SPCEs were also validated against standard plaque assays, and very good agreement was found between both methods (R2 = 0.9993, n = 6), suggesting that our assay could be used to assess patient infectivity. The assay currently takes 70 min from sampling to results.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/diagnosis , Humans , Immunoassay/methods , Nucleocapsid Proteins , Sensitivity and Specificity
7.
ACS Sens ; 6(11): 4067-4075, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34694794

ABSTRACT

Rapid and inexpensive serological tests for SARS-CoV-2 antibodies are needed to conduct population-level seroprevalence surveillance studies and can improve diagnostic reliability when used in combination with viral tests. Here, we report a novel low-cost electrochemical capillary-flow device to quantify IgG antibodies targeting SARS-CoV-2 nucleocapsid proteins (anti-N antibody) down to 5 ng/mL in low-volume (10 µL) human whole blood samples in under 20 min. No sample preparation is needed as the device integrates a blood-filtration membrane for on-board plasma extraction. The device is made of stacked layers of a hydrophilic polyester and double-sided adhesive films, which create a passive microfluidic circuit that automates the steps of an enzyme-linked immunosorbent assay (ELISA). The sample and reagents are sequentially delivered to a nitrocellulose membrane that is modified with a recombinant SARS-CoV-2 nucleocapsid protein. When present in the sample, anti-N antibodies are captured on the nitrocellulose membrane and detected via chronoamperometry performed on a screen-printed carbon electrode. As a result of this quantitative electrochemical readout, no result interpretation is required, making the device ideal for point-of-care (POC) use by non-trained users. Moreover, we show that the device can be coupled to a near-field communication potentiostat operated from a smartphone, confirming its true POC potential. The novelty of this work resides in the integration of sensitive electrochemical detection with capillary-flow immunoassay, providing accuracy at the point of care. This novel electrochemical capillary-flow device has the potential to aid the diagnosis of infectious diseases at the point of care.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies, Viral , Humans , Immunoassay , Nucleocapsid Proteins , Point-of-Care Systems , Reproducibility of Results , Seroepidemiologic Studies
8.
Analyst ; 146(13): 4340-4347, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34106115

ABSTRACT

Sensitive, reliable and cost-effective detection of pathogens has wide ranging applications in clinical diagnostics and therapeutics, water and food safety, environmental monitoring, biosafety and epidemiology. Nucleic acid amplification tests (NAATs) such as PCR and isothermal amplification methods provide excellent analytical performance and significantly faster turnaround times than conventional culture-based methods. However, the inherent cost and complexity of NAATs limit their application in resource-limited settings and the developing world. To help address this urgent need, we have developed a sensitive method for nucleic acid analysis based on padlock probe rolling circle amplification (PLRCA), nuclease protection (NP) and lateral flow detection (LFA), referred to as PLAN-LFA, that can be used in resource-limited settings. The assay involves solution-phase hybridization of a padlock probe to target, sequence-specific ligation of the probe to form a circular template that undergoes isothermal rolling circle amplification in the presence of a polymerase and a labeled probe DNA. The RCA product is a long, linear concatenated single-stranded DNA that contains binding sites for the labeled probe. The sample is then exposed to a nuclease which selectively cleaves single-stranded DNA, the double-stranded labeled probe is protected from nuclease digestion and detected in a lateral flow immunoassay format to provide a visual, colorimetric readout of results. We have developed specific assays targeting beta-lactamase resistance gene for monitoring of antimicrobial resistance and Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2, the novel coronavirus discovered in 2019) using the PLAN-LFA platform. The assay provides a limit of detection of 1.1 pM target DNA (or 1.3 × 106 copies per reaction). We also demonstrate the versatility and robustness of the method by performing analysis on DNA and RNA targets, and perform analysis in complex sample matrices like saliva, plant tissue extract and bacterial culture without any sample pretreatment steps.


