1
|
Desire CT, Arrua RD, Strudwick XL, Kopecki Z, Cowin AJ, Hilder EF. The development of microfluidic-based western blotting: Technical advances and future perspectives. J Chromatogr A 2023; 1691:463813. [PMID: 36709548 DOI: 10.1016/j.chroma.2023.463813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
Over the past two decades significant technical advancement in the field of western blotting has been made possible through the utilization of microfluidic technologies. In this review we provide a critical overview of these advancements, highlighting the advantages and disadvantages of each approach. Particular attention is paid to the development of now commercially available systems, including those for single cell analysis. This review also discusses more recent developments, including algorithms for automation and/or improved quantitation, the utilization of different materials/chemistries, use of projection electrophoresis, and the development of triBlots. Finally, the review includes commentary on future advances in the field based on current developments, and the potential of these systems for use as point-of-care devices in healthcare.
Collapse
Affiliation(s)
- Christopher T Desire
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - R Dario Arrua
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Zlatko Kopecki
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Emily F Hilder
- Future Industries Institute, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia.
| |
Collapse
|
2
|
Booth PPM, Lamb DT, Anderson JP, Furtaw MD, Kennedy RT. Capillary electrophoresis Western blot using inkjet transfer to membrane. J Chromatogr A 2022; 1679:463389. [PMID: 35933772 DOI: 10.1016/j.chroma.2022.463389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/15/2022] [Accepted: 07/29/2022] [Indexed: 10/16/2022]
Abstract
Traditional Western blots are commonly used to separate and assay proteins; however, they have limitations including a long, cumbersome process and large sample requirements. Here, we describe a system for Western blotting where capillary gel electrophoresis is used to separate sodium dodecyl sulfate-protein complexes. The capillary outlet is threaded into a piezoelectric inkjetting head that deposits the separated proteins in a quasi-continuous stream of <100 pL droplets onto a moving membrane. Through separations at 400 V/cm and protein capture on a membrane moving at 2 mm/min, we are able to detect actin with a limit of detection at 8 pM, or an estimated 5 fg injected. Separation and membrane capture of sample containing 10 proteins ranging in molecular weights from 11 - 250 kDa was achieved in 15 min. The system was demonstrated with Western blots for actin, β-tubulin, ERK1/2, and STAT3 in human A431 epidermoid carcinoma cell lysate.
Collapse
|
3
|
Xie H, Ding X. The Intriguing Landscape of Single-Cell Protein Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105932. [PMID: 35199955 PMCID: PMC9036017 DOI: 10.1002/advs.202105932] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/27/2022] [Indexed: 05/15/2023]
Abstract
Profiling protein expression at single-cell resolution is essential for fundamental biological research (such as cell differentiation and tumor microenvironmental examination) and clinical precision medicine where only a limited number of primary cells are permitted. With the recent advances in engineering, chemistry, and biology, single-cell protein analysis methods are developed rapidly, which enable high-throughput and multiplexed protein measurements in thousands of individual cells. In combination with single cell RNA sequencing and mass spectrometry, single-cell multi-omics analysis can simultaneously measure multiple modalities including mRNAs, proteins, and metabolites in single cells, and obtain a more comprehensive exploration of cellular signaling processes, such as DNA modifications, chromatin accessibility, protein abundance, and gene perturbation. Here, the recent progress and applications of single-cell protein analysis technologies in the last decade are summarized. Current limitations, challenges, and possible future directions in this field are also discussed.
Collapse
Affiliation(s)
- Haiyang Xie
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| |
Collapse
|
4
|
Eills J, Hale W, Utz M. Synergies between Hyperpolarized NMR and Microfluidics: A Review. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2022; 128:44-69. [PMID: 35282869 DOI: 10.1016/j.pnmrs.2021.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 06/14/2023]
Abstract
Hyperpolarized nuclear magnetic resonance and lab-on-a-chip microfluidics are two dynamic, but until recently quite distinct, fields of research. Recent developments in both areas increased their synergistic overlap. By microfluidic integration, many complex experimental steps can be brought together onto a single platform. Microfluidic devices are therefore increasingly finding applications in medical diagnostics, forensic analysis, and biomedical research. In particular, they provide novel and powerful ways to culture cells, cell aggregates, and even functional models of entire organs. Nuclear magnetic resonance is a non-invasive, high-resolution spectroscopic technique which allows real-time process monitoring with chemical specificity. It is ideally suited for observing metabolic and other biological and chemical processes in microfluidic systems. However, its intrinsically low sensitivity has limited its application. Recent advances in nuclear hyperpolarization techniques may change this: under special circumstances, it is possible to enhance NMR signals by up to 5 orders of magnitude, which dramatically extends the utility of NMR in the context of microfluidic systems. Hyperpolarization requires complex chemical and/or physical manipulations, which in turn may benefit from microfluidic implementation. In fact, many hyperpolarization methodologies rely on processes that are more efficient at the micro-scale, such as molecular diffusion, penetration of electromagnetic radiation into a sample, or restricted molecular mobility on a surface. In this review we examine the confluence between the fields of hyperpolarization-enhanced NMR and microfluidics, and assess how these areas of research have mutually benefited one another, and will continue to do so.
Collapse
Affiliation(s)
- James Eills
- Institute for Physics, Johannes Gutenberg University, D-55090 Mainz, Germany; GSI Helmholtzzentrum für Schwerionenforschung GmbH, Helmholtz-Institut Mainz, 55128 Mainz, Germany.
| | - William Hale
- Department of Chemistry, University of Florida, 32611, USA
| | - Marcel Utz
- School of Chemistry, University of Southampton, SO17 1BJ, UK.
| |
Collapse
|
5
|
Maqbool I, Ali SI, Paul V, Muzaffer U. Immunognetics: a tool for the identification of novel therapeutic targets in immune disorders. CLINICAL APPLICATIONS OF IMMUNOGENETICS 2022:355-376. [DOI: 10.1016/b978-0-323-90250-2.00015-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
He JY, Chen ZH, Deng HL, Yuan R, Xu WJ. Antibody-powered DNA switches to initiate the hybridization chain reaction for the amplified fluorescence immunoassay. Analyst 2021; 146:5067-5073. [PMID: 34297024 DOI: 10.1039/d1an01045j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Designing antibody-powered DNA nanodevice switches is crucial and fascinating to perform a variety of functions in response to specific antibodies as regulatory inputs, achieving highly sensitive detection by integration with simple amplified methods. In this work, we report a unique DNA-based conformational switch, powered by a targeted anti-digoxin mouse monoclonal antibody (anti-Dig) as a model, to rationally initiate the hybridization chain reaction (HCR) for enzyme-free signal amplification. As a proof-of-concept, both a fluorophore Cy3-labeled reporter hairpin (RH) in the 3' terminus and a single-stranded helper DNA (HS) were individually hybridized with a recognition single-stranded DNA (RS) modified with Dig hapten, while the unpaired loop of RH was hybridized with the exposed 3'-toehold of HS, isothermally self-assembling an intermediate metastable DNA structure. The introduction of target anti-Dig drove the concurrent conjugation with two tethered Dig haptens, powering the directional switch of this DNA structure into a stable conformation. In this case, the unlocked 3'-stem of RH was implemented to unfold the 5'-stem of the BHQ-2-labeled quench hairpin (QH), rationally initiating the HCR between them by the overlapping complementary hybridization. As a result, numerous pairs of Cy3 and BHQ-2 in the formed long double helix were located in spatial proximity. In response to this, the significant quenching of the fluorescence intensity of Cy3 by BHQ-2 was dependent on the variable concentration of anti-Dig, achieving a highly sensitive quantification down to the picomolar level based on a simplified protocol integrated with enzyme-free amplification.
