Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.847
Filter
Add more filters

Publication year range
1.
Cell ; 164(6): 1105-1109, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26967278

ABSTRACT

While studies of cultured cells have led to new insights into biological control, greater understanding of human pathophysiology requires the development of experimental systems that permit analysis of intercellular communications and tissue-tissue interactions in a more relevant organ context. Human organs-on-chips offer a potentially powerful new approach to confront this long-standing problem.


Subject(s)
Microfluidic Analytical Techniques/methods , Organ Culture Techniques , Tissue Engineering/methods , Blood-Brain Barrier , Humans , Neoplasms/physiopathology
2.
Nature ; 618(7967): 1057-1064, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37344592

ABSTRACT

Translation regulation is critical for early mammalian embryonic development1. However, previous studies had been restricted to bulk measurements2, precluding precise determination of translation regulation including allele-specific analyses. Here, to address this challenge, we developed a novel microfluidic isotachophoresis (ITP) approach, named RIBOsome profiling via ITP (Ribo-ITP), and characterized translation in single oocytes and embryos during early mouse development. We identified differential translation efficiency as a key mechanism regulating genes involved in centrosome organization and N6-methyladenosine modification of RNAs. Our high-coverage measurements enabled, to our knowledge, the first analysis of allele-specific ribosome engagement in early development. These led to the discovery of stage-specific differential engagement of zygotic RNAs with ribosomes and reduced translation efficiency of transcripts exhibiting allele-biased expression. By integrating our measurements with proteomics data, we discovered that ribosome occupancy in germinal vesicle-stage oocytes is the predominant determinant of protein abundance in the zygote. The Ribo-ITP approach will enable numerous applications by providing high-coverage and high-resolution ribosome occupancy measurements from ultra-low input samples including single cells.


Subject(s)
Embryonic Development , Isotachophoresis , Microfluidic Analytical Techniques , Protein Biosynthesis , Ribosome Profiling , Ribosomes , Single-Cell Analysis , Animals , Mice , Proteomics , Ribosomes/metabolism , RNA, Messenger/genetics , Single-Cell Analysis/methods , Alleles , Microfluidic Analytical Techniques/methods , Oocytes/growth & development , Oocytes/metabolism , Isotachophoresis/methods , Ribosome Profiling/methods , Centrosome , Zygote/growth & development , Zygote/metabolism
3.
Nature ; 605(7910): 464-469, 2022 05.
Article in English | MEDLINE | ID: mdl-35585345

ABSTRACT

Chain reactions, characterized by initiation, propagation and termination, are stochastic at microscopic scales and underlie vital chemical (for example, combustion engines), nuclear and biotechnological (for example, polymerase chain reaction) applications1-5. At macroscopic scales, chain reactions are deterministic and limited to applications for entertainment and art such as falling dominoes and Rube Goldberg machines. On the other hand, the microfluidic lab-on-a-chip (also called a micro-total analysis system)6,7 was visualized as an integrated chip, akin to microelectronic integrated circuits, yet in practice remains dependent on cumbersome peripherals, connections and a computer for automation8-11. Capillary microfluidics integrate energy supply and flow control onto a single chip by using capillary phenomena, but programmability remains rudimentary with at most a handful (eight) operations possible12-19. Here we introduce the microfluidic chain reaction (MCR) as the conditional, structurally programmed propagation of capillary flow events. Monolithic chips integrating a MCR are three-dimensionally printed, and powered by the free energy of a paper pump, autonomously execute liquid handling algorithms step-by-step. With MCR, we automated (1) the sequential release of 300 aliquots across chained, interconnected chips, (2) a protocol for severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) antibodies detection in saliva and (3) a thrombin generation assay by continuous subsampling and analysis of coagulation-activated plasma with parallel operations including timers, iterative cycles of synchronous flow and stop-flow operations. MCRs are untethered from and unencumbered by peripherals, encode programs structurally in situ and can form a frugal, versatile, bona fide lab-on-a-chip with wide-ranging applications in liquid handling and point-of-care diagnostics.