Subject(s)
COVID-19 , SARS-CoV-2 , DNA Probes , Humans , Nucleic Acid Amplification Techniques , Nucleic Acid Hybridization
9.
Analyst ; 146(6): 1932-1939, 2021 Mar 21.
Article in English | MEDLINE | ID: mdl-33492316

ABSTRACT

Capillary-driven microfluidic devices are of significant interest for on-site analysis because they do not require external pumps and can be made from inexpensive materials. Among capillary-driven devices, those made from paper and polyester film are among the most common and have been used in a wide array of applications. However, since capillary forces are the only driving force, flow is difficult to control, and passive flow control methods such as changing the geometry must be used to accomplish various analytical applications. This study presents several new flow control methods that can be utilized in a laminate capillary-driven microfluidic device to increase available functionality. First, we introduce push and burst valve systems that can stop and start flow. These valves can stop flow for >30 min and be opened by either pressing the channel or inflowing other fluids to the valve region. Next, we propose flow control methods for Y-shaped channels that enable more functions. In one example, we demonstrate the ability to accurately control concentration to create laminar, gradient, and fully mixed flows. In a second example, flow velocity in the main channel is controlled by adjusting the length of the inlet channel. In addition, the flow velocity is constant as the inlet length increases. Finally, the flow velocity in the Y-shaped device as a function of channel height and fluid properties such as viscosity and surface tension was examined. As in previous studies on capillary-driven channels, the flow rate was affected by each parameter. The fluidic control tools presented here will enable new designs and functions for low cost point of need assays across a variety of fields.

10.
Anal Methods ; 12(43): 5177-5185, 2020 11 21.
Article in English | MEDLINE | ID: mdl-33073789

ABSTRACT

Microfluidic magnetophoresis is a powerful technique that is used to separate and/or isolate cells of interest from complex matrices for analysis. However, mechanical pumps are required to drive flow, limiting portability and making translation to point-of-care (POC) settings difficult. Microfluidic paper-based analytical devices (µPADs) offer an alternative to traditional microfluidic devices that do not require external pumps to generate flow. However, µPADs are not typically used for particle analysis because most particles become trapped in the porous fiber network. Here we report the ability of newly developed fast-flow microfluidic paper-based analytical devices (ffPADs) to perform magnetophoresis. ffPADs use capillary action in a gap between stacked layers of paper and transparency sheets to drive flow at higher velocities than traditional µPADs. The multi-layer ffPADs allow particles and cells to move through the gap without being trapped in the paper layers. We first demonstrate that ffPADs enable magnetic particle separations in a µPAD with a neodymium permanent magnet and study key factors that affect performance. To demonstrate utility, E. coli was used as a model analyte and was isolated from human urine before detection with a fluorescently labeled antibody. A capture efficiency of 61.5% was then obtained of E. coli labeled magnetic beads in human urine. Future studies will look at the improvement of the capture efficiency and to make this assay completely off-chip without the need of a fluorescent label. The assay and device described here demonstrate the first example of magnetophoresis in a paper based, pump free microfluidic device.


Subject(s)
Microfluidic Analytical Techniques , Paper , Capillary Action , Escherichia coli , Humans , Lab-On-A-Chip Devices
11.
Anal Bioanal Chem ; 412(13): 3051-3061, 2020 May.
Article in English | MEDLINE | ID: mdl-32193587