Collapse
Affiliation(s)
- Jia-Yang He
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China.
| | - Ze-Hui Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China.
| | - Hui-Lin Deng
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China.
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China.
| | - Wen-Ju Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R. China.
| |
Collapse
|
7
|
Shlyapnikov YM, Kanev IL, Shlyapnikova EA. Rapid Ultrasensitive Gel-Free Immunoblotting with Magnetic Labels. Anal Chem 2020; 92:4146-4153. [PMID: 32023039 DOI: 10.1021/acs.analchem.0c00314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Immunoblotting is widely used for the detection of proteins using specific antibodies. We present here a new immunoblotting method, which is characterized by exceptional sensitivity, rapidness, and low consumption of antibodies. A thin conductive layer between touching hydrophilic cellulose membranes instead of polyacrylamide gel is used for the electrophoretic separation of proteins. Contrary to common Western blotting, the separation occurs in nondenaturing conditions. The membrane surface is smoothed by deposition of the cellulose layer and modified with azidophenyl groups, allowing for the photochemical in situ immobilization of proteins, which are carried out after the electrophoresis. Thus, the additional step of transferring the protein from the gel onto the membrane is eliminated. Specific protein bands are then visualized by decoration with magnetic beads. The limit of detection of interleukin IL-1β reaches 0.3 fg or ∼104 molecules, whereas the total blotting time is about 5 min. The application of the technique is demonstrated by the detection of IL-1β, total IgA, and IgA specific to Mycobacterium tuberculosis antigen in the exhaled breath samples, obtained from healthy subjects and tuberculosis patients.
Collapse
Affiliation(s)
- Yuri M Shlyapnikov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Igor L Kanev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| | - Elena A Shlyapnikova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290 Russia
| |
Collapse
|
8
|
Shin K, Yu H, Kim J. Determination of diffusion coefficient and partition coefficient of photoinitiator 2-hydroxy-2-methylpropiophenone in nanoporous polydimethylsiloxane network and aqueous poly(ethylene glycol) diacrylate solution. J IND ENG CHEM 2017. [DOI: 10.1016/j.jiec.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
9
|
Clayton KN, Lee D, Wereley ST, Kinzer-Ursem TL. Measuring biotherapeutic viscosity and degradation on-chip with particle diffusometry. LAB ON A CHIP 2017; 17:4148-4159. [PMID: 29115357 DOI: 10.1039/c7lc00507e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In the absence of efficient ways to test drug stability and efficacy, pharmaceuticals that have been stored outside of set temperature conditions are destroyed, often at great cost. This is especially problematic for biotherapeutics, which are highly sensitive to temperature fluctuations. Current platforms for assessing the stability of protein-based biotherapeutics in high throughput and in low volumes are unavailable outside of research and development laboratories and are not efficient for use in production, quality control, distribution, or clinical settings. In these alternative environments, microanalysis platforms could provide significant advantages for the characterization of biotherapeutic degradation. Here we present particle diffusometry (PD), a new technique to study degradation of biotherapeutic solutions. PD uses a simple microfluidic chip and microscope setup to calculate the Brownian motion of particles in a quiescent solution using a variation of particle image velocimetry (PIV) fundamentals. We show that PD can be used to measure the viscosity of protein solutions to discriminate native protein from degraded samples as well as to determine the change in viscosity as a function of therapeutic concentration. PD viscosity analysis is applied to two particularly important biotherapeutic preparations: insulin, a commonly used protein for diabetic patients, and monoclonal antibodies which are an emerging class of biotherapeutics used to treat a variety of diseases such as autoimmune disorders and cancer. PD-based characterization of solution viscosity is a new tool for biotherapeutic analysis, and owing to its easy setup could readily be implemented at key points of the pharmaceutical delivery chain and in clinical settings.
Collapse
Affiliation(s)
- K N Clayton
- School of Mechanical Engineering and Birck Nanotechnology Center, Purdue University, West Lafayette, 47907, USA
| | | | | | | |
Collapse
|
10
|
Abdel-Sayed P, Yamauchi KA, Gerver RE, Herr AE. Fabrication of an Open Microfluidic Device for Immunoblotting. Anal Chem 2017; 89:9643-9648. [PMID: 28825964 DOI: 10.1021/acs.analchem.7b02406] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Given the wide adoption of polydimethylsiloxane (PDMS) for the rapid fabrication of microfluidic networks and the utility of polyacrylamide gel electrophoresis (PAGE), we develop a technique for fabrication of PAGE molecular sieving gels in PDMS microchannel networks. In developing the fabrication protocol, we trade-off constraints on materials properties of these two polymer materials: PDMS is permeable to O2 and the presence of O2 inhibits the polymerization of polyacrylamide. We present a fabrication method compatible with performing PAGE protein separations in a composite PDMS-glass microdevice, that toggles from an "enclosed" microchannel for PAGE and blotting to an "open" PA gel lane for immunoprobing and readout. To overcome the inhibitory effects of O2, we coat the PDMS channel with a 10% benzophenone solution, which quenches the inhibiting effect of O2 when exposed to UV, resulting in a PAGE-in-PDMS device. We then characterize the PAGE separation performance. Using a ladder of small-to-mid mass proteins (Trypsin Inhibitor (TI); Ovalbumin (OVA); Bovine Serum Albumin (BSA)), we observe resolution of the markers in <60 s, with separation resolution exceeding 1.0 and CVs of 8.4% for BSA-OVA and 2.4% for OVA-TI, with comparable reproducibility to glass microdevice PAGE. We show that benzophenone groups incorporated into the gel through methacrylamide can be UV-activated multiple times to photocapture protein. PDMS microchannel network is reversibly bonded to a glass slide allowing direct access to separated proteins and subsequent in situ diffusion-driven immunoprobing and total protein Sypro red staining. We see this PAGE-in-PDMS fabrication technique as expanding the application and use of microfluidic PAGE without the need for a glass microfabrication infrastructure.
Collapse
Affiliation(s)
- Philippe Abdel-Sayed
- Department of Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| | - Kevin A Yamauchi
- Department of Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| | - Rachel E Gerver
- Department of Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| | - Amy E Herr
- Department of Bioengineering, University of California Berkeley , Berkeley, California 94720, United States
| |
Collapse
|
11
|
Sanders BJ, Kim DC, Dunn RC. Recent Advances in Microscale Western Blotting. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2016; 8:7002-7013. [PMID: 28392839 PMCID: PMC5383213 DOI: 10.1039/c6ay01947a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Western blotting is a ubiquitous tool used extensively in the clinical and research settings to identify proteins and characterize their levels. It has rapidly become a mainstay in research laboratories due to its specificity, low cost, and ease of use. The specificity arises from the orthogonal processes used to identify proteins. Samples are first separated based on size and then probed with antibodies specific for the protein of interest. This confirmatory approach helps avoid pitfalls associated with antibody cross-reactivity and specificity issues. While the technique has evolved since its inception, the last decade has witnessed a paradigm shift in Western blotting technology. The introduction of capillary and microfluidic platforms has significantly decreased time and sample requirements while enabling high-throughput capabilities. These advances have enabled Western analysis down to the single cell level in highly parallel formats, opening vast new opportunities for studying cellular heterogeneity. Recent innovations in microscale Western blotting are surveyed, and the potential for enhancing detection using advances in label-free biosensing is briefly discussed.
