Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Commun Biol ; 7(1): 677, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830977

ABSTRACT

We present a quantitative sandwich immunoassay for CD63 Extracellular Vesicles (EVs) and a constituent surface cargo, EGFR and its activity state, that provides a sensitive, selective, fluorophore-free and rapid alternative to current EV-based diagnostic methods. Our sensing design utilizes a charge-gating strategy, with a hydrophilic anion exchange membrane functionalized with capture antibodies and a charged silica nanoparticle reporter functionalized with detection antibodies. With sensitivity and robustness enhancement by the ion-depletion action of the membrane, this hydrophilic design with charged reporters minimizes interference from dispersed proteins, thus enabling direct plasma analysis without the need for EV isolation or sensor blocking. With a LOD of 30 EVs/µL and a high relative sensitivity of 0.01% for targeted proteomic subfractions, our assay enables accurate quantification of the EV marker, CD63, with colocalized EGFR by an operator/sample insensitive universal normalized calibration. We analysed untreated clinical samples of Glioblastoma to demonstrate this new platform. Notably, we target both total and "active" EGFR on EVs; with a monoclonal antibody mAb806 that recognizes a normally hidden epitope on overexpressed or mutant variant III EGFR. Analysis of samples yielded an area-under-the-curve (AUC) value of 0.99 and a low p-value of 0.000033, surpassing the performance of existing assays and markers.


Subject(s)
ErbB Receptors , Extracellular Vesicles , Glioblastoma , Tetraspanin 30 , Humans , Glioblastoma/blood , Glioblastoma/diagnosis , Glioblastoma/metabolism , Tetraspanin 30/metabolism , ErbB Receptors/metabolism , Extracellular Vesicles/metabolism , Immunoassay/methods , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Brain Neoplasms/blood , Brain Neoplasms/metabolism , Brain Neoplasms/diagnosis
2.
J Biotechnol ; 383: 27-38, 2024 Mar 10.
Article in English | MEDLINE | ID: mdl-38336281

ABSTRACT

The widespread adoption of genetically modified (GM) crops has escalated concerns about their safety and ethical implications, underscoring the need for efficient GM crop detection methods. Conventional detection methods, such as polymerase chain reaction, can be costly, lab-bound, and time-consuming. To overcome these challenges, we have developed RapiSense, a cost-effective, portable, and sensitive biosensor platform. This sensor generates a measurable voltage shift (0.1-1 V) in the system's current-voltage characteristics, triggered by an increase in membrane's negative charge upon hybridization of DNA/RNA targets with a specific DNA probe. Probes designed to identify the herbicide resistance gene hygromycin phosphotransferase show a detection range from ∼1 nM to ∼10 µM and can discriminate between complementary, non-specific, and mismatched nucleotide targets. The incorporation of a small membrane sensor to detect fragmented RNA samples substantially improve the platform's sensitivity. In this study, RapiSense has been effectively used to detect specific DNA and fragmented RNA in transgenic variants of Arabidopsis, sweet potato, and rice, showcasing its potential for rapid, on-site GM crop screening.


Subject(s)
Crops, Agricultural , RNA , Plants, Genetically Modified/genetics , Crops, Agricultural/genetics , Polymerase Chain Reaction/methods , DNA
3.
bioRxiv ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37905113

ABSTRACT

We present a novel quantitative immunoassay for CD63 EVs (extracellular vesicles) and a constituent surface cargo, EGFR and its activity state, that provides a sensitive, selective, fluorophore-free and rapid alternative to current EV-based diagnostic methods. Our sensing design utilizes a charge-gating strategy, with a hydrophilic anion exchange membrane and a charged silica nanoparticle reporter. With sensitivity and robustness enhancement by the ion-depletion action of the membrane, this hydrophilic design with charged reporters minimizes interference from dispersed proteins and fluorophore degradation, thus enabling direct plasma analysis. With a limit of detection of 30 EVs/µL and a high relative sensitivity of 0.01% for targeted proteomic subfractions, our assay enables accurate quantification of the EV marker, CD63, with colocalized EGFR by an operator/sample insensitive universal normalized calibration. Glioblastoma necessitates improved non-invasive diagnostic approaches for early detection and monitoring. Notably, we target both total and "active" EGFR on EVs; with a monoclonal antibody mAb806 that recognizes a normally hidden epitope on overexpressed or mutant variant III EGFR. This approach offers direct glioblastoma detection from untreated human patient samples. Analysis of glioblastoma clinical samples yielded an area-under-the-curve (AUC) value of 0.99 and low p-value of 0.000033, significantly surpassing the performance of existing assays and markers.