ABSTRACT

Pathogen detection is crucial for human, animal, and environmental health; crop protection; and biosafety. Current culture-based methods have long turnaround times and lack sensitivity. Nucleic acid amplification tests offer high specificity and sensitivity. However, their cost and complexity remain a significant hurdle to their applications in resource-limited settings. Thus, point-of-need molecular diagnostic platforms that can be used by minimally trained personnel are needed. The nuclease protection assay (NPA) is a nucleic acid hybridization-based technique that does not rely on amplification, can be paired with other methods to improve specificity, and has the potential to be developed into a point-of-need device. In traditional NPAs, hybridization of an anti-sense probe to the target sequence is followed by single-strand nuclease digestion. The double-stranded target-probe hybrids are protected from nuclease digestion, precipitated, and visualized using autoradiography or other methods. We have developed a paper-based nuclease protection assay (PB-NPA) that can be implemented in field settings as the detection approach requires limited equipment and technical expertise. The PB-NPA uses a lateral flow format to capture the labeled target-probe hybrids onto a nitrocellulose membrane modified with an anti-label antibody. A colorimetric enzyme-substrate pair is used for signal visualization, producing a test line. The nuclease digestion of non-target and mismatched DNA provides high specificity while signal amplification with the reporter enzyme-substrate provides high sensitivity. We have also developed an on-chip sample pretreatment step utilizing chitosan-modified paper to eliminate possible interferents from the reaction and preconcentrate nucleic acids, thereby significantly reducing the need for auxiliary equipment. Graphical abstract.


Subject(s)
Lab-On-A-Chip Devices , Nucleic Acids/analysis , Paper , Point-of-Care Systems , DNA/chemistry , Limit of Detection , Oligonucleotide Array Sequence Analysis
12.
Plant Physiol ; 181(2): 547-564, 2019 10.
Article in English | MEDLINE | ID: mdl-31391208

ABSTRACT

Individual cells of cyanobacteria or algae are supplied with light in a highly irregular fashion when grown in industrial-scale photobioreactors (PBRs). These conditions coincide with significant reductions in growth rate compared to the static light environments commonly used in laboratory experiments. We grew a dense culture of the model cyanobacterium Synechocystis sp. PCC 6803 under a sinusoidal light regime in a bench-top PBR (the Phenometrics environmental PBR [ePBR]). We developed a computational fluid dynamics model of the ePBR, which predicted that individual cells experienced rapid fluctuations (∼6 s) between 2,000 and <1 µmol photons m-2 s-1, caused by vertical mixing and self-shading. The daily average light exposure of a single cell was 180 µmol photons m-2 s-1 Physiological measurements across the day showed no in situ occurrence of nonphotochemical quenching, and there was no significant photoinhibition. An ex situ experiment showed that up to 50% of electrons derived from PSII were diverted to alternative electron transport in a rapidly changing light environment modeled after the ePBR. Collectively, our results suggest that modification of nonphotochemical quenching may not increase cyanobacterial productivity in PBRs with rapidly changing light. Instead, tuning the rate of alternative electron transport and increasing the processing rates of electrons downstream of PSI are potential avenues to enhance productivity. The approach presented here could be used as a template to investigate the photophysiology of any aquatic photoautotroph in a natural or industrially relevant mixing regime.


Subject(s)
Photobioreactors , Synechocystis/radiation effects , Cell Division , Circadian Rhythm , Hydrodynamics , Light , Oxygen/metabolism , Photosynthesis , Pigmentation , Synechocystis/metabolism
13.
Anal Chem ; 91(14): 8966-8972, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31276368

ABSTRACT

Microfluidic paper-based analytical devices (µPADs) are simple but powerful analytical tools that are gaining significant recent attention due to their many advantages over more traditional monitoring tools. These include being inexpensive, portable, pump-free, and having the ability to store reagents. One major limitation of these devices is slow flow rates, which are controlled by capillary action in the hydrophilic pores of cellulosic paper. Recent investigations have advanced the flow rates in µPADs through the generation of a gap or channel between two closely spaced paper sheets. This multilayered format has opened up µPADs to new applications and detection schemes, where large gap sizes (>300 µm) provide at least 169× faster flow rates than single-layer µPADs, but do not conform to established mathematical models for fluid transport in porous materials, such as the classic Lucas-Washburn equation. In the present study, experimental investigations and analytical modeling are applied to elucidate the driving forces behind the rapid flow rates in these devices. We investigate a range of hypotheses for the systems fluid dynamics and establish a theoretical model to predict the flow rate in multilayered µPADs that takes into account viscous dissipation within the paper. Device orientation, sample addition method, and the gap height are found to be critical concerns when modeling the imbibition in multilayered devices.