Collapse
Affiliation(s)
- Brittany J Sanders
- Ralph Adams Institute of Bioanalytical Chemistry, Department of Chemistry, University of Kansas
| | - Daniel C Kim
- Ralph Adams Institute of Bioanalytical Chemistry, Department of Chemistry, University of Kansas
| | - Robert C Dunn
- Ralph Adams Institute of Bioanalytical Chemistry, Department of Chemistry, University of Kansas
| |
Collapse
|
12
|
Potential use of SERS-assisted theranostic strategy based on Fe3O4/Au cluster/shell nanocomposites for bio-detection, MRI, and magnetic hyperthermia. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 64:199-207. [DOI: 10.1016/j.msec.2016.03.090] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/01/2016] [Accepted: 03/23/2016] [Indexed: 01/01/2023]
|
13
|
Jin S, Furtaw MD, Chen H, Lamb DT, Ferguson SA, Arvin NE, Dawod M, Kennedy RT. Multiplexed Western Blotting Using Microchip Electrophoresis. Anal Chem 2016; 88:6703-10. [PMID: 27270033 DOI: 10.1021/acs.analchem.6b00705] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Western blotting is a commonly used protein assay that combines the selectivity of electrophoretic separation and immunoassay. The technique is limited by long time, manual operation with mediocre reproducibility, and large sample consumption, typically 10-20 μg per assay. Western blots are also usually used to measure only one protein per assay with an additional housekeeping protein for normalization. Measurement of multiple proteins is possible; however, it requires stripping membranes of antibody and then reprobing with a second antibody. Miniaturized alternatives to Western blot based on microfluidic or capillary electrophoresis have been developed that enable higher-throughput, automation, and greater mass sensitivity. In one approach, proteins are separated by electrophoresis on a microchip that is dragged along a polyvinylidene fluoride membrane so that as proteins exit the chip they are captured on the membrane for immunoassay. In this work, we improve this method to allow multiplexed protein detection. Multiple injections made from the same sample can be deposited in separate tracks so that each is probed with a different antibody. To further enhance multiplexing capability, the electrophoresis channel dimensions were optimized for resolution while keeping separation and blotting times to less than 8 min. Using a 15 μm deep × 50 μm wide × 8.6 cm long channel, it is possible to achieve baseline resolution of proteins that differ by 5% in molecular weight, e.g., ERK1 (44 kDa) from ERK2 (42 kDa). This resolution allows similar proteins detected by cross-reactive antibodies in a single track. We demonstrate detection of 11 proteins from 9 injections from a single Jurkat cell lysate sample consisting of 400 ng of total protein using this procedure. Thus, multiplexed Western blots are possible without cumbersome stripping and reprobing steps.
Collapse
Affiliation(s)
- Shi Jin
- Department of Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Michael D Furtaw
- LI-COR Biosciences , 4647 Superior Street, Lincoln, Nebraska 68504, United States
| | - Huaxian Chen
- LI-COR Biosciences , 4647 Superior Street, Lincoln, Nebraska 68504, United States
| | - Don T Lamb
- LI-COR Biosciences , 4647 Superior Street, Lincoln, Nebraska 68504, United States
| | - Stephen A Ferguson
- Department of Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Natalie E Arvin
- Department of Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Mohamed Dawod
- Department of Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
14
|
Liu CY, Lu DC, Jiang YW, Yen YK, Chang SC, Wang AB. Easy and Fast Western Blotting by Thin-Film Direct Coating with Suction. Anal Chem 2016; 88:6349-56. [PMID: 27254752 DOI: 10.1021/acs.analchem.6b00699] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Thin-film direct coating (TDC) has been successfully used in Western blotting (WB). In this study, the advanced technique of TDC with suction (TDCS) was developed to reduce the consumption amount of antibody by a factor of up to 10(4) in comparison with the amount consumed by the conventional WB using the capillary tube without any need of special micromachining processes. The operation time for completely finishing a high-quality WB can be reduced from 3 h in conventional WB to about 5 min or even less by TDCS. In addition, the signal-to-noise ratio of the immunoblotting by TDCS can be markedly increased. TDCS WB showed a high linearity within a 6-log2 dynamic range for detecting 90-6000 ng of purified recombinant glutathione-S-transferase (GST) proteins and could particularly detect extrinsic GST proteins added in crude Escherichia coli or 293T cell lysates. Moreover, a protein mixture containing bovine serum albumin, GST, and ubiquitin could be specifically probed in parallel with their corresponding antibodies through multichannel TDCS WB. This simple and innovative TDCS WB offers various potential applications in simultaneously finishing multiple antibody-antigen screenings in a fast and single experiment.
Collapse
Affiliation(s)
- Chao-Yuan Liu
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| | - De-Chao Lu
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| | - Yi-Wei Jiang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| | - Yi-Kuang Yen
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| | - Shih-Chung Chang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| | - An-Bang Wang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University , Taipei 106, Taiwan
| |
Collapse
|
15
|
|
16
|
Shameli SM, Ren CL. Microfluidic two-dimensional separation of proteins combining temperature gradient focusing and sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Anal Chem 2015; 87:3593-7. [PMID: 25787346 DOI: 10.1021/acs.analchem.5b00380] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A two-dimensional separation system is presented combining scanning temperature gradient focusing (TGF) and sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) in a PDMS/glass microfluidic chip. Denatured proteins are first focused and separated in a 15 mm long channel via TGF with a temperature range of 16-47 °C and a pressure scanning rate of -0.5 Pa/s and then further separated via SDS-PAGE in a 25 mm long channel. A side channel is designed at the intersection between the two dimensions to continuously inject SDS into the gel, allowing SDS molecules to be compiled within the focused bands. Separation experiments are performed using several fluorescently labeled proteins with single point detection. Experimental results show a dramatic improvement in peak capacity over one-dimensional separation techniques.
Collapse
Affiliation(s)
- Seyed Mostafa Shameli
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Ave. West, Waterloo, Ontario Canada, N2L 3G1
| | - Carolyn L Ren
- Department of Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Ave. West, Waterloo, Ontario Canada, N2L 3G1
| |
Collapse
|
17
|
Yang AQ, Wang D, Wang X, Han Y, Ke XB, Wang HJ, Zhou X, Ren L. Rational design of Au nanorods assemblies for highly sensitive and selective SERS detection of prostate specific antigen. RSC Adv 2015. [DOI: 10.1039/c5ra01322d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A simple SERS immunosensor based on AuNRs assembly was developed for rapid detection of specific antigen in early diagnostics.