4.
ACS Nano ; 17(10): 9388-9404, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37071723

ABSTRACT

Extracellular nanocarriers (extracellular vesicles (EVs), lipoproteins, and ribonucleoproteins) of protein and nucleic acids mediate intercellular communication and are clinically adaptable as distinct circulating biomarkers. However, the overlapping size and density of the nanocarriers have so far prevented their efficient physical fractionation, thus impeding independent downstream molecular assays. Here, we report a bias-free high-throughput and high-yield continuous isoelectric fractionation nanocarrier fractionation technique based on their distinct isoelectric points. This nanocarrier fractionation platform is enabled by a robust and tunable linear pH profile provided by water-splitting at a bipolar membrane and stabilized by flow without ampholytes. The linear pH profile that allows easy tuning is a result of rapid equilibration of the water dissociation reaction and stabilization by flow. The platform is automated with a machine learning procedure to allow recalibration for different physiological fluids and nanocarriers. The optimized technique has a resolution of 0.3 ΔpI, sufficient to separate all nanocarriers and even subclasses of nanocarriers. Its performance is then evaluated with several biofluids, including plasma, urine, and saliva samples. Comprehensive, high-purity (plasma: >93%, urine: >95% and saliva: >97%), high-yield (plasma: >78%, urine: >87% and saliva: >96%), and probe-free isolation of ribonucleoproteins in 0.75 mL samples of various biofluids in 30 min is demonstrated, significantly outperforming affinity-based and highly biased gold standards having low yield and day-long protocols. Binary fractionation of EVs and different lipoproteins is also achieved with similar performance.


Subject(s)
Body Fluids , Extracellular Vesicles , Saliva/metabolism , Ribonucleoproteins , Body Fluids/chemistry , Extracellular Vesicles/metabolism , Lipoproteins/analysis , Lipoproteins/metabolism
5.
Nat Commun ; 14(1): 557, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36732521

ABSTRACT

Cardiovascular disease-related deaths (one-third of global deaths) can be reduced with a simple screening test for better biomarkers than the current lipid and lipoprotein profiles. We propose using a highly atheroprotective subset of HDL with colocalized PON1 (PON1-HDL) for superior cardiovascular risk assessment. However, direct quantification of HDL proteomic subclasses are complicated by the peroxides/antioxidants associated with HDL interfering with redox reactions in enzymatic calorimetric and electrochemical immunoassays. Hence, we developed an enzyme-free Nanoparticle-Gated Electrokinetic Membrane Sensor (NGEMS) platform for quantification of PON1-HDL in plasma within 60 min, with a sub-picomolar limit of detection, 3-4 log dynamic range and without needing sample pretreatment or individual-sample calibration. Using NGEMS, we report our study on human plasma PON1-HDL as a cardiovascular risk marker with AUC~0.99 significantly outperforming others (AUC~0.6-0.8), including cholesterol/triglycerides tests. Validation for a larger cohort can establish PON1-HDL as a biomarker that can potentially reshape cardiovascular landscape.


Subject(s)
Cardiovascular Diseases , Humans , Cardiovascular Diseases/diagnosis , Proteomics , Risk Factors , Lipoproteins , Heart Disease Risk Factors , Aryldialkylphosphatase , Cholesterol, HDL
6.
Sci Rep ; 13(1): 184, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604528