Subject(s)
COVID-19 , Microfluidic Analytical Techniques , Humans , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Microfluidics/methods , Polymerase Chain Reaction , SARS-CoV-2/genetics
4.
Proc Natl Acad Sci U S A ; 121(19): e2315168121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683997

ABSTRACT

Accurate prediction of the efficacy of immunotherapy for cancer patients through the characterization of both genetic and phenotypic heterogeneity in individual patient cells holds great promise in informing targeted treatments, and ultimately in improving care pathways and clinical outcomes. Here, we describe the nanoplatform for interrogating living cell host-gene and (micro-)environment (NICHE) relationships, that integrates micro- and nanofluidics to enable highly efficient capture of circulating tumor cells (CTCs) from blood samples. The platform uses a unique nanopore-enhanced electrodelivery system that efficiently and rapidly integrates stable multichannel fluorescence probes into living CTCs for in situ quantification of target gene expression, while on-chip coculturing of CTCs with immune cells allows for the real-time correlative quantification of their phenotypic heterogeneities in response to immune checkpoint inhibitors (ICI). The NICHE microfluidic device provides a unique ability to perform both gene expression and phenotypic analysis on the same single cells in situ, allowing us to generate a predictive index for screening patients who could benefit from ICI. This index, which simultaneously integrates the heterogeneity of single cellular responses for both gene expression and phenotype, was validated by clinically tracing 80 non-small cell lung cancer patients, demonstrating significantly higher AUC (area under the curve) (0.906) than current clinical reference for immunotherapy prediction.


Subject(s)
Neoplastic Cells, Circulating , Humans , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Microfluidics/methods , Single-Cell Analysis/methods , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/blood , Phenotype , Cell Line, Tumor , Immunotherapy/methods , Gene Expression Profiling/methods , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/blood , Microfluidic Analytical Techniques/methods , Microfluidic Analytical Techniques/instrumentation
5.
Nature ; 582(7811): 277-282, 2020 06.
Article in English | MEDLINE | ID: mdl-32349121

ABSTRACT

The great majority of globally circulating pathogens go undetected, undermining patient care and hindering outbreak preparedness and response. To enable routine surveillance and comprehensive diagnostic applications, there is a need for detection technologies that can scale to test many samples1-3 while simultaneously testing for many pathogens4-6. Here, we develop Combinatorial Arrayed Reactions for Multiplexed Evaluation of Nucleic acids (CARMEN), a platform for scalable, multiplexed pathogen detection. In the CARMEN platform, nanolitre droplets containing CRISPR-based nucleic acid detection reagents7 self-organize in a microwell array8 to pair with droplets of amplified samples, testing each sample against each CRISPR RNA (crRNA) in replicate. The combination of CARMEN and Cas13 detection (CARMEN-Cas13) enables robust testing of more than 4,500 crRNA-target pairs on a single array. Using CARMEN-Cas13, we developed a multiplexed assay that simultaneously differentiates all 169 human-associated viruses with at least 10 published genome sequences and rapidly incorporated an additional crRNA to detect the causative agent of the 2020 COVID-19 pandemic. CARMEN-Cas13 further enables comprehensive subtyping of influenza A strains and multiplexed identification of dozens of HIV drug-resistance mutations. The intrinsic multiplexing and throughput capabilities of CARMEN make it practical to scale, as miniaturization decreases reagent cost per test by more than 300-fold. Scalable, highly multiplexed CRISPR-based nucleic acid detection shifts diagnostic and surveillance efforts from targeted testing of high-priority samples to comprehensive testing of large sample sets, greatly benefiting patients and public health9-11.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Microfluidic Analytical Techniques/methods , Virus Diseases/diagnosis , Virus Diseases/virology , Animals , Betacoronavirus/genetics , Betacoronavirus/isolation & purification , Drug Resistance, Viral/genetics , Genome, Viral/genetics , HIV/classification , HIV/genetics , HIV/isolation & purification , Humans , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/isolation & purification , Microfluidic Analytical Techniques/instrumentation , RNA, Guide, Kinetoplastida/genetics , SARS-CoV-2 , Sensitivity and Specificity
6.
Annu Rev Biomed Eng ; 26(1): 119-139, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38316063

ABSTRACT

Recent advances in single-cell and multicellular microfluidics technology have provided powerful tools for studying cancer biology and immunology. The ability to create controlled microenvironments, perform high-throughput screenings, and monitor cellular interactions at the single-cell level has significantly advanced our understanding of tumor biology and immune responses. We discuss cutting-edge multicellular and single-cell microfluidic technologies and methodologies utilized to investigate cancer-immune cell interactions and assess the effectiveness of immunotherapies. We explore the advantages and limitations of the wide range of 3D spheroid and single-cell microfluidic models recently developed, highlighting the various approaches in device generation and applications in immunotherapy screening for potential opportunities for point-of-care approaches.