Collapse
Affiliation(s)
- An-qi Yang
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| | - Dong Wang
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| | - Xiang Wang
- State Key Laboratory for Physical Chemistry of Solid Surfaces
- Department of Chemistry
- College of Chemistry and Chemical Engineering
- Xiamen University
- Xiamen
| | - Yu Han
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| | - Xue-bin Ke
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| | - Hong-jun Wang
- Department of Chemistry
- Chemical Biology and Biomedical Engineering
- Stevens Institute of Technology
- Hoboken
- USA
| | - Xi Zhou
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| | - Lei Ren
- Department of Biomaterials
- College of Materials
- Xiamen University
- Xiamen
- People's Republic of China
| |
Collapse
|
18
|
Gerver R, Herr AE. Microfluidic Western blotting of low-molecular-mass proteins. Anal Chem 2014; 86:10625-32. [PMID: 25268977 PMCID: PMC4222625 DOI: 10.1021/ac5024588] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/30/2014] [Indexed: 12/01/2022]
Abstract
We describe a microfluidic Western blot assay (μWestern) using a Tris tricine discontinuous buffer system suitable for analyses of a wide molecular mass range (6.5-116 kDa). The Tris tricine μWestern is completed in an enclosed, straight glass microfluidic channel housing a photopatterned polyacrylamide gel that incorporates a photoactive benzophenone methacrylamide monomer. Upon brief ultraviolet (UV) light exposure, the hydrogel toggles from molecular sieving for size-based separation to a covalent immobilization scaffold for in situ antibody probing. Electrophoresis controls all assay stages, affording purely electronic operation with no pumps or valves needed for fluid control. Electrophoretic introduction of antibody into and along the molecular sieving gel requires that the probe must traverse through (i) a discontinuous gel interface central to the transient isotachophoresis used to achieve high-performance separations and (ii) the full axial length of the separation gel. In-channel antibody probing of small molecular mass species is especially challenging, since the gel must effectively sieve small proteins while permitting effective probing with large-molecular-mass antibodies. To create a well-controlled gel interface, we introduce a fabrication method that relies on a hydrostatic pressure mismatch between the buffer and polymer precursor solution to eliminate the interfacial pore-size control issues that arise when a polymerizing polymer abuts a nonpolymerizing polymer solution. Combined with a new swept antibody probe plug delivery scheme, the Tris tricine μWestern blot enables 40% higher separation resolution as compared to a Tris glycine system, destacking of proteins down to 6.5 kDa, and a 100-fold better signal-to-noise ratio (SNR) for small pore gels, expanding the range of applicable biological targets.
Collapse
Affiliation(s)
- Rachel
E. Gerver
- University of California Berkeley
and University of California San
Francisco Graduate Program in Bioengineering, and Department of Bioengineering, University of California Berkeley, Berkeley, California 94720, United States
| | - Amy E. Herr
- University of California Berkeley
and University of California San
Francisco Graduate Program in Bioengineering, and Department of Bioengineering, University of California Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
19
|
Vashist SK, Lam E, Hrapovic S, Male KB, Luong JHT. Immobilization of Antibodies and Enzymes on 3-Aminopropyltriethoxysilane-Functionalized Bioanalytical Platforms for Biosensors and Diagnostics. Chem Rev 2014; 114:11083-130. [DOI: 10.1021/cr5000943] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sandeep Kumar Vashist
- HSG-IMIT - Institut für Mikro- und Informationstechnik, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
- Laboratory for MEMS Applications, Department of Microsystems Engineering - IMTEK, University of Freiburg, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
| | - Edmond Lam
- National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | | | - Keith B. Male
- National Research Council Canada, Montreal, Quebec H4P 2R2, Canada
| | - John H. T. Luong
- Innovative Chromatography Group, Irish Separation Science Cluster (ISSC), Department of Chemistry and Analytical, Biological Chemistry Research Facility (ABCRF), University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Chung M, Kim D, Herr AE. Polymer sieving matrices in microanalytical electrophoresis. Analyst 2014; 139:5635-54. [DOI: 10.1039/c4an01179a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
Eker B, Temiz Y, Delamarche E. Heterogeneous integration of gels into microfluidics using a mesh carrier. Biomed Microdevices 2014; 16:829-35. [DOI: 10.1007/s10544-014-9886-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Sung JH, Srinivasan B, Esch MB, McLamb WT, Bernabini C, Shuler ML, Hickman JJ. Using physiologically-based pharmacokinetic-guided "body-on-a-chip" systems to predict mammalian response to drug and chemical exposure. Exp Biol Med (Maywood) 2014; 239:1225-39. [PMID: 24951471 DOI: 10.1177/1535370214529397] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The continued development of in vitro systems that accurately emulate human response to drugs or chemical agents will impact drug development, our understanding of chemical toxicity, and enhance our ability to respond to threats from chemical or biological agents. A promising technology is to build microscale replicas of humans that capture essential elements of physiology, pharmacology, and/or toxicology (microphysiological systems). Here, we review progress on systems for microscale models of mammalian systems that include two or more integrated cellular components. These systems are described as a "body-on-a-chip", and utilize the concept of physiologically-based pharmacokinetic (PBPK) modeling in the design. These microscale systems can also be used as model systems to predict whole-body responses to drugs as well as study the mechanism of action of drugs using PBPK analysis. In this review, we provide examples of various approaches to construct such systems with a focus on their physiological usefulness and various approaches to measure responses (e.g. chemical, electrical, or mechanical force and cellular viability and morphology). While the goal is to predict human response, other mammalian cell types can be utilized with the same principle to predict animal response. These systems will be evaluated on their potential to be physiologically accurate, to provide effective and efficient platform for analytics with accessibility to a wide range of users, for ease of incorporation of analytics, functional for weeks to months, and the ability to replicate previously observed human responses.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Chemical Engineering, Hongik University, Seoul 121-791, Republic of Korea
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Mandy Brigitte Esch
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - William T McLamb
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Catia Bernabini
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
23
|
Yen YK, Jiang YW, Chang SC, Wang AB. Western Blotting by Thin-Film Direct Coating. Anal Chem 2014; 86:5164-70. [DOI: 10.1021/ac5010162] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yi-Kuang Yen
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University, Taipei 106, Taiwan
| | - Yi-Wei Jiang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University, Taipei 106, Taiwan
| | - Shih-Chung Chang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University, Taipei 106, Taiwan
| | - An-Bang Wang
- Institute of Applied Mechanics and ‡Department of Biochemical Science & Technology, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
24
|
Tentori AM, Herr AE. Performance implications of chemical mobilization after microchannel IEF. Electrophoresis 2014; 35:1453-60. [DOI: 10.1002/elps.201400023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/18/2014] [Accepted: 02/19/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Augusto M. Tentori
- The UC Berkeley/UCSF Graduate Program in Bioengineering; Berkeley CA USA
| | - Amy E. Herr
- The UC Berkeley/UCSF Graduate Program in Bioengineering; Berkeley CA USA
- Department of Bioengineering; UC Berkeley; Berkeley CA USA
| |
Collapse
|
25
|
Baratchi S, Khoshmanesh K, Sacristán C, Depoil D, Wlodkowic D, McIntyre P, Mitchell A. Immunology on chip: Promises and opportunities. Biotechnol Adv 2014; 32:333-46. [DOI: 10.1016/j.biotechadv.2013.11.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 11/04/2013] [Accepted: 11/17/2013] [Indexed: 01/09/2023]
|
26
|
Chang HN, Leroueil PR, Selwa K, Gasper CJ, Tsuchida RE, Wang JJ, McHugh WM, Cornell TT, Baker JR, Goonewardena SN. Profiling inflammatory responses with microfluidic immunoblotting. PLoS One 2013; 8:e81889. [PMID: 24312374 PMCID: PMC3842271 DOI: 10.1371/journal.pone.0081889] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 10/17/2013] [Indexed: 11/18/2022] Open
Abstract
Rapid profiling of signaling pathways has been a long sought after goal in biological sciences and clinical medicine. To understand these signaling pathways, their protein components must be profiled. The protein components of signaling pathways are typically profiled with protein immunoblotting. Protein immunoblotting is a powerful technique but has several limitations including the large sample requirements, high amounts of antibody, and limitations in assay throughput. To overcome some of these limitations, we have designed a microfluidic protein immunoblotting device to profile multiple signaling pathways simultaneously. We show the utility of this approach by profiling inflammatory signaling pathways (NFκB, JAK-STAT, and MAPK) in cell models and human samples. The microfluidic immunoblotting device can profile proteins and protein modifications with 5380-fold less antibody compared to traditional protein immunoblotting. Additionally, this microfluidic device interfaces with commonly available immunoblotting equipment, has the ability to multiplex, and is compatible with several protein detection methodologies. We anticipate that this microfluidic device will complement existing techniques and is well suited for life science applications.