ABSTRACT

Droplet microfluidics offers a platform from which new digital molecular assay, disease screening, wound healing and material synthesis technologies have been proposed. However, the current commercial droplet generation, assembly and imaging technologies are too expensive and rigid to permit rapid and broad-range tuning of droplet features/cargoes. This rapid prototyping bottleneck has limited further expansion of its application. Herein, an inexpensive home-made pipette droplet microfluidics kit is introduced. This kit includes elliptical pipette tips that can be fabricated with a simple DIY (Do-It-Yourself) tool, a unique tape-based or 3D printed shallow-center imaging chip that allows rapid monolayer droplet assembly/immobilization and imaging with a smart-phone camera or miniature microscope. The droplets are generated by manual or automatic pipetting without expensive and lab-bound microfluidic pumps. The droplet size and fluid viscosity/surface tension can be varied significantly because of our particular droplet generation, assembly and imaging designs. The versatility of this rapid prototyping kit is demonstrated with three representative applications that can benefit from a droplet microfluidic platform: (1) Droplets as microreactors for PCR reaction with reverse transcription to detect and quantify target RNAs. (2) Droplets as microcompartments for spirulina culturing and the optical color/turbidity changes in droplets with spirulina confirm successful photosynthetic culturing. (3) Droplets as templates/molds for controlled synthesis of gold-capped polyacrylamide/gold composite Janus microgels. The easily fabricated and user-friendly portable kit is hence ideally suited for design, training and educational labs.


Subject(s)
Microfluidic Analytical Techniques , Microgels , Microfluidics/methods , Microfluidic Analytical Techniques/methods , Cell Encapsulation , Polymerase Chain Reaction
7.
Lab Chip ; 23(2): 285-294, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36524732

ABSTRACT

Ribonucleoproteins (RNPs), particularly microRNA-induced silencing complex (miRISC), have been associated with cancer-related gene regulation. Specific RNA-protein associations in miRISC complexes or those found in let-7 lin28A complexes can downregulate tumor-suppressing genes and can be directly linked to cancer. The high protein-RNA electrostatic binding affinity is a particular challenge for the quantification of the associated microRNAs (miRNAs). We report here the first microfluidic point-of-care assay that allows direct quantification of RNP-associated RNAs, which has the potential to greatly advance RNP profiling for liquid biopsy. Key to the technology is an integrated cation-anion exchange membrane (CEM/AEM) platform for rapid and irreversible dissociation (k = 0.0025 s-1) of the RNP (Cas9-miR-21) complex and quantification of its associated miR-21 in 40 minutes. The CEM-induced depletion front is used to concentrate the RNP at the depletion front such that the high electric field (>100 V cm-1) within the concentration boundary layer induces irreversible dissociation of the low KD (∼0.5 nM) complex, with ∼100% dissociation even though the association rate (kon = 6.1 s-1) is 1000 times higher. The high field also electrophoretically drives the dissociated RNA out of the concentrated zone without reassociation. A detection limit of 1.1 nM is achieved for Cy3 labelled miR-21.


Subject(s)
MicroRNAs , Microfluidics , Neoplasms , Humans , Gene Expression Regulation , Microfluidics/instrumentation , MicroRNAs/chemistry , Ribonucleoproteins/chemistry
8.
Commun Biol ; 5(1): 1358, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36496485

ABSTRACT

Superparamagnetic nanobeads offer several advantages over microbeads for immunocapture of nanocarriers (extracellular vesicles, lipoproteins, and viruses) in a bioassay: high-yield capture, reduction in incubation time, and higher capture capacity. However, nanobeads are difficult to "pull-down" because their superparamagnetic feature requires high nanoscale magnetic field gradients. Here, an electrodeposited track-etched membrane is shown to produce a unique superparamagnetic nano-edge ring with multiple edges around nanopores. With a uniform external magnetic field, the induced monopole and dipole of this nano edge junction combine to produce a 10× higher nanobead trapping force. A dense nanobead suspension can be filtered through the magnetic nanoporous membrane (MNM) at high throughput with a 99% bead capture rate. The yield of specific nanocarriers in heterogeneous media by nanobeads/MNM exceeds 80%. Reproducibility, low loss, and concentration-independent capture rates are also demonstrated. This MNM material hence expands the application of nanobead immunocapture to physiological samples.