Subject(s)
Microfluidics , Neoplasms , Point-of-Care Systems , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Microfluidics/methods , Tumor Microenvironment , Immunotherapy/methods , Spheroids, Cellular , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Cell Communication , Animals , Lab-On-A-Chip Devices
7.
Chem Rev ; 123(9): 5571-5611, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37126602

ABSTRACT

Novel and improved biocatalysts are increasingly sourced from libraries via experimental screening. The success of such campaigns is crucially dependent on the number of candidates tested. Water-in-oil emulsion droplets can replace the classical test tube, to provide in vitro compartments as an alternative screening format, containing genotype and phenotype and enabling a readout of function. The scale-down to micrometer droplet diameters and picoliter volumes brings about a >107-fold volume reduction compared to 96-well-plate screening. Droplets made in automated microfluidic devices can be integrated into modular workflows to set up multistep screening protocols involving various detection modes to sort >107 variants a day with kHz frequencies. The repertoire of assays available for droplet screening covers all seven enzyme commission (EC) number classes, setting the stage for widespread use of droplet microfluidics in everyday biochemical experiments. We review the practicalities of adapting droplet screening for enzyme discovery and for detailed kinetic characterization. These new ways of working will not just accelerate discovery experiments currently limited by screening capacity but profoundly change the paradigms we can probe. By interfacing the results of ultrahigh-throughput droplet screening with next-generation sequencing and deep learning, strategies for directed evolution can be implemented, examined, and evaluated.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics , Microfluidics/methods , Biological Assay , Emulsions , Water , Kinetics , High-Throughput Screening Assays/methods , Microfluidic Analytical Techniques/methods
8.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Article in English | MEDLINE | ID: mdl-35074872

ABSTRACT

Cell-cell interactions are important to numerous biological systems, including tissue microenvironments, the immune system, and cancer. However, current methods for studying cell combinations and interactions are limited in scalability, allowing just hundreds to thousands of multicell assays per experiment; this limited throughput makes it difficult to characterize interactions at biologically relevant scales. Here, we describe a paradigm in cell interaction profiling that allows accurate grouping of cells and characterization of their interactions for tens to hundreds of thousands of combinations. Our approach leverages high-throughput droplet microfluidics to construct multicellular combinations in a deterministic process that allows inclusion of programmed reagent mixtures and beads. The combination droplets are compatible with common manipulation and measurement techniques, including imaging, barcode-based genomics, and sorting. We demonstrate the approach by using it to enrich for chimeric antigen receptor (CAR)-T cells that activate upon incubation with target cells, a bottleneck in the therapeutic T cell engineering pipeline. The speed and control of our approach should enable valuable cell interaction studies.


Subject(s)
Biological Assay/methods , Cell Communication/physiology , Microfluidic Analytical Techniques/methods , Microfluidics/methods , Animals , Cell Communication/genetics , Genomics/methods , Humans
9.
J Proteome Res ; 23(8): 3096-3107, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38417049

ABSTRACT

Fluorescence-activated cell sorting (FACS) is a specialized technique to isolate specific cell subpopulations with a high level of recovery and accuracy. However, the cell sorting procedure can impact the viability and metabolic state of cells. Here, we performed a comparative study and evaluated the impact of traditional high-pressure charged droplet-based and microfluidic chip-based sorting on the metabolic and phosphoproteomic profile of different cell types. While microfluidic chip-based sorted cells more closely resembled the unsorted control group for most cell types tested, the droplet-based sorted cells showed significant metabolic and phosphoproteomic alterations. In particular, greater changes in redox and energy status were present in cells sorted with the droplet-based cell sorter along with larger shifts in proteostasis. 13C-isotope tracing analysis on cells recovering postsorting revealed that the sorter-induced suppression of mitochondrial TCA cycle activity recovered faster in the microfluidic chip-based sorted group. Apart from this, amino acid and lipid biosynthesis pathways were suppressed in sorted cells, with minimum impact and faster recovery in the microfluidic chip-based sorted group. These results indicate microfluidic chip-based sorting has a minimum impact on metabolism and is less disruptive compared to droplet-based sorting.