Subject(s)
Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Paper , Microfluidic Analytical Techniques/instrumentation , Models, Chemical
14.
Biosens Bioelectron ; 131: 46-52, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30822687

ABSTRACT

Detection of viral infection is commonly performed using serological techniques like the enzyme-linked immunosorbent assay (ELISA) to detect antibody responses. Such assays may also be used to determine the infection phase based on isotype prevalence. However, ELISAs demonstrate limited sensitivity and are difficult to perform at the point of care. Here, we present a novel technique for label-free, rapid detection of ultra-low concentrations of virus specific antibodies. We have developed a simple, robust capacitive biosensor using microwires coated with Zika or Chikungunya virus envelope antigen. With little discernable nonspecific binding, the sensor can detect as few as 10 antibody molecules in a small volume (10 molecules/30 µL) within minutes. It can also be used to rapidly, specifically, and accurately determine the isotype of antigen-specific antibodies. Finally, we demonstrate that anti-Zika virus antibody can be sensitively and specifically detected in dilute mouse serum and can be isotyped using the sensor. Overall, our findings suggest that our microwire sensor platform has the potential to be used as a reliable, sensitive, and inexpensive diagnostic tool to detect immune responses at the point of care.


Subject(s)
Antibodies, Viral/isolation & purification , Biosensing Techniques , Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , Antibodies, Viral/blood , Antibody Formation/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Viral Envelope Proteins/blood , Viral Envelope Proteins/isolation & purification , Zika Virus/pathogenicity , Zika Virus Infection/virology
15.
Anal Chem ; 90(12): 7777-7783, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29790331

ABSTRACT

Viral pathogens are a serious health threat around the world, particularly in resource limited settings, where current sensing approaches are often insufficient and slow, compounding the spread and burden of these pathogens. Here, we describe a label-free, point-of-care approach toward detection of virus particles, based on a microfluidic paper-based analytical device with integrated microwire Au electrodes. The device is initially characterized through capturing of streptavidin modified nanoparticles by biotin-modified microwires. An order of magnitude improvement in detection limits is achieved through use of a microfluidic device over a classical static paper-based device, due to enhanced mass transport and capturing of particles on the modified electrodes. Electrochemical impedance spectroscopy detection of West Nile virus particles was carried out using antibody functionalized Au microwires, achieving a detection limit of 10.2 particles in 50 µL of cell culture media. No increase in signal is found on addition of an excess of a nonspecific target (Sindbis). This detection motif is significantly cheaper (∼$1 per test) and faster (∼30 min) than current methods, while achieving the desired selectivity and sensitivity. This sensing motif represents a general platform for trace detection of a wide range of biological pathogens.


Subject(s)
Electrochemical Techniques , Paper , Virion/chemistry , Virion/isolation & purification , West Nile virus/chemistry , West Nile virus/isolation & purification , Gold/chemistry , Molecular Structure
16.
Lab Chip ; 18(5): 793-802, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29431751

ABSTRACT

Microfluidic paper-based analytical devices (µPADs) are a versatile and inexpensive point-of-care (POC) technology, but their widespread adoption has been limited by slow flow rates and the inability to carry out complex in field analytical measurements. In the present work, we investigate multilayer µPADs as a means to generate enhanced flow rates within self-pumping paper devices. Through optical and electrochemical measurements, the fluid dynamics are investigated and compared to established flow theories within µPADs. We demonstrate a ∼145-fold increase in flow rate (velocity = 1.56 cm s-1, volumetric flow rate = 1.65 mL min-1, over 5.5 cm) through precise control of the channel height in a 2 layer paper device, as compared to archetypical 1 layer µPAD designs. These design considerations are then applied to a self-pumping sequential injection device format, known as a three-dimensional paper network (3DPN). These 3DPN devices are characterized through flow injection analysis of a ferrocene complex and anodic stripping detection of cadmium, exhibiting a 5× enhancement in signal compared to stationary measurements.