Collapse
Affiliation(s)
- Huai-Ning Chang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pascale R. Leroueil
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Katherine Selwa
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - C. J. Gasper
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ryan E. Tsuchida
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jason J. Wang
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Walker M. McHugh
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Timothy T. Cornell
- Division of Pediatric Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sascha N. Goonewardena
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
27
|
Wang T, Zhang M, Dreher DD, Zeng Y. Ultrasensitive microfluidic solid-phase ELISA using an actuatable microwell-patterned PDMS chip. LAB ON A CHIP 2013; 13:4190-7. [PMID: 23989677 DOI: 10.1039/c3lc50783a] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Quantitative detection of low abundance proteins is of significant interest for biological and clinical applications. Here we report an integrated microfluidic solid-phase ELISA platform for rapid and ultrasensitive detection of proteins with a wide dynamic range. Compared to the existing microfluidic devices that perform affinity capture and enzyme-based optical detection in a constant channel volume, the key novelty of our design is two-fold. First, our system integrates a microwell-patterned assay chamber that can be pneumatically actuated to significantly reduce the volume of chemifluorescent reaction, markedly improving the sensitivity and speed of ELISA. Second, monolithic integration of on-chip pumps and the actuatable assay chamber allow programmable fluid delivery and effective mixing for rapid and sensitive immunoassays. Ultrasensitive microfluidic ELISA was demonstrated for insulin-like growth factor 1 receptor (IGF-1R) across at least five orders of magnitude with an extremely low detection limit of 21.8 aM. The microwell-based solid-phase ELISA strategy provides an expandable platform for developing the next-generation microfluidic immunoassay systems that integrate and automate digital and analog measurements to further improve the sensitivity, dynamic ranges, and reproducibility of proteomic analysis.
Collapse
Affiliation(s)
- Tanyu Wang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, United States.
| | | | | | | |
Collapse
|
28
|
Araz MK, Apori AA, Salisbury CM, Herr AE. Microfluidic barcode assay for antibody-based confirmatory diagnostics. LAB ON A CHIP 2013; 13:3910-3920. [PMID: 23925585 DOI: 10.1039/c3lc50229e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Confirmatory diagnostics offer high clinical sensitivity and specificity typically by assaying multiple disease biomarkers. Employed in clinical laboratory settings, such assays confirm a positive screening diagnostic result. These important multiplexed confirmatory assays require hours to complete. To address this performance gap, we introduce a simple 'single inlet, single outlet' microchannel architecture with multiplexed analyte detection capability. A streptavidin-functionalized, channel-filling polyacrylamide gel in a straight glass microchannel operates as a 3D scaffold for a purely electrophoretic yet heterogeneous immunoassay. Biotin and biotinylated capture reagents are patterned in discrete regions along the axis of the microchannel resulting in a barcode-like pattern of reagents and spacers. To characterize barcode fabrication, an empirical study of patterning behaviour was conducted across a range of electromigration and binding reaction timescales. We apply the heterogeneous barcode immunoassay to detection of human antibodies against hepatitis C virus and human immunodeficiency virus antigens. Serum was electrophoresed through the barcode patterned gel, allowing capture of antibody targets. We assess assay performance across a range of Damkohler numbers. Compared to clinical immunoblots that require 4-10 h long sample incubation steps with concomitant 8-20 h total assay durations; directed electromigration and reaction in the microfluidic barcode assay leads to a 10 min sample incubation step and a 30 min total assay duration. Further, the barcode assay reports clinically relevant sensitivity (25 ng ml(-1) in 2% human sera) comparable to standard HCV confirmatory diagnostics. Given the low voltage, low power and automated operation, we see the streamlined microfluidic barcode assay as a step towards rapid confirmatory diagnostics for a low-resource clinical laboratory setting.
Collapse
Affiliation(s)
- M Kursad Araz
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | |
Collapse
|
29
|
Signal amplification strategy for sensitive immunoassay of prostate specific antigen (PSA) based on ferrocene incorporated polystyrene spheres. Anal Chim Acta 2013; 793:19-25. [DOI: 10.1016/j.aca.2013.07.054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 07/19/2013] [Accepted: 07/25/2013] [Indexed: 12/13/2022]
|
30
|
Kim D, Herr AE. Protein immobilization techniques for microfluidic assays. BIOMICROFLUIDICS 2013; 7:41501. [PMID: 24003344 PMCID: PMC3747845 DOI: 10.1063/1.4816934] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 07/16/2013] [Indexed: 05/07/2023]
Abstract
Microfluidic systems have shown unequivocal performance improvements over conventional bench-top assays across a range of performance metrics. For example, specific advances have been made in reagent consumption, throughput, integration of multiple assay steps, assay automation, and multiplexing capability. For heterogeneous systems, controlled immobilization of reactants is essential for reliable, sensitive detection of analytes. In most cases, protein immobilization densities are maximized, while native activity and conformation are maintained. Immobilization methods and chemistries vary significantly depending on immobilization surface, protein properties, and specific assay goals. In this review, we present trade-offs considerations for common immobilization surface materials. We overview immobilization methods and chemistries, and discuss studies exemplar of key approaches-here with a specific emphasis on immunoassays and enzymatic reactors. Recent "smart immobilization" methods including the use of light, electrochemical, thermal, and chemical stimuli to attach and detach proteins on demand with precise spatial control are highlighted. Spatially encoded protein immobilization using DNA hybridization for multiplexed assays and reversible protein immobilization surfaces for repeatable assay are introduced as immobilization methods. We also describe multifunctional surface coatings that can perform tasks that were, until recently, relegated to multiple functional coatings. We consider the microfluidics literature from 1997 to present and close with a perspective on future approaches to protein immobilization.
Collapse
Affiliation(s)
- Dohyun Kim
- Department of Mechanical Engineering, Myongji University, 116 Myongji-ro, Cheoin-gu, Yongin-si, Gyeonggi-do 449-728, South Korea
| | | |
Collapse
|
31
|
Duncombe TA, Herr AE. Photopatterned free-standing polyacrylamide gels for microfluidic protein electrophoresis. LAB ON A CHIP 2013; 13:2115-2123. [PMID: 23609800 DOI: 10.1039/c3lc50269d] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Designed for compatibility with slab-gel polyacrylamide gel electrophoresis (PAGE) reagents and instruments, we detail development of free-standing polyacrylamide gel (fsPAG) microstructures supporting electrophoretic performance rivalling that of microfluidic platforms. For the protein electrophoresis study described here, fsPAGE lanes are comprised of a sample reservoir and contiguous separation gel. No enclosed microfluidic channels are employed. The fsPAG devices (120 μm tall) are directly photopatterned atop of and covalently attached to planar polymer or glass surfaces. Leveraging the fast <1 h design-prototype-test cycle - significantly faster than mold based fabrication techniques - we optimize the fsPAG architecture to minimize injection dispersion for rapid (<1 min) and short (1 mm) protein separations. The facile fabrication and prototyping of the fsPAGE provides researchers a powerful tool for developing custom analytical assays. We highlight the utility of assay customization by fabricating a polyacrylamide gel with a spatial pore-size distribution and demonstrate the resulting enhancement in separation performance over a uniform gel. Further, we up-scale from a unit separation to an array of 96 concurrent fsPAGE assays in 10 min run time driven by one electrode pair. The fsPAG array layout matches that of a 96-well plate to facilitate integration of the planar free standing gel array with multi-channel pipettes while remaining compatible with conventional slab-gel PAGE reagents, such as staining for label-free protein detection. Notably, the entire fsPAGE workflow from fabrication, to operation, and readout uses readily available materials and instruments - making this technique highly accessible.
Collapse
Affiliation(s)
- Todd A Duncombe
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, 342 Stanley Hall, Berkeley, California 94720, USA
| | | |
Collapse
|
32
|
Jin S, Anderson GJ, Kennedy RT. Western blotting using microchip electrophoresis interfaced to a protein capture membrane. Anal Chem 2013; 85:6073-9. [PMID: 23672369 DOI: 10.1021/ac400940x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Western blotting is a commonly used assay for proteins. Despite the utility of the method, it is also characterized by long analysis times, manual operation, and lack of established miniaturized counterpart. We report a new way to Western blot that addresses these limitations. In the method, sodium dodecyl sulfate (SDS)-protein complexes are separated by sieving electrophoresis in a microfluidic device or chip. The chip is interfaced to a moving membrane so that proteins are captured in discrete zones as they migrate from the chip. Separations of SDS-protein complexes in the molecular weight range of 11-155 kDa were completed in 2 min with 4 × 10(4) theoretical plates at 460 V/cm. Migration time and peak area relative standard deviations were 3-6% and 0.2%, respectively. Detection limit for actin was 0.7 nM. Assays for actin, AMP-kinase, carbonic anhydrase, and lysozyme are shown to demonstrate versatility of the method. Total analysis time including immunoassay was 22-32 min for a single sample. Because processing membrane for immunoassay is the slow step of the assay, sequential injections from different reservoirs on the chip and capture in different tracks on the same membrane allow increased throughput. As a demonstration, 9 injections were collected on one membrane and analyzed in 43 min (~5 min/sample). Further improvements in throughput are possible with more reservoirs or parallel channels.
Collapse
Affiliation(s)
- Shi Jin
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | |
Collapse
|
33
|
Tentori AM, Hughes AJ, Herr AE. Microchamber integration unifies distinct separation modes for two-dimensional electrophoresis. Anal Chem 2013; 85:4538-45. [PMID: 23565932 PMCID: PMC3714212 DOI: 10.1021/ac4001767] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
By combining isoelectric focusing (IEF) with subsequent gel electrophoresis, two-dimensional electrophoresis (2DE) affords more specific characterization of proteins than each constituent unit separation. In a new approach to integrating the two assay dimensions in a microscope slide-sized glass device, we introduce microfluidic 2DE using photopatterned polyacrylamide (PA) gel elements housed in a millimeter-scale, 20-μm-deep chamber. The microchamber minimizes information loss inherent to channel network architectures commonly used for microfluidic 2DE. To define the IEF axis along a "lane" at the top of the chamber, we used free solution carrier ampholytes and immobilized acrylamido buffers in the PA gels. This approach yielded high-resolution (0.1 pH unit) and rapid (<20 min) IEF. Next, protein transfer to the second dimension was accomplished by chemical mobilization perpendicular to the IEF axis. Mobilization drove focused proteins off the IEF lane and into a region for protein gel electrophoresis. Using fluorescently labeled proteins, we observed transfer-induced band broadening factors ~7.5-fold lower than those observed in microchannel networks. Both native polyacrylamide gel electrophoresis (PAGE) and pore-limit electrophoresis (PLE) were studied as the second assay dimension and completed in <15 min. PLE yields protein molecular mass information without the need for ionic surfactant or reducing agents, simplifying device design and operation. Microchamber-based 2DE unifies two independent separation dimensions in a single device with minimal transfer-associated information losses. Peak capacities for the total assay ranged from 256 to 35 with <1 h assay duration. The rapid microchamber 2DE assay has the potential to bridge an existing gap in targeted proteomics for protein biomarker validation and systems biology that may complement recent innovation in mass spectrometry.
Collapse
Affiliation(s)
- Augusto M. Tentori
- The UC Berkeley/UCSF Graduate Program in Bioengineering, Berkeley, CA, USA
| | - Alex J. Hughes
- The UC Berkeley/UCSF Graduate Program in Bioengineering, Berkeley, CA, USA
| | - Amy E. Herr
- The UC Berkeley/UCSF Graduate Program in Bioengineering, Berkeley, CA, USA
| |
Collapse
|
34
|
Na N, Liu T, Yang X, Sun B, Ouyang J, Ouyang J. A simple cellulose acetate membrane-based small lanes technique for protein electrophoresis. Anal Bioanal Chem 2012; 404:753-62. [DOI: 10.1007/s00216-012-6168-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 05/30/2012] [Accepted: 05/30/2012] [Indexed: 11/30/2022]
|
35
|
Azadi G, Gustafson E, Wessel GM, Tripathi A. Rapid detection and quantification of specific proteins by immunodepletion and microfluidic separation. Biotechnol J 2012; 7:1008-13. [PMID: 22539461 DOI: 10.1002/biot.201100378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 03/18/2012] [Accepted: 04/20/2012] [Indexed: 11/11/2022]
Abstract
Conventional immunoblotting techniques are labor intensive, time consuming and rely on the elution of target protein after depletion. Here we describe a new method for detection and quantification of proteins, independent of washing and elution. In this method, the target protein is first captured by immunodepletion with antibody-coated microbeads. In the second step, both the supernatant after immunodepletion and the untreated protein sample are directly analyzed by microfluidic electrophoresis without further processing. Subsequently, the detection and quantification are performed by comparing the electropherograms of these two samples. This method was tested using an Escherichia coli lysate with a FLAG-tagged protein and anti-FLAG magnetic beads. An incubation of as short as one min was sufficient for detectable depletion (66%) by microchip electrophoresis. Longer incubation (up to 60 min) resulted in more depletion of the target band (82%). Our results show that only 19% of the target is recovered after elution from the beads. By eliminating multiple wash and elution steps, our method is faster, less labor intensive, and highly reproducible. The target protein can still be easily identified even in the case of nonspecific binding at low concentrations. This work highlights the advantages of integrating immunodepletion techniques on a microfluidic platform.
Collapse
Affiliation(s)
- Glareh Azadi
- Center for Biomedical Engineering, School of Engineering and Division of Biology & Medicine, Brown University, Providence, RI 02912, USA
| | | | | | | |
Collapse
|
36
|
Zhang J, Sajid M, Na N, Huang L, He D, Ouyang J. The application of Au nanoclusters in the fluorescence imaging of human serum proteins after native PAGE: Enhancing detection by low-temperature plasma treatment. Biosens Bioelectron 2012; 35:313-318. [DOI: 10.1016/j.bios.2012.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/06/2012] [Indexed: 11/28/2022]
|
37
|
van der Meer AD, van den Berg A. Organs-on-chips: breaking the in vitro impasse. Integr Biol (Camb) 2012; 4:461-70. [PMID: 22388577 DOI: 10.1039/c2ib00176d] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In vitro models of biological tissues are indispensable tools for unraveling human physiology and pathogenesis. They usually consist of a single layer of a single cell type, which makes them robust and suitable for parallelized research. However, due to their simplicity, in vitro models are also less valid as true reflections of the complex biological tissues of the human body. Even though the realism of the models can be increased by including more cell types, this will inevitably lead to a decrease in robustness and throughput. The constant trade-off between realism and simplicity has led to an impasse in the development of new in vitro models. Organs-on-chips, a class of microengineered in vitro tissue models, have the potential to break the in vitro impasse. These models combine an artificially engineered, physiologically realistic cell culture microenvironment with the potential for parallelization and increased throughput. They are robust, because the engineered physiological, organ-level features such as tissue organization, geometry, soluble gradients and mechanical stimulation are well-defined and controlled. Moreover, their microfluidic properties and integrated sensors pave the way for high-throughput studies. In this review, we define the in vitro impasse, we explain why organs-on-chips have the potential to break the impasse and we formulate a view on the future of the field. We focus on the design philosophy of organs-on-chips, the integration of technology and biology and on how to connect to the potential end-users.
Collapse
Affiliation(s)
- Andries D van der Meer
- BIOS/Lab on a Chip group, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands
| | | |
Collapse
|
38
|
Cho SW, Kang DK, Choo JB, Demllo AJ, Chang SI. Recent advances in microfluidic technologies for biochemistry and molecular biology. BMB Rep 2011; 44:705-12. [DOI: 10.5483/bmbrep.2011.44.11.705] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
39
|
Appleyard DC, Chapin SC, Srinivas RL, Doyle PS. Bar-coded hydrogel microparticles for protein detection: synthesis, assay and scanning. Nat Protoc 2011; 6:1761-74. [DOI: 10.1038/nprot.2011.400] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
40
|
Karns K, Herr AE. Human tear protein analysis enabled by an alkaline microfluidic homogeneous immunoassay. Anal Chem 2011; 83:8115-22. [PMID: 21910436 DOI: 10.1021/ac202061v] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The ability to probe the protein content of human tear fluid has enormous potential for deepening our understanding of ocular and systemic disease pathology and enabling novel noninvasive tear-based diagnostic technologies. To overcome current challenges in tear proteomic measurements, we report on the first microfluidic homogeneous immunoassay capable of making rapid, quantitative, and specific measurements of endogenous tear protein biomarkers in human tear fluid. Lactoferrin (Lf) is a tear-specific biomarker for Sjögren's syndrome (SS), a serious systemic autoimmune disease currently diagnosed through rudimentary volumetric and surface chemistry measurements and an invasive lip biopsy. We detail optimization of a homogeneous electrophoretic immunoassay for Lf in <1 μL of tear fluid at clinically relevant concentrations. In particular, we present assay development details and a final assay that enables quantification of Lf in <5 s in a clinically relevant range for SS diagnostics. Characterization suggests the on-chip assay is accurate to within 15% of ELISA, specific (<15% nonspecific signal), and with a lower limit of detection of 3 ± 2 nM Lf in human tear matrix. Additionally, we develop and characterize a protocol for eluting proteins from nitrocellulose Schirmer strips, the clinical de facto standard for tear collection and storage. We relate on-chip measured Lf concentrations back to ocular surface concentrations for the first time to our knowledge. Taken in sum, this work details important steps toward (1) expanding the set of proteins quantified by electrophoretic immunoassays to encompass a wider range of isoelectric points than has been reported, (2) creating a first-in-kind translatable assay with clinical relevance to SS diagnostics, and (3) expanding the analytical toolkit available for rapid tear protein measurements, as is relevant to the advancement of basic research and clinical medicine.
Collapse
Affiliation(s)
- Kelly Karns
- Graduate Program in Bioengineering, The University of California, Berkeley-University of California, San Francisco, Berkeley, California 94720, United States
| | | |
Collapse
|
41
|
Tentori AM, Herr AE. Photopatterned materials in bioanalytical microfluidic technology. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2011; 21:54001. [PMID: 21857772 PMCID: PMC3156436 DOI: 10.1088/0960-1317/21/5/054001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Microfluidic technologies are playing an increasingly important role in biological inquiry. Sophisticated approaches to the microanalysis of biological specimens rely, in part, on the fine fluid and material control offered by microtechnology, as well as a sufficient capacity for systems integration. A suite of techniques that utilize photopatterning of polymers on fluidic surfaces, within fluidic volumes, and as primary device structures underpins recent technological innovation in bioanalysis. Well-characterized photopatterning approaches enable previously fabricated or commercially fabricated devices to be customized by the user in a straight-forward manner, making the tools accessible to laboratories that do not focus on microfabrication technology innovation. In this review of recent advances, we summarize reported microfluidic devices with photopatterned structures and regions as platforms for a diverse set of biological measurements and assays.
Collapse
|
42
|
Abstract
We introduce and characterize multiplexed native Western blotting in an automated and unified microfluidic format. While slab gel Western blotting is slow and laborious, conventional multiplexed blotting ("reblotting": probing one sample with multiple antibodies) requires even more resources. Here we detail three key advances that enable an automated and rapid microfluidic alternative to slab gel reblotting. First, we introduce both assay and microdevice designs that integrate protein blotting against multiple antibody blotting regions with native polyacrylamide gel electrophoresis. This microfluidic integration strategy overcomes nonspecific material losses inherent to harsh antibody stripping steps typically needed for conventional reblotting; said conditions can severely limit analyte quantitation. Second, to inform rational design of the multiplexed microfluidic device we develop an analytical model for analyte capture on the blotting regions. Comparison to empirical observations is reported, with capture efficiencies of >85%. Third, we introduce label free detection that makes simultaneous and quantitative multiplexed measurements possible without the need for prelabeling of sample. Assay linear dynamic range spans 8-800 nM with assay completion in 5 min. Owing to the speed, automation, enhanced quantitation capability, and the difficulty of conventional slab gel Western reblotting, microfluidic multiplexed native Western blotting should find use in systems biology, in particular in analyses of protein isoforms and multimeric protein complexes.
Collapse
Affiliation(s)
- Samuel Q Tia
- Department of Bioengineering, University of California, Berkeley, 94720, United States
| | | | | | | |
Collapse
|
43
|
Anderson GJ, M Cipolla C, Kennedy RT. Western blotting using capillary electrophoresis. Anal Chem 2011; 83:1350-5. [PMID: 21265514 DOI: 10.1021/ac102671n] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A microscale Western blotting system based on separating sodium-dodecyl sulfate protein complexes by capillary gel electrophoresis followed by deposition onto a blotting membrane for immunoassay is described. In the system, the separation capillary is grounded through a sheath capillary to a mobile X-Y translation stage which moves a blotting membrane past the capillary outlet for protein deposition. The blotting membrane is moistened with a methanol and buffer mixture to facilitate protein adsorption. Although discrete protein zones could be detected, bands were broadened by ∼1.7-fold by transfer to membrane. A complete Western blot for lysozyme was completed in about one hour with 50 pg mass detection limit from low microgram per milliliter samples. These results demonstrate substantial reduction in time requirements and improvement in mass sensitivity compared to conventional Western blots. Western blotting using capillary electrophoresis shows promise to analyze low volume samples with reduced reagents and time, while retaining the information content of a typical Western blot.
Collapse
|
44
|
Abstract
This protocol describes regional photopatterning of polyacrylamide gels in glass microfluidic devices as a platform for seamless integration of multiple assay steps. The technology enables rapid, automated protein immunoblotting, demonstrated in this study for native western blotting. The fabrication procedure is straightforward and requires approximately 3 h from the start of gel photopatterning to completion of native protein western blotting, a substantial time savings over slab-gel immunoblotting. The assay itself requires less than 5 min. Importantly, all assay stages are programmably controlled by a high-voltage power supply and monitored by an epifluorescence microscope equipped with a charge-coupled device camera. Our approach overcomes severe limitations associated with conventional immunoblotting, including multiple steps requiring manual intervention, low throughput and substantial consumption of reagents. We also describe a simple chemical recycling protocol so that glass chips can be reused. The fabrication technique described forms the basis for a diverse suite of bioanalytical tools, including DNA/RNA blotting and multidimensional separations.
Collapse
Affiliation(s)
- Mei He
- Department of Bioengineering, University of California, Berkeley, California, USA
| | | |
Collapse
|
45
|
Abstract
We develop a novel method for Western blot based on microfluidics, incorporating the internal molecular weight marker, loading control, and antibody titration in the same protocol. Compared with the conventional method which could detect only one protein, the microfluidic Western blot could analyze at least 10 proteins simultaneously from a single sample, and it requires only about 1% of the amount of antibody used in conventional Western blot.
Collapse
|
46
|
Oita I, Halewyck H, Thys B, Rombaut B, Vander Heyden Y, Mangelings D. Microfluidics in macro-biomolecules analysis: macro inside in a nano world. Anal Bioanal Chem 2010; 398:239-64. [PMID: 20549494 PMCID: PMC7079953 DOI: 10.1007/s00216-010-3857-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2010] [Revised: 05/13/2010] [Accepted: 05/18/2010] [Indexed: 12/26/2022]
Abstract
Use of microfluidic devices in the life sciences and medicine has created the possibility of performing investigations at the molecular level. Moreover, microfluidic devices are also part of the technological framework that has enabled a new type of scientific information to be revealed, i.e. that based on intensive screening of complete sets of gene and protein sequences. A deeper bioanalytical perspective may provide quantitative and qualitative tools, enabling study of various diseases and, eventually, may offer support for the development of accurate and reliable methods for clinical assessment. This would open the way to molecule-based diagnostics, i.e. establish accurate diagnosis and disease prognosis based on identification and/or quantification of biomacromolecules, for example proteins or nucleic acids. Finally, the development of disposable and portable devices for molecule-based diagnosis would provide the perfect translation of the science behind life-science research into practical applications dedicated to patients and health practitioners. This review provides an analytical perspective of the impact of microfluidics on the detection and characterization of bio-macromolecules involved in pathological processes. The main features of molecule-based diagnostics and the specific requirements for the diagnostic devices are discussed. Further, the techniques currently used for testing bio-macromolecules for potential diagnostic purposes are identified, emphasizing the newest developments. Subsequently, the challenges of this type of application and the status of commercially available devices are highlighted, and future trends are noted.
Collapse
Affiliation(s)
- Iuliana Oita
- Department of Analytical Chemistry and Pharmaceutical Technology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| | - Hadewych Halewyck
- Department of Pharmaceutical Biotechnology & Molecular Biology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| | - Bert Thys
- Department of Pharmaceutical Biotechnology & Molecular Biology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| | - Bart Rombaut
- Department of Pharmaceutical Biotechnology & Molecular Biology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| | - Yvan Vander Heyden
- Department of Analytical Chemistry and Pharmaceutical Technology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| | - Debby Mangelings
- Department of Analytical Chemistry and Pharmaceutical Technology, Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel-VUB, Laarbeeklaan 103, Brussels, 1090 Belgium
| |
Collapse
|
47
|
Hughes AJ, Herr AE. Quantitative enzyme activity determination with zeptomole sensitivity by microfluidic gradient-gel zymography. Anal Chem 2010; 82:3803-11. [PMID: 20353191 DOI: 10.1021/ac100201z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We describe a sensitive zymography technique that utilizes an automated microfluidic platform to report enzyme molecular weight, amount, and activity (including k(cat) and K(m)) from dilute protein mixtures. Calf intestinal alkaline phosphatase (CIP) is examined in detail as a model enzyme system, and the method is also demonstrated for horseradish peroxidase (HRP). The 40 min assay has a detection limit of 5 zmol ( approximately 3 000 molecules) of CIP. Two-step pore-limit electrophoresis with enzyme assay (PLENZ) is conducted in a single, straight microchannel housing a polyacrylamide (PA) pore-size gradient gel. In the first step, pore limit electrophoresis (PLE) sizes and pseudoimmobilizes resolved proteins. In the second step, electrophoresis transports both charged and neutral substrates into the PLE channel to the entrapped proteins. Arrival of substrate at the resolved enzyme band generates fluorescent product that reveals enzyme molecular weight against a fluorescent protein ladder. Additionally, the PLENZ zymography assay reports the kinetic properties of CIP in a fully quantitative manner. In contrast to covalent enzyme immobilization, physical pseudoimmobilization of CIP in the PA gel does not significantly reduce its maximum substrate turnover rate. However, an 11-fold increase in the Michaelis constant (over the free solution value) is observed, consistent with diffusional limitations on substrate access to the enzyme active site. PLENZ offers a robust platform for rapid and multiplexed functional analysis of heterogeneous protein samples in drug discovery, clinical diagnostics, and biocatalyst engineering.
Collapse
Affiliation(s)
- Alex J Hughes
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | | |
Collapse
|
48
|
He M, Herr AE. Polyacrylamide gel photopatterning enables automated protein immunoblotting in a two-dimensional microdevice. J Am Chem Soc 2010; 132:2512-3. [PMID: 20131779 DOI: 10.1021/ja910164d] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We demonstrate a two-dimensional microfluidic architecture that integrates polyacrylamide gel electrophoresis (PAGE) with immunoblotting in a fully automated format. This assay is designed to overcome performance limitations of conventional slab-gel immunoblotting, including multiple manual interventions, low-throughput transfer and blotting, and substantial consumption of reagents and sample. To unify PAGE with blotting in one device, this microfluidic approach makes use of high-resolution regional photopatterning of multiple polyacrylamide gel elements, and automated electrophoretic transport. A complete native immunoblot of free prostate specific antigen from human seminal fluid is demonstrated in less than 5 min. Further, the characterization of post-PAGE electrophoretic transfer showed high efficiency and minimal sample dispersion.
Collapse
Affiliation(s)
- Mei He
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | | |
Collapse
|