Subject(s)
Flow Cytometry , Multiomics , Animals , Humans , Cell Separation/methods , Citric Acid Cycle , Flow Cytometry/methods , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Microfluidic Analytical Techniques/instrumentation , Microfluidics/methods , Proteomics/methods
10.
Anal Chem ; 96(15): 5815-5823, 2024 04 16.
Article in English | MEDLINE | ID: mdl-38575144

ABSTRACT

Microfluidic techniques are widely applied in biomolecular analysis and disease diagnostic assays. While the volume of the sample that is directly used in such assays is often only femto-to microliters, the "dead volume" of solutions supplied in syringes and tubing can be much larger, even up to milliliters, increasing overall reagent use and making analysis significantly more expensive. To reduce the difficulty and cost, we designed a new chip using a low volume solution for analysis and applied it to obtain real-time data for protein-protein interaction measurements. The chip takes advantage of air/aqueous two-phase droplet flow, on-chip rapid mixing within milliseconds, and a droplet capture method, that ultimately requires only 2 µL of reagent solution. The interaction is analyzed by particle diffusometry, a nonintrusive and precise optical detection method to analyze the properties of microparticle diffusion in solution. Herein, we demonstrate on-chip characterization of human immunodeficiency virus p24 antibody-antigen protein binding kinetics imaged via fluorescence microscopy and analyzed by PD. The measured kon and koff are 1 × 106 M-1 s-1 and 3.3 × 10-4 s-1, respectively, and agree with independent measurement via biolayer interferometry and previously calculated p24-antibody binding kinetics. This new microfluidic chip and the protein-protein interaction analysis method can also be applied in other fields that require low-volume solutions to perform accurate measurement, analysis, and detection.


Subject(s)
Microfluidic Analytical Techniques , Microfluidics , Humans , Kinetics , Diffusion , Indicators and Reagents , Microfluidic Analytical Techniques/methods
11.
Anal Chem ; 96(2): 766-774, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38158582

ABSTRACT

Microfluidic chips have emerged as a promising tool for sorting and enriching circulating tumor cells (CTCs) in blood, while the efficacy and purity of CTC sorting greatly depend on chip design. Herein, a novel cascaded phase-transfer microfluidic chip was developed for high-efficiency sorting, purification, release, and detection of MCF-7 cells (as a model CTC) in blood samples. MCF-7 cells were specifically captured by EpCAM aptamer-modified magnetic beads and then introduced into the designed cascaded phase-transfer microfluidic chip that consisted of three functional regions (sorting, purification, and release zone). In the sorting zone, the MCF-7 cells moved toward the inner wall of the channel and entered the purification zone for primary separation from white blood cells; in the purification zone, the MCF-7 cells were transferred to the phosphate-buffered saline flow under the interaction of Dean forces and central magnetic force, achieving high purification of MCF-7 cells from blood samples; in the release zone, MCF-7 cells were further transferred into the nuclease solution and fixed in groove by the strong magnetic force and hydrodynamic force, and the continuously flowing nuclease solution cleaved the aptamer on the trapped MCF-7 cells, causing gentle release of MCF-7 cells for subsequent inductively coupled plasma mass spectrometry (ICP-MS) detection or further cultivation. By measurement of the endogenous element Zn in the cells using ICP-MS for cell counting, an average cell recovery of 84% for MCF-7 cells was obtained in spiked blood samples. The developed method was applied in the analysis of real blood samples from healthy people and breast cancer patients, and CTCs were successfully detected in all tested patient samples (16/16). Additionally, the removal of the magnetic probes on the cell surface significantly improved cell viability up to 99.3%. Therefore, the developed cascaded phase-transfer microfluidic chip ICP-MS system possessed high integration for CTCs analysis with high cell viability, cell recovery, and purity, showing great advantages in early clinical cancer diagnosis.