Subject(s)
Extracellular Vesicles , Reproducibility of Results , Magnetic Fields , Membranes
9.
Small ; 18(28): e2201330, 2022 07.
Article in English | MEDLINE | ID: mdl-35670145

ABSTRACT

Current biomarkers for myocardial infarction (MI) diagnosis are typically late markers released upon cell death, incapable of distinguishing between ischemic and reperfusion injury and can be symptoms of other pathologies. Circulating microRNAs (miRNAs) have recently been proposed as alternative biomarkers for MI diagnosis; however, detecting the changes in the human cardiac miRNA profile during MI is extremely difficult. Here, to study the changes in miRNA levels during acute MI, a heart-on-chip model with a cardiac channel, containing human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes in human heart decellularized matrix and collagen, and a vascular channel, containing hiPSC-derived endothelial cells, is developed. This model is exposed to anoxia followed by normoxia to mimic ischemia and reperfusion, respectively. Using a highly sensitive miRNA biosensor that the authors developed, the exact same increase in miR-1, miR-208b, and miR-499 levels in the MI-on-chip and the time-matched human blood plasma samples collected before and after ischemia and reperfusion, is shown. That the surface marker profile of exosomes in the engineered model changes in response to ischemic and reperfusion injury, which can be used as biomarkers to detect MI, is also shown. Hence, the MI-on-chip model developed here can be used in biomarker discovery.


Subject(s)
Exosomes , Induced Pluripotent Stem Cells , MicroRNAs , Myocardial Infarction , Reperfusion Injury , Biomarkers/metabolism , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Hypoxia/metabolism , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Reperfusion , Reperfusion Injury/diagnosis
10.
Methods Mol Biol ; 2394: 3-18, 2022.
Article in English | MEDLINE | ID: mdl-35094318

ABSTRACT

We report a highly sensitive and selective CNT-switch liquid biopsy platform that detects and quantifies protein biomarker expressions from circulating tumor cells in blood for early detection of metastatic breast cancer and its relapse. This platform first isolates and enriches more than 99% of tumor cells with an off-chip micro-size membrane filtration technique and then conducts on-chip detection of the membrane and internal protein biomarkers of the tumor cells with high sensitivity and selectivity. High sensitivity is achieved with complete association of the antibody-antigen-antibody (Ab-Ag-Ab) complex by precisely and rapidly assembling carbon nanotubes (CNTs) across two parallel electrodes via sequential DC electrophoresis and dielectrophoresis (DEP) deposition. Each bridged CNT acts as a switch that connects the electrodes and closes the circuit to generate an electrical signal. The high selectivity is achieved with a critical hydrodynamic shear rate that irreversibly removes non-target linkers of the aligned CNTs. At present, we are able to detect the protein biomarkers from 5 spiked breast cancer tumor cells of different types within 7.5 ml of human blood samples. This demonstrates the potential of this platform as an inexpensive and noninvasive alternative to MRI scans and tissue biopsies currently used to detect early metastatic breast cancer and its relapse.


Subject(s)
Nanotubes, Carbon , Neoplastic Cells, Circulating , Biomarkers , Electrophoresis , Humans , Neoplasm Recurrence, Local
11.
ACS Biomater Sci Eng ; 8(11): 4618-4621, 2022 11 14.
Article in English | MEDLINE | ID: mdl-34932307

ABSTRACT

Ion-depletion action of an ion-selective membrane produces a moat channel that electrically insulates a cell colony and elevates the cell medium potential uniformly to synchronously activate and deactivate the voltage-gated ion channels of all cells. The result is robust synchronization with strong intercellular electrical communication and the discovery of ion channel deactivation that is only possible when the cells are in communication. The study suggests that the collective response of a cell colony to external stimuli is distinct from that of a single cell. Cell proliferation must hence be guided with strong intercellular communication and proper exogenous stimuli.


Subject(s)
Cell Culture Techniques , Ion Channels , Ion Channels/metabolism
12.
Lab Chip ; 21(20): 3876-3887, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34546237