Subject(s)
Cadmium/analysis , Microfluidic Analytical Techniques/instrumentation , Paper , Point-of-Care Systems , Electrochemical Techniques/instrumentation , Optical Imaging/instrumentation
17.
Adv Sci (Weinh) ; 4(10): 1700153, 2017 10.
Article in English | MEDLINE | ID: mdl-29051857

ABSTRACT

The ability to study individual bacteria or subcellular organelles using inertial microfluidics is still nascent. This is due, in no small part, to the significant challenges associated with concentrating and separating specific sizes of micrometer and sub-micrometer bioparticles in a microfluidic format. In this study, using a rigid polymeric microfluidic network with optimized microchannel geometry dimensions, it is demonstrated that 2 µm, and even sub-micrometer, particles can be continuously and accurately focused to stable equilibrium positions. Suspensions have been processed at flow rates up to 1400 µL min-1 in an ultrashort 4 mm working channel length. A wide range of suspension concentrations-from 0.01 to 1 v/v%-have been systematically investigated, with yields greater than 97%, demonstrating the potential of this technology for large-scale implementation. Additionally, the ability of this chip to separate micrometer- and sub-micrometer-sized particles and to focus bioparticles (cyanobacteria) has been demonstrated. This study pushes the microfluidic inertial focusing particle range down to sub-micrometer length scales, enabling novel routes for investigation of individual microorganisms and subcellular organelles.

19.
Biosens Bioelectron ; 87: 646-653, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27619528

ABSTRACT

This paper presents a label-free affinity-based capacitive biosensor using interdigitated electrodes. Using an optimized process of DNA probe preparation to minimize the effect of contaminants in commercial thiolated DNA probe, the electrode surface was functionalized with the 24-nucleotide DNA probes based on the West Nile virus sequence (Kunjin strain). The biosensor has the ability to detect complementary DNA fragments with a detection limit down to 20 DNA target molecules (1.5aM range), making it suitable for a practical point-of-care (POC) platform for low target count clinical applications without the need for amplification. The reproducibility of the biosensor detection was improved with efficient covalent immobilization of purified single-stranded DNA probe oligomers on cleaned gold microelectrodes. In addition to the low detection limit, the biosensor showed a dynamic range of detection from 1µL-1 to 105µL-1 target molecules (20 to 2 million targets), making it suitable for sample analysis in a typical clinical application environment. The binding results presented in this paper were validated using fluorescent oligomers.


Subject(s)
DNA Probes/chemistry , DNA, Single-Stranded/chemistry , DNA/analysis , Electric Capacitance , Electrochemical Techniques/instrumentation , Immobilized Nucleic Acids/chemistry , Nucleic Acid Hybridization , Base Sequence , Biosensing Techniques/instrumentation , Electrodes , Equipment Design , Gold , Humans , Limit of Detection , Point-of-Care Systems , Reproducibility of Results , Sulfhydryl Compounds/chemistry , West Nile Fever/virology , West Nile virus/chemistry
20.
Lab Chip ; 13(2): 208-11, 2013 Jan 21.
Article in English | MEDLINE | ID: mdl-23172274

ABSTRACT

Chemical gradients drive a diverse set of biological processes ranging from nerve transduction to ovulation. At present, the most common method for quantifying chemical gradients is microscopy. Here, a new concept for probing spatial and temporal chemical gradients is reported that uses a multi-layer microfluidic device to measure analyte concentration as a function of lateral position in a microfluidic channel using electrochemistry in a format that is readily adaptable to multi-analyte sensing.


Subject(s)
Electrochemical Techniques/instrumentation , Microfluidic Analytical Techniques/instrumentation , Dopamine/analysis , Electrochemical Techniques/methods , Equipment Design , Fluorescein/chemistry , Microelectrodes , Microfluidic Analytical Techniques/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...