Subject(s)
Microfluidic Analytical Techniques , Neoplastic Cells, Circulating , Humans , Neoplastic Cells, Circulating/pathology , Microfluidics , Cell Separation/methods , Cell Line, Tumor , Microfluidic Analytical Techniques/methods , Magnetic Phenomena
12.
Anal Chem ; 96(4): 1622-1629, 2024 01 30.
Article in English | MEDLINE | ID: mdl-38215213

ABSTRACT

The microfluidic chip-based nucleic acid detection method significantly improves the sensitivity since it precisely controls the microfluidic flow in microchannels. Nonetheless, significant challenges still exist in improving the detection efficiency to meet the demand for rapid detection of trace substances. This work provides a novel magnetic herringbone (M-HB) structure in a microfluidic chip, and its advantage in rapid and sensitive detection is verified by taking complementary DNA (cDNA) sequences of human immunodeficiency virus (HIV) detection as an example. The M-HB structure is designed based on controlling the magnetic field distribution in the micrometer scale and is formed by accumulation of magnetic microbeads (MMBs). Hence, M-HB is similar to a nanopore microstructure, which has a higher contact area and probe density. All of the above is conducive to improving sensitivity in microfluidic chips. The M-HB chip is stable and easy to form, which can linearly detect cDNA sequences of HIV quantitatively ranging from 1 to 20 nM with a detection limit of 0.073 nM. Compared to the traditional herringbone structure, this structure is easier to form and release by controlling the magnetic field, which is flexible and helps in further study. Results show that this chip can sensitively detect the cDNA sequences of HIV in blood samples, demonstrating that it is a powerful platform to rapidly and sensitively detect multiple nucleic acid-related viruses of infectious diseases.


Subject(s)
HIV Infections , Microfluidic Analytical Techniques , Humans , DNA, Complementary , Microspheres , HIV , Magnetic Phenomena , HIV Infections/diagnosis , Microfluidic Analytical Techniques/methods
13.
Small ; 20(26): e2308950, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38441226

ABSTRACT

Monoclonal antibody (mAb) discovery plays a prominent role in diagnostic and therapeutic applications. Droplet microfluidics has become a standard technology for high-throughput screening of antibody-producing cells due to high droplet single-cell confinement frequency and rapid analysis and sorting of the cells of interest with their secreted mAbs. In this work, a new method is described for on-demand co-encapsulation of cells that eliminates the difficulties associated with washing in between consecutive steps inside the droplets and enables the washing and addition of fresh media. The new platform identifies hybridoma cells that are expressing antibodies of interest using antibody-characterization assays to find the best-performing or rare-cell antibody candidates.


Subject(s)
Antibodies, Monoclonal , Microfluidics , Antibodies, Monoclonal/chemistry , Microfluidics/methods , Animals , Hybridomas/cytology , Single-Cell Analysis/methods , Mice , Humans , Automation , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
14.
Int J Exp Pathol ; 105(3): 90-99, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38717047

ABSTRACT

Management of lung cancer today obligates a mutational analysis of the epidermal growth factor receptor (EGFR) gene particularly when Tyrosine Kinase Inhibitor (TKI) therapy is being considered as part of prognostic stratification. This study evaluates the performance of automated microfluidics-based EGFR mutation detection and its significance in clinical diagnostic settings. Formalin-fixed, paraffin-embedded (FFPE) samples from NSCLC patients (n = 174) were included in a two-phase study. Phase I: Validation of the platform by comparing the results with conventional real-time PCR and next-generation sequencing (NGS) platform. Phase II: EGFR mutation detection on microfluidics-based platform as part of routine diagnostics workup. The microfluidics-based platform demonstrates 96.5% and 89.2% concordance with conventional real-time PCR and NGS, respectively. The system efficiently detects mutations across the EGFR gene with 88.23% sensitivity and 100% specificity. Out of 144 samples analysed in phase II, the platform generated valid results in 94% with mutation detected in 41% of samples. This microfluidics-based platform can detect as low as 5% mutant allele fractions from the FFPE samples. Therefore the microfluidics-based platform is a rapid, complete walkaway, with minimum tissue requirement (two sections of 5 µ thickness) and technical skill requirement. The method can detect clinically actionable EGFR mutations efficiently and can be considered a reliable diagnostic platform in resource-limited settings. From receiving samples to reporting the results this platform provides accurate data without much manual intervention. The study helped to devise an algorithm that emphasizes effective screening of the NSCLC cases for EGFR mutations with varying tumour content. Thus it helps in triaging the cases judiciously before proceeding with multigene testing.