ABSTRACT

Micro RNAs (miRNAs) have shown great potential as rapid and discriminating biomarkers for acute myocardial infarction (AMI) diagnosis. We have developed a multiplexed ion-exchange membrane-based miRNA (MIX·miR) preconcentration/sensing amplification-free platform for quantifying in parallel a panel of miRNAs, including miR-1, miR-208b, and miR-499, from the same plasma samples from: 1) reference subjects with no evident coronary artery disease (NCAD); 2) subjects with stable coronary artery disease (CAD); and 3) subjects experiencing ST-elevation myocardial infarction (STEMI) prior to (STEMI-pre) and following (STEMI-PCI) percutaneous coronary intervention. The picomolar limit of detection from raw plasma and 3-decade dynamic range of MIX·miR permits detection of the miRNA panel in untreated samples from disease patients and its precise standard curve, provided by large 0.1 to 1 V signals and eliminates individual sensor calibration. The use of molecular concentration feature reduces the assay time to less than 30 minutes and increases the detection sensitivity by bringing all targets close to the sensors. miR-1 was low for NCAD patients but more than one order of magnitude above the normal value for all samples from three categories (CAD, STEMI-pre, and STEMI-PCI) of patients with CAD. In fact, miR-1 expression levels of stable CAD, STEMI-pre and STEMI-PCI are each more than 10-fold higher than the previous class, in that order, well above the 95% confidence level of MIX·miR. Its overexpression estimate is significantly higher than the PCR benchmark. This suggests that, in contrast to protein biomarkers of myocardial injury, miR-1 appears to differentiate ischemia from both reperfusion injury and non-AMI CAD patients. The battery-operated MIX·miR can be a portable and low-cost AMI diagnostic device, particularly useful in settings where cardiac catheterization is not readily available to determine the status of coronary reperfusion.


Subject(s)
Coronary Artery Disease , MicroRNAs , Myocardial Infarction , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction , Coronary Artery Disease/diagnosis , Coronary Artery Disease/genetics , Humans , MicroRNAs/genetics , Myocardial Infarction/diagnosis
13.
Lab Chip ; 21(18): 3614, 2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34519324

ABSTRACT

Correction for 'Constant-potential environment for activating and synchronizing cardiomyocyte colonies with on-chip ion-depleting perm-selective membranes' by Vivek Yadav et al., Lab Chip, 2020, 20, 4273-4284, DOI: 10.1039/D0LC00809E.

14.
Anal Chem ; 93(16): 6456-6462, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33861566

ABSTRACT

Rapid point-of-care (POC) quantification of low virus RNA load would significantly reduce the turn-around time for the PCR test and help contain a fast-spreading epidemic. Herein, we report a droplet digital PCR (ddPCR) platform that can achieve this sensitivity and rapidity without bulky lab-bound equipment. The key technology is a flattened pipette tip with an elliptical cross-section, which extends a high aspect-ratio microfluidic chip design to pipette scale, for rapid (<5 min) generation of several thousand monodispersed droplets ∼150 to 350 µm in size with a CV of ∼2.3%. A block copolymer surfactant (polyoxyalkylene F127) is used to stabilize these large droplets in oil during thermal cycling. At this droplet size and number, positive droplets can be counted by eye or imaged by a smartphone with appropriate illumination/filtering to accurately quantify up to 100 target copies. We demonstrate with 2019 nCoV-PCR assay LODs of 3.8 copies per 20 µL of sample and a dynamic range of 4-100 copies. The ddPCR platform is shown to be inhibitor resistant with spiked saliva samples, suggesting RNA extraction may not be necessary. It represents a rapid 1.5-h POC quantitative PCR test that requires just a pipette equipped with elliptical pipette tip, a commercial portable thermal cycler, a smartphone, and a portable trans-illuminator, without bulky and expensive micropumps and optical detectors that prevent POC application.


Subject(s)
COVID-19 , Point-of-Care Systems , Humans , Real-Time Polymerase Chain Reaction , SARS-CoV-2 , Viral Load
15.
Talanta ; 225: 122021, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33592751

ABSTRACT

Frequent on-line and automated monitoring of multiple protein biomarkers level secreted in the culture media during tissue growth is essential for the successful development of Tissue Engineering and Regenerative Medicine (TERM) products. Here, we present a low-cost, rapid, reliable, and integrable anion-exchange membrane-(AEM) based multiplexed sensing platform for this application. Unlike the gold-standard manual ELISA test, incubation/wash steps are optimized for each target and precisely metered in microfluidic chips to enhance selectivity. Unlike optical detection and unreliable visual detection for the ELISA test, which require standardization for every usage, the AEM ion current signal also offers robustness, endowed by the pH and ionic strength control capability of the ion-selective membrane, such that a universal standard curve can be used to calibrate all runs. The electrical signal is enhanced by highly charged silica nanoparticle reporters, which also act as hydrodynamic shear amplifiers to enhance selectivity during wash. This AEM-based sensing platform is tested with vascular protein biomarkers, Endothelin-1 (ET-1), Angiogenin (ANG) and Placental Growth Factor (PlGF). The limit of detection and three-decade dynamic range are comparable to ELISA assay but with a significantly reduced assay time of 1 h vs 7 h, due to the elimination of calibration and blocking steps. Optimized protocol for each target renders the detection highly reliable with more than 98% confidence. The multiplexed detection capability of the platform is also demonstrated by simultaneous detection of ET-1, ANG and PlGF in 40 µl of the vascular endothelial cell culture supernatants using three-membrane AEM sensor and the performance is validated against ELISA.


Subject(s)
Hydrodynamics , Silicon Dioxide , Biomarkers , Enzyme-Linked Immunosorbent Assay , Placenta Growth Factor
16.
Nat Commun ; 12(1): 140, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420061

ABSTRACT

Solid-state nanopores allow high-throughput single-molecule detection but identifying and even registering all translocating small molecules remain key challenges due to their high translocation speeds. We show here the same electric field that drives the molecules into the pore can be redirected to selectively pin and delay their transport. A thin high-permittivity dielectric coating on bullet-shaped polymer nanopores permits electric field leakage at the pore tip to produce a voltage-dependent surface field on the entry side that can reversibly edge-pin molecules. This mechanism renders molecular entry an activated process with sensitive exponential dependence on the bias voltage and molecular rigidity. This sensitivity allows us to selectively prolong the translocation time of short single-stranded DNA molecules by up to 5 orders of magnitude, to as long as minutes, allowing discrimination against their double-stranded duplexes with 97% confidence.


Subject(s)
DNA, Single-Stranded/metabolism , High-Throughput Screening Assays/methods , Nanopores , Single Molecule Imaging/methods , Aluminum Oxide/chemistry , High-Throughput Screening Assays/instrumentation , Polymers/chemistry , Single Molecule Imaging/instrumentation , Surface Properties
17.
J Phys Chem B ; 125(7): 1906-1915, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33410691

ABSTRACT

The presence of a small number (∼1000) of charged nanoparticles or macromolecules on the surface of an oppositely charged perm-selective membrane is shown to sensitively gate the ionic current through the membrane at a particular voltage, thus producing a voltage signal much larger than thermal noise. We show that, at sufficiently high voltages, surface vortices appear on the membrane surface and sustain an ion-depleted boundary layer that controls the diffusion length and ion current. An asymmetric vortex bifurcation occurs beyond a critical voltage to reduce the diffusion length and the differential resistance by half. Surface nanoparticles and molecules only affect this transition voltage in the membrane I-V curve. It is shown to shift by 2 ln10 (RT/F) ∼ 0.12 V for every decade increase in bulk target concentration, independent of sensor dimension and target/probe pair. Such universal features of the surface charge-sensitive nonlinear and nonequilibrium conductance allow us to develop very robust (a 2-3 decade dynamic range for highly heterogeneous samples with built-in control) yet sensitive (subpicomolar) and selective biosensors for highly charged molecules like nucleic acids and endotoxins-and for proteins with charged nanoparticle reporters.


Subject(s)
Biosensing Techniques , Nanoparticles , Nucleic Acids , Ion Transport
18.
ACS Appl Mater Interfaces ; 12(49): 54459-54472, 2020 Dec 09.
Article in English | MEDLINE | ID: mdl-33215917

ABSTRACT

One-pot synthesis of novel hydrogel-based anion exchange membranes (AEMs), with only a single-phase monomer mixture, was used to eliminate surface heterogeneity and generate reproducible electroconvective microvortices in the over-limiting region of the current-voltage characteristic (CVC) curves. Diallyldimethylammonium chloride (DDA) was used as the main component to provide the cation charge groups, and 2-hydroxyethyl methacrylate (HEMA) and ethylene glycol dimethyl acrylate (EGDMA) were used as the auxiliary structure monomers. The uniform membrane structure allowed reproducible and sensitive DNA detection and quantification, as probe-target surface complexes can gate the ion flux and produce large voltage shifts in the over-limiting region. Suppressed membrane curvature due to controlled swelling is a crucial part to avoid the reduction of depletion region for maintaining the influence of target gene hybridization. Fourier-transform infrared (FTIR) spectroscopy verified the synthesized membrane structure, with a residual vinyl group that allows easy carboxylation via additional photografting reaction. Consequently, a significantly higher DNA probe functionalization efficiency is obtained on the homogeneous AEMs, evidenced by the increasing nitrogen element content and bonding via X-ray photoelectron spectroscopy (XPS). The DDA content was optimized to provide a sufficient coulomb force between AEM and nucleic acid backbone to promote the specific binding efficiency but without high dimensional swelling which might change the surface geometry and restrict the voltage shifting for sensing in the over-limiting region, and the optimal DDA/HEMA ratio was found to be 4/10. The synthesized AEM sensor for recombinant 35S promoter sequence identification exhibited a reproducible calibration standard curve with dynamic range between 30 fM and 1 µM and high selectivity with only 0.01 V shift for 1 µM nontarget oligo.


Subject(s)
Anion Exchange Resins/chemistry , Biosensing Techniques/methods , DNA/analysis , Membranes, Artificial , DNA/metabolism , DNA Probes/chemistry , DNA Probes/metabolism , DNA, Plant/analysis , DNA, Plant/metabolism , Hydrogels/chemistry , Limit of Detection , Methacrylates/chemistry , Microfluidics , Nucleic Acid Hybridization , Plants, Genetically Modified/genetics , Reproducibility of Results , Glycine max/genetics , Surface Properties
19.
Lab Chip ; 20(22): 4273-4284, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33090162

ABSTRACT

In this study, an ion depleted zone created by an ion-selective membrane was used to impose a high and uniform constant extracellular potential over an entire ∼1000 cell rat cardiomyocyte (rCM) colony on-a-chip to trigger synchronized voltage-gated ion channel activities while preserving cell viability, thus extending single-cell voltage-clamp ion channel studies to an entire normalized colony. Image analysis indicated that rCM beating was strengthened and accelerated (by a factor of ∼2) within minutes of ion depletion and the duration of contraction and relaxation phases was significantly reduced. After the initial synchronization, the entire colony responds collectively to external potential changes such that beating over the entire colony can be activated or deactivated within 0.1 s. These newly observed collective dynamic responses, due to simultaneous ion channel activation/deactivation by a uniform constant-potential extracellular environment, suggest that perm-selective membrane modules on cell culture chips can facilitate studies of extracellular cardiac cell electrical communication and how ion-channel related pathologies affect cardiac cell synchronization. The future applications of this new technology can lead to better drug screening platforms for cardiotoxicity as well as platforms that can facilitate synchronized maturation of pluripotent stem cells into colonies with high electrical connectivity.


Subject(s)
Ion Channels , Myocytes, Cardiac , Animals , Drug Evaluation, Preclinical , Rats
20.
Electrophoresis ; 41(21-22): 1878-1892, 2020 11.
Article in English | MEDLINE | ID: mdl-32180242

ABSTRACT

Liquid biopsy, screening cancer non-invasively and frequently by detecting and quantifying molecular markers in physiological fluids, would significantly improve cancer survival rate but it remains a distant goal. The key obstacles presented by the highly heterogeneous samples are rapid/high-yield purification and precise/selective marker capture by their antibody and oligo probes. As irregular expressions of these molecular biomarkers are the key signals, quantifying only those from the cancer cells would greatly enhance the performance of the screening tests. The recent discovery that the biomarkers are carried by nanocarriers, such as exosomes, with cell-specific membrane proteins suggests that such selection may be possible, although a new suite of fractionation and quantification technologies would need to be developed. Although under-appreciated, membrane microfluidics has made considerable contributions to resolving these issues. We review the progress made so far, based on ion-selective, track-etched, and gel membranes and advanced electrophoretic and nano-filtration designs, in this perspective and suggest future directions.


Subject(s)
Biomarkers, Tumor/analysis , Early Detection of Cancer/methods , Extracellular Vesicles/metabolism , Liquid Biopsy/methods , Microfluidic Analytical Techniques/methods , Extracellular Vesicles/chemistry , High-Throughput Screening Assays/methods , Humans , Nanopores
SELECTION OF CITATIONS
SEARCH DETAIL
...