Subject(s)
Carcinoma, Non-Small-Cell Lung , ErbB Receptors , High-Throughput Nucleotide Sequencing , Lung Neoplasms , Mutation , Humans , ErbB Receptors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/genetics , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Microfluidics/methods , Real-Time Polymerase Chain Reaction/methods , Microfluidic Analytical Techniques/methods , Paraffin Embedding
15.
Electrophoresis ; 45(1-2): 69-100, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37259641

ABSTRACT

Proteins are important molecules involved in an immensely large number of biological processes. Being capable of manipulating proteins is critical for developing reliable and affordable techniques to analyze and/or detect them. Such techniques would enable the production of therapeutic agents for the treatment of diseases or other biotechnological applications (e.g., bioreactors or biocatalysis). Microfluidic technology represents a potential solution to protein manipulation challenges because of the diverse phenomena that can be exploited to achieve micro- and nanoparticle manipulation. In this review, we discuss recent contributions made in the field of protein manipulation in microfluidic systems using different physicochemical principles and techniques, some of which are miniaturized versions of already established macro-scale techniques.


Subject(s)
Microfluidic Analytical Techniques , Nanoparticles , Microfluidics/methods , Microfluidic Analytical Techniques/methods , Nanoparticles/chemistry , Lab-On-A-Chip Devices
16.
Electrophoresis ; 45(13-14): 1233-1242, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38161241

ABSTRACT

The efficient and precise extraction of target particles is a crucial prerequisite for achieving accurate detection and analysis in microfluidic cell analysis. In this study, a symmetrical contraction-expansion microchannel with sheath flow was designed, aiming to extract target larger particles from particles of different sizes within the channel. This paper conducted numerical simulations to investigate the three-dimensional migration mechanisms of particles and performed experimental studies to examine the separation performance of particles with different sizes under varying flow rate ratios and different numbers of contraction-expansion structures. The experimental results indicate that at moderate sample flow rates and higher flow rate ratios, microchannels with fewer contraction-expansion structures are likely to achieve better performance in extracting target particles compared to microchannels with a greater number of these structures. Our work advances the application of viscoelastic contraction-expansion microchannels in particle separation. This device is easy to set up in parallel and significantly enhances throughput, providing an accurate and efficient solution for future particle separation applications.


Subject(s)
Equipment Design , Microfluidic Analytical Techniques , Particle Size , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Computer Simulation , Viscosity
17.
Electrophoresis ; 45(15-16): 1428-1442, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38430203

ABSTRACT

To explore the optoelectronic wetting droplet transport mechanism, a transient numerical model of optoelectrowetting (OEW) under the coupling of flow and electric fields is established. The study investigates the impact of externally applied voltage, dielectric constant of the dielectric layer, and interfacial tension between the two phases on the dynamic behavior of droplets during transport. The proposed model employs an improved Young's equation to calculate the instantaneous voltage and contact angle of the droplet on the dielectric layer. Results indicate that, under the influence of OEW, significant variations in the interface contact angle of droplets occur in bright and dark regions, inducing droplet movement. Moreover, the dynamic behavior of droplet transport is closely associated with various parameters, including externally applied voltage, dielectric layer material, and interfacial tension between the two phases, all of which impact the contact angle and, consequently, the transport process. By summarizing the influence patterns of the three key parameters studied, the optimization of droplet transport performance is achieved. The study employs two-dimensional simulation models to emulate the droplet motion under the influence of the electric field, investigating the OEW droplet transport mechanism. The continuous movement of droplets involves three stages: initial wetting, continuous transport, and reaching a steady position. The findings contribute theoretical support for the efficient design of digital microfluidic devices for OEW droplet movement and the selection of key parameters for droplet manipulation.


Subject(s)
Wettability , Models, Theoretical , Computer Simulation , Surface Tension , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
18.
Electrophoresis ; 45(7-8): 752-763, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38143284

ABSTRACT

We report the possibilities of achieving highly controlled segregation of ion-enriched and ion-depleted regions in straight nanochannels. This is achieved via harnessing the interplay of an axial gradient of the induced transverse electric field on account of electrical double layer phenomenon and the localized thickening of the fluid because of intensified electric fields due to the large spatial gradients of the electrical potential in extreme confinements. By considering alternate surface patches of different charge densities over pre-designed axial spans, we illustrate how these effects can be exploited to realize selectively ion-enriched and ion-depleted zones. Physically, this is attributed to setting up of an axial concentration gradient that delves on the ionic advection due to the combined effect of an externally applied electric field and induced back-pressure gradient along the channel axis and electro-migration due to the combinatorial influences of the applied and the induced electrostatic fields. With an explicit handle on the pertinent parameters, our results offer insights on the possible means of imposing delicate controls on the solute-enrichment and depletion phenomena, a paradigm that remained unexplored thus far.


Subject(s)
Ions , Static Electricity , Ions/chemistry , Nanotechnology/methods , Surface Properties , Nanostructures/chemistry , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Electromagnetic Fields
19.
Electrophoresis ; 45(15-16): 1379-1388, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38343171

ABSTRACT

Most biological fluids (such as blood, saliva, and lymph) in nature have certain viscoelasticity and are beginning to be used as the carrying fluids for viscoelastic microfluidics. However, the particle-focusing mechanisms in these new biological viscoelastic fluids are still unclear. In this work, the particle-focusing mechanisms in λ-DNA solutions were systematically explored. We first explored the particle focusing dynamics in a square cross-section under varied flow rates to uncover the effects of flow rate on particle focusing. Three focusing stages, from the classic five-position viscoelastic focusing to single-stream focusing and finally to multiplex-stream focusing, were clearly demonstrated. In addition, the particle focusing process along the channel length was demonstrated, and a first-fast-and-then-slow focusing process was clearly observed. Then, the effects of λ-DNA concentrations on particle focusing were explored and compared using the solutions with 0-25 ppm λ-DNA. Finally, we discussed the inferences of blockage ratio on particle focusing by changing the particle diameter and cross-sectional dimensions. Our work may provide a deeper understanding on the particle focusing mechanisms in biological viscoelastic fluids and lays a foundation for the subsequent particle counting and analysis and the development of low-cost portable flow cytometers.


Subject(s)
Microfluidic Analytical Techniques , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Viscosity , DNA, Viral/analysis , Particle Size , Bacteriophage lambda/chemistry , Equipment Design
20.
Electrophoresis ; 45(7-8): 676-686, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38350722

ABSTRACT

Understanding electrokinetic transport in nanochannels and nanopores is essential for emerging biological and electrochemical applications. The viscoelectric effect is an important mechanism implicated in the increase of local viscosity due to the polarization of a solvent under a strong electric field. However, most analyses of the viscoelectric effect have been limited to numerical analyses. In this work, we present a set of analytical solutions applicable to the physical description of viscoelectric effects in nanochannel electrokinetic systems. To achieve such closed-form solutions, we employ the Debye-Hückel approximation of small diffuse charge layer potentials compared to the thermal potential. We analyze critical parameters, including electroosmotic flow profiles, electroosmotic mobility, flow rate, and channel conductance. We compare and benchmark our analytical solutions with published predictions from numerical models. Importantly, we leverage these analytical solutions to identify essential thermophysical and nondimensional parameters that govern the behavior of these systems. We identify scaling parameters and relations among surface charge density, ionic strength, and nanochannel height.


Subject(s)
Electroosmosis , Electroosmosis/methods , Viscosity , Nanotechnology/methods , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Nanopores , Osmolar Concentration , Nanostructures/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL