1
|
Dagher M, Ongo G, Robichaud N, Kong J, Rho W, Teahulos I, Tavakoli A, Bovaird S, Merjaneh S, Tan A, Edwardson K, Scheepers C, Ng A, Hajjar A, Sow B, Vrouvides M, Lee A, DeCorwin-Martin P, Rasool S, Huang J, Erps T, Coffin S, Han Y, Chandrasekaran SN, Miller L, Kost-Alimova M, Skepner A, Singh S, Carpenter AE, Munzar J, Juncker D. nELISA: A high-throughput, high-plex platform enables quantitative profiling of the inflammatory secretome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.04.17.535914. [PMID: 37131604 PMCID: PMC10153206 DOI: 10.1101/2023.04.17.535914] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We present the nELISA, a high-throughput, high-fidelity, and high-plex protein profiling platform. DNA oligonucleotides are used to pre-assemble antibody pairs on spectrally encoded microparticles and perform displacement-mediated detection. Spatial separation between non-cognate antibodies prevents the rise of reagent-driven cross-reactivity, while read-out is performed cost-efficiently and at high-throughput using flow cytometry. nELISA can measure both protein concentration and their post-translational modifications. We assembled an inflammatory panel of 191 targets that were multiplexed without cross-reactivity nor impact on performance vs 1-plex signals, with sensitivities as low as 0.1 pg/mL and measurements spanning 7 orders of magnitude. We then performed a large-scale inflammatory-secretome perturbation screen of peripheral blood mononuclear cells (PBMCs), with cytokines as both perturbagens and readouts, measuring 7,392 samples and generating ~1.4M protein data points in under a week; a significant advance in throughput compared to other highly multiplexed immunoassays. We uncovered 447 significant cytokine responses, including multiple putatively novel ones, that were conserved across donors and stimulation conditions. We validate nELISA for phenotypic screening, where its capacity to faithfully report hundreds of proteins make it a powerful tool across multiple stages of drug discovery.
Collapse
Affiliation(s)
- Milad Dagher
- Nomic Bio, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | | | | | - Jinglin Kong
- Nomic Bio, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | - Woojong Rho
- Nomic Bio, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | - Ivan Teahulos
- Nomic Bio, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | | | | | | | | | | | | | - Andy Ng
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | | - Yu Han
- Broad Institute of MIT and Harvard, Imaging Platform, Cambridge, MA, USA
| | | | - Lisa Miller
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA, USA
| | - Maria Kost-Alimova
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA, USA
| | - Shantanu Singh
- Broad Institute of MIT and Harvard, Imaging Platform, Cambridge, MA, USA
| | - Anne E. Carpenter
- Broad Institute of MIT and Harvard, Imaging Platform, Cambridge, MA, USA
| | - Jeffrey Munzar
- Nomic Bio, Montreal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | - David Juncker
- Victor Phillip Dahdaleh Institute of Genomic Medicine, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Zhang SJ, Wu C, Walt DR. A Multiplexed Digital Platform Enables Detection of Attomolar Protein Levels with Minimal Cross-Reactivity. ACS NANO 2024; 18:29891-29901. [PMID: 39422558 DOI: 10.1021/acsnano.4c10340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Protein-based biomarkers are essential for disease diagnostics, yet their low abundance in biofluids often presents significant detection challenges for traditional enzyme-linked immunosorbent assay (ELISA) techniques. While various ultrasensitive methods such as digital ELISA have improved sensitivity, multiplex assays still suffer from considerable cross-reactivities that can compromise result accuracies. To address this challenge, we have developed barcoded Molecular On-bead Signal Amplification for Individual Counting (barcoded MOSAIC), a multiplexed digital ELISA technology that markedly reduces cross-reactivity by pairing barcoded detection antibodies with specific bead types. This approach enables the simultaneous detection of eight analytes from less than 9 μL of blood, with sensitivities ranging from midpicomolar to low-attomolar levels and a collective dynamic range exceeding seven logs across multiple analytes within a single multiplex assay. Additionally, barcoded MOSAIC is compatible with standard immunoassay reagents and workflows, utilizing a rapid, automatable flow cytometric readout for quantification, which makes it a highly accessible benchtop platform that is readily adoptable by both research and clinical laboratories, setting the stage for future translation into point-of-care applications.
Collapse
Affiliation(s)
- Stephanie J Zhang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Connie Wu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| |
Collapse
|
3
|
Musicò A, Bergamaschi G, Strada A, Frigerio R, Gagni P, Cretich M, Gori A. Hybrid Peptide-Agarose Hydrogels for 3D Immunoassays. Methods Mol Biol 2023; 2578:53-62. [PMID: 36152280 DOI: 10.1007/978-1-0716-2732-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Recent advances in biosensing analytical platforms have brought relevant outcomes for novel diagnostic and therapy-oriented applications. In this context, 3D droplet microarrays, where hydrogels are used as matrices to stably entrap biomolecules onto analytical surfaces, potentially provide relevant advantages over conventional 2D assays, such as increased loading capacity, lower nonspecific binding, and enhanced signal-to-noise ratio. Here, we describe a hybrid hydrogel composed of a self-assembling peptide and commercial agarose (AG) as a suitable matrix for 3D microarray bioassays. The hybrid hydrogel is printable and self-adhesive and allows analyte diffusion. As a showcase example, we describe its application in a diagnostic immunoassay for the detection of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Angelo Musicò
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy.
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Greta Bergamaschi
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| | - Alessandro Strada
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| | - Roberto Frigerio
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| | - Paola Gagni
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| | - Marina Cretich
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| | - Alessandro Gori
- National Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche (SCITEC-CNR), Milan, Italy
| |
Collapse
|
4
|
Devadhasan JP, Summers AJ, Gu J, Smith S, Thomas B, Fattahi A, Helton J, Pandit SG, Gates-Hollingsworth M, Hau D, Pflughoeft KJ, Montgomery DC, Atta S, Vo-Dinh T, AuCoin D, Zenhausern F. Point-of-care vertical flow immunoassay system for ultra-sensitive multiplex biothreat-agent detection in biological fluids. Biosens Bioelectron 2023; 219:114796. [PMID: 36257115 DOI: 10.1016/j.bios.2022.114796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
This paper presents simple, fast, and sensitive detection of multiple biothreat agents by paper-based vertical flow colorimetric sandwich immunoassay for detection of Yersinia pestis (LcrV and F1) and Francisella tularensis (lipopolysaccharide; LPS) antigens using a vertical flow immunoassay (VFI) prototype with portable syringe pump and a new membrane holder. The capture antibody (cAb) printing onto nitrocellulose membrane and gold-labelled detection antibody (dAb) were optimized to enhance the assay sensitivity and specificity. Even though the paper pore size was relaxed from previous 0.1 μm to the current 0.45 μm for serum samples, detection limits as low as 0.050 ng/mL for LcrV and F1, and 0.100 ng/mL for FtLPS have been achieved in buffer and similarly in diluted serum (with LcrV and F1 LODs remained the same and LPS LOD reduced to 0.250 ng/mL). These were 40, 80, and 50X (20X for LPS in serum) better than those from lateral flow configuration. Furthermore, the comparison of multiplex format demonstrated low cross-reactivity and equal sensitivity to that of the singleplex assay. The optimized VFI platform thus provides a portable and rapid on-site monitoring system for multiplex biothreat detection with the potential for high sensitivity, specificity, reproducibility, and multiplexing capability, supporting its utility in remote and resource-limited settings.
Collapse
Affiliation(s)
- Jasmine Pramila Devadhasan
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Alexander Jarrett Summers
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Jian Gu
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA; Department of Basic Medical Sciences, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA.
| | - Stanley Smith
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Baiju Thomas
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Ali Fattahi
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - James Helton
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA
| | - Sujata G Pandit
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | | | - Derrick Hau
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Kathryn J Pflughoeft
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Douglas C Montgomery
- School of Computing and Augmented Intelligence, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe AZ, 85281, USA
| | - Supriya Atta
- Fitzpatrick Institute for Photonics, Departments of Biomedical Engineering and Chemistry, Duke University, Durham, NC, 27708-0281, USA
| | - Tuan Vo-Dinh
- Fitzpatrick Institute for Photonics, Departments of Biomedical Engineering and Chemistry, Duke University, Durham, NC, 27708-0281, USA
| | - David AuCoin
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA; Department of Basic Medical Sciences, The University of Arizona, College of Medicine-Phoenix, Phoenix, AZ, 85004, USA; Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson AZ, 85721-0020, USA.
| |
Collapse
|
5
|
Immune-Proteome Profiling in Classical Hodgkin Lymphoma Tumor Diagnostic Tissue. Cancers (Basel) 2021; 14:cancers14010009. [PMID: 35008176 PMCID: PMC8750205 DOI: 10.3390/cancers14010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 11/16/2022] Open
Abstract
In classical Hodgkin Lymphoma (cHL), immunoediting via protein signaling is key to evading tumor surveillance. We aimed to identify immune-related proteins that distinguish diagnostic cHL tissues (=diagnostic tumor lysates, n = 27) from control tissues (reactive lymph node lysates, n = 30). Further, we correlated our findings with the proteome plasma profile between cHL patients (n = 26) and healthy controls (n = 27). We used the proximity extension assay (PEA) with the OlinkTM multiplex Immuno-Oncology panel, consisting of 92 proteins. Univariate, multivariate-adjusted analysis and Benjamini–Hochberg’s false discovery testing (=Padj) were performed to detect significant discrepancies. Proteins distinguishing cHL cases from controls were more numerous in plasma (30 proteins) than tissue (17 proteins), all Padj < 0.05. Eight of the identified proteins in cHL tissue (PD-L1, IL-6, CCL17, CCL3, IL-13, MMP12, TNFRS4, and LAG3) were elevated in both cHL tissues and cHL plasma compared with control samples. Six proteins distinguishing cHL tissues from controls tissues were significantly correlated to PD-L1 expression in cHL tissue (IL-6, MCP-2, CCL3, CCL4, GZMB, and IFN-gamma, all p ≤0.05). In conclusion, this study introduces a distinguishing proteomic profile in cHL tissue and potential immune-related markers of pathophysiological relevance.
Collapse
|
6
|
Goecker ZC, Legg KM, Salemi MR, Herren AW, Phinney BS, McKiernan HE, Parker GJ. Alternative LC-MS/MS Platforms and Data Acquisition Strategies for Proteomic Genotyping of Human Hair Shafts. J Proteome Res 2021; 20:4655-4666. [PMID: 34491751 DOI: 10.1021/acs.jproteome.1c00209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein is a major component of all biological evidence. Proteomic genotyping is the use of genetically variant peptides (GVPs) that contain single-amino-acid polymorphisms to infer the genotype of matching nonsynonymous single-nucleotide polymorphisms for the individual from whom the protein sample originated. This can be used to statistically associate an individual to evidence found at a crime scene. The utility of the inferred genotype increases as the detection of GVPs increases, which is the direct result of technology transfer to mass spectrometry platforms typically available. Digests of single (2 cm) human hair shafts from three European and two African subjects were analyzed using data-dependent acquisition on a Q-Exactive Plus Hybrid Quadrupole-Orbitrap system, data-independent acquisition and a variant of parallel reaction monitoring (PRM) on an Orbitrap Fusion Lumos Tribrid system, and multiple reaction monitoring (MRM) on an Agilent 6495 triple quadrupole system. In our hands, average GVP detection from a selected panel of 24 GVPs increased from 6.5 ± 1.1 and 3.1 ± 0.8 using data-dependent and -independent acquisition to 9.5 ± 0.7 and 11.7 ± 1.7 using PRM and MRM (p < 0.05), respectively. PRM resulted in a 1.3-fold increase in detection sensitivity, and MRM resulted in a 1.6-fold increase in detection sensitivity. This increase in biomarker detection has a functional impact on the statistical association of a protein sample and an individual. Increased biomarker sensitivity, using Markov Chain Monte Carlo modeling, produced a median-estimated random match probability of over 1 in 10 trillion from a single hair using targeted proteomics. For PRM and MRM, detected GVPs were validated by the inclusion of stable isotope-labeled peptides in each sample, which served also as a detection trigger. This research accomplishes two aims: the demonstration of utility for alternative analytical platforms in proteomic genotyping and the establishment of validation methods for the evaluation of inferred genotypes.
Collapse
Affiliation(s)
- Zachary C Goecker
- Department of Environmental Toxicology, University of California, Davis, California 95616, United States
| | - Kevin M Legg
- The Center for Forensic Science Research and Education, Willow Grove, Pennsylvania 19090, United States
| | - Michelle R Salemi
- Proteomics Core Facility, University of California, Davis, California 95616, United States
| | - Anthony W Herren
- Proteomics Core Facility, University of California, Davis, California 95616, United States
| | - Brett S Phinney
- Proteomics Core Facility, University of California, Davis, California 95616, United States
| | - Heather E McKiernan
- The Center for Forensic Science Research and Education, Willow Grove, Pennsylvania 19090, United States
| | - Glendon J Parker
- Department of Environmental Toxicology, University of California, Davis, California 95616, United States
| |
Collapse
|
7
|
Vinod SP, Vignesh R, Priyanka M, Tirumurugaan KG, Sivaselvam SN, Raj GD. Generation of single stranded DNA with selective affinity to bovine spermatozoa. Anim Biosci 2021; 34:1579-1589. [PMID: 32882770 PMCID: PMC8495356 DOI: 10.5713/ajas.20.0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 07/23/2020] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE This study was conducted to generate single stranded DNA oligonucleotides with selective affinity to bovine spermatozoa, assess its binding potential and explore its potential utility in trapping spermatozoa from suspensions. METHODS A combinatorial library of 94 mer long oligonucleotide was used for systematic evolution of ligands by exponential enrichment (SELEX) with bovine spermatozoa. The amplicons from sixth and seventh rounds of SELEX were sequenced, and the reads were clustered employing cluster database at high identity with tolerance (CD-HIT) and FASTAptamer. The enriched nucleotides were predicted for secondary structures by Mfold, motifs by Multiple Em for Motif Elicitation and 5' labelled with biotin/6-FAM to determine the binding potential and binding pattern. RESULTS We generated 14.1 and 17.7 million reads from sixth and seventh rounds of SELEX respectively to bovine spermatozoa. The CD-HIT clustered 78,098 and 21,196 reads in the top ten clusters and FASTAptamer identified 2,195 and 4,405 unique sequences in the top three clusters from the sixth and seventh rounds, respectively. The identified oligonucleotides formed secondary structures with delta G values between -1.17 to -26.18 kcal/mol indicating varied stability. Confocal imaging with the oligonucleotides from the seventh round revealed different patterns of binding to bovine spermatozoa (fluorescence of the whole head, spot of fluorescence in head and mid- piece and tail). Use of a 5'-biotin tagged oligonucleotide from the sixth round at 100 pmol with 4×106 spermatozoa could trap almost 80% from the suspension. CONCLUSION The binding patterns and ability of the identified oligonucleotides confirms successful optimization of the SELEX process and generation of aptamers to bovine spermatozoa. These oligonucleotides provide a quick approach for selective capture of spermatozoa from complex samples. Future SELEX rounds with X- or Y- enriched sperm suspension will be used to generate oligonucleotides that bind to spermatozoa of a specific sex type.
Collapse
Affiliation(s)
- Sivadasan Pathiyil Vinod
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| | - Rajamani Vignesh
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| | - Mani Priyanka
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| | - Krishnaswamy Gopalan Tirumurugaan
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| | - Salem Nagalingam Sivaselvam
- Department of Animal Genetics and Breeding, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| | - Gopal Dhinakar Raj
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Chennai – 600051, India
| |
Collapse
|
8
|
Zhang Y, Zhou N. Electrochemical Biosensors Based on Micro‐fabricated Devices for Point‐of‐Care Testing: A Review. ELECTROANAL 2021. [DOI: 10.1002/elan.202100281] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Yuting Zhang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Biotechnology Jiangnan University Wuxi 214122 China
| | - Nandi Zhou
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Biotechnology Jiangnan University Wuxi 214122 China
| |
Collapse
|
9
|
Normandeau F, Ng A, Beaugrand M, Juncker D. Spatial Bias in Antibody Microarrays May Be an Underappreciated Source of Variability. ACS Sens 2021; 6:1796-1806. [PMID: 33973474 DOI: 10.1021/acssensors.0c02613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antibody microarrays enable multiplexed protein detection with minimal reagent consumption, but they continue to be plagued by lack of reproducibility. Chemically functionalized glass slides are used as substrates, yet antibody binding spatial inhomogeneity across the slide has not been analyzed in antibody microarrays. Here, we characterize spatial bias across five commercial slides patterned with nine overlapping dense arrays (by combining three buffers and three different antibodies), and we measure signal variation for both antibody immobilization and the assay signal, generating 270 heatmaps. Spatial bias varied across models, and the coefficient of variation ranged from 4.6 to 50%, which was unexpectedly large. Next, we evaluated three layouts of spot replicates-local, random, and structured random-for their capacity to predict assay variation. Local replicates are widely used but systematically underestimate the whole-slide variation by up to seven times; structured random replicates gave the most accurate estimation. Our results highlight the risk and consequences of using local replicates: the underappreciation of spatial bias as a source of variability, poor assay reproducibility, and possible overconfidence in assay results. We recommend the detailed characterization of spatial bias for antibody microarrays and the description and use of distributed positive replicates for research and clinical applications.
Collapse
Affiliation(s)
- Frédéric Normandeau
- McGill Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Andy Ng
- McGill Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Maiwenn Beaugrand
- McGill Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - David Juncker
- McGill Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
10
|
Gilboa T, Maley AM, Ogata AF, Wu C, Walt DR. Sequential Protein Capture in Multiplex Single Molecule Arrays: A Strategy for Eliminating Assay Cross-Reactivity. Adv Healthc Mater 2021; 10:e2001111. [PMID: 32893488 PMCID: PMC8238389 DOI: 10.1002/adhm.202001111] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/12/2020] [Indexed: 12/31/2022]
Abstract
Measurements of multiple biomolecules within the same biological sample are important for many clinical applications to enable accurate disease diagnosis or classification. These disease-related biomarkers often exist at very low levels in biological fluids, necessitating ultrasensitive measurement methods. Single-molecule arrays (Simoa), a bead-based digital enzyme-linked immunosorbent assay, is the current state of the art for ultrasensitive protein detection and can detect sub-femtomolar protein concentrations, but its ability to achieve high-order multiplexing without cross-reactivity remains a challenge. Here, a sequential protein capture approach for multiplex Simoa assays is implemented to eliminate cross-reactivity between binding reagents by sequentially capturing each protein analyte and then incubating each capture bead with only its corresponding detection antibody. This strategy not only reduces cross-reactivity to background levels and significantly improves measurement accuracies, but also enables higher-order multiplexing. As a proof of concept, the sequential multiplex Simoa assay is used to measure five different cytokines in plasma samples from Coronavirus Disease 2019 (COVID-19) patients. The ultrasensitive sequential multiplex Simoa assays will enable the simultaneous measurements of multiple low-abundance analytes in a time- and cost-effective manner and will prove especially critical in many cases where sample volumes are limited.
Collapse
Affiliation(s)
- Tal Gilboa
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Adam M Maley
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Alana F Ogata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Connie Wu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
11
|
He Z, Huffman J, Curtin K, Garner KL, Bowdridge EC, Li X, Nurkiewicz TR, Li P. Composable Microfluidic Plates (cPlate): A Simple and Scalable Fluid Manipulation System for Multiplexed Enzyme-Linked Immunosorbent Assay (ELISA). Anal Chem 2021; 93:1489-1497. [PMID: 33326204 DOI: 10.1021/acs.analchem.0c03651] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Enzyme-linked immunosorbent assay (ELISA) is the gold standard method for protein biomarkers. However, scaling up ELISA for multiplexed biomarker analysis is not a trivial task due to the lengthy procedures for fluid manipulation and high reagent/sample consumption. Herein, we present a highly scalable multiplexed ELISA that achieves a similar level of performance to commercial single-target ELISA kits as well as shorter assay time, less consumption, and simpler procedures. This ELISA is enabled by a novel microscale fluid manipulation method, composable microfluidic plates (cPlate), which are comprised of miniaturized 96-well plates and their corresponding channel plates. By assembling and disassembling the plates, all of the fluid manipulations for 96 independent ELISA reactions can be achieved simultaneously without any external fluid manipulation equipment. Simultaneous quantification of four protein biomarkers in serum samples is demonstrated with the cPlate system, achieving high sensitivity and specificity (∼ pg/mL), short assay time (∼1 h), low consumption (∼5 μL/well), high scalability, and ease of use. This platform is further applied to probe the levels of three protein biomarkers related to vascular dysfunction under pulmonary nanoparticle exposure in rat's plasma. Because of the low cost, portability, and instrument-free nature of the cPlate system, it will have great potential for multiplexed point-of-care testing in resource-limited regions.
Collapse
Affiliation(s)
- Ziyi He
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Justin Huffman
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Kathrine Curtin
- Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Krista L Garner
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia 26506, United States.,Center for Inhalation Toxicology, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia 26506, United States.,Center for Inhalation Toxicology, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Xiaojun Li
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia 26506, United States.,Center for Inhalation Toxicology, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Peng Li
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| |
Collapse
|
12
|
Abstract
Recent advances in biosensing analytical platforms have brought relevant outcomes for novel diagnostic and therapy-oriented applications. In this context, hydrogels have emerged as appealing matrices to locally confine biomolecules onto sensing surfaces under solution mimetic conditions, preserving their structural integrity and function. Here, we describe the application of a self-assembling peptide hydrogel as a suitable matrix for 3D microarray bioassays. The hydrogel is printable and self-adhesive and allows for fast analyte diffusion. As a showcase example, we describe its application in a diagnostic immunoassay for the detection of arbovirus infection.
Collapse
|
13
|
Paquet-Mercier F, Juncker D, Bergeron S. Precise Chip-to-Chip Reagent Transfer for Cross-Reactivity-Free Multiplex Sandwich Immunoassays. Methods Mol Biol 2021; 2237:141-149. [PMID: 33237415 DOI: 10.1007/978-1-0716-1064-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Common multiplex sandwich immunoassays suffer from cross-reactivity due to the mixing of detection antibodies and the combinatorial, undesired interaction between all reagents and analytes. Here we present the snap chip to perform antibody colocalization microarrays that eliminates undesirable interactions by running an array of singleplex assays realized by sequestering detection antibodies in individual nanodroplets. When detecting proteins in biological fluids, the absence of cross-reactivity allows a higher level of multiplexing, reduced background, increased sensitivity, and ensures accurate and specific results. The use of the snap chip is illustrated by measuring highly related analytes such as proteins isoforms and phospho-proteins, both particularly prone to cross-reactivity, in a single experiment. The main steps of the protocol are preparation of sample, incubation on an assay slide harboring the microarrayed capture antibodies, transfer of the microarrayed detection antibodies on their cognate spots, and measurement of the assay results by fluorescence.
Collapse
Affiliation(s)
| | - David Juncker
- McGill University and Génome Québec Innovation Centre, Montreal, QC, Canada
- Biomedical Engineering Department, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
14
|
Bime C, Camp SM, Casanova N, Oita RC, Ndukum J, Lynn H, Garcia JGN. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020; 226:105-115. [PMID: 32599095 PMCID: PMC7319618 DOI: 10.1016/j.trsl.2020.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Recent innovations in translational research have ushered an exponential increase in the discovery of novel biomarkers, thereby elevating the hope for deeper insights into "personalized" medicine approaches to disease phenotyping and care. However, a critical gap exists between the fast pace of biomarker discovery and the successful translation to clinical use. This gap underscores the fundamental biomarker conundrum across various acute and chronic disorders: how does a biomarker address a specific unmet need? Additionally, the gap highlights the need to shift the paradigm from a focus on biomarker discovery to greater translational impact and the need for a more streamlined drug approval process. The unmet need for biomarkers in acute respiratory distress syndrome (ARDS) is for reliable and validated biomarkers that minimize heterogeneity and allow for stratification of subject selection for enrollment in clinical trials of tailored therapies. This unmet need is particularly highlighted by the ongoing SARS-CoV-2/COVID-19 pandemic. The unprecedented numbers of COVID-19-induced ARDS cases has strained health care systems across the world and exposed the need for biomarkers that would accelerate drug development and the successful phenotyping of COVID-19-infected patients at risk for development of ARDS and ARDS mortality. Accordingly, this review discusses the current state of ARDS biomarkers in the context of the drug development pipeline and highlight gaps between biomarker discovery and clinical implementation while proposing potential paths forward. We discuss potential ARDS biomarkers by category and by context of use, highlighting progress in the development continuum. We conclude by discussing challenges to successful translation of biomarker candidates to clinical impact and proposing possible novel strategies.
Collapse
Affiliation(s)
- Christian Bime
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona.
| | - Sara M Camp
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Nancy Casanova
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Radu C Oita
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Juliet Ndukum
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Heather Lynn
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
15
|
Multiplexed Immunosensor Based on the Amperometric Transduction for Monitoring of Marine Pollutants in Sea Water. SENSORS 2020; 20:s20195532. [PMID: 32992549 PMCID: PMC7584025 DOI: 10.3390/s20195532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Abstract
Environmental pollutants vigilance is one of the main problems that the aquaculture industry has to face with the objective to ensure the quality of their products and prevent entrance in the food chain that finally may arrive to the consumer. Contaminants such as hormones, antibiotics or biocides are especially relevant due to their toxicity, pharmacological effect or hormonal activity that can be considered harmful for the final consumer. The contaminants can be detected in the environment where the food is growing, and their concentration can be found (i.e., seawater) in the range of µg·L−1, ng·L−1 or even in lower concentrations. Thus, sensitive and selective methods for their monitoring are required to avoid their arrival in the food chain. Here, the development of a multiplexed amperometric biosensor is described, based on the use of specific antibodies to reach the necessary detectability to measure the targeted contaminants directly in seawater. The multiplexed immunosensor allows the detection of four relevant pollutants, such as el Irgarol 1051, sulfapyridine, chloramphenicol and estradiol, reaching an IC50 of 5.04 ± 0.29, 3.45 ± 0.29, 4.17 ± 0.44 and 5.94 ± 0.28 µg·L−1, directly measured in seawater.
Collapse
|
16
|
Günther A, Becker M, Göpfert J, Joos T, Schneiderhan-Marra N. Comparison of Bead-Based Fluorescence Versus Planar Electrochemiluminescence Multiplex Immunoassays for Measuring Cytokines in Human Plasma. Front Immunol 2020; 11:572634. [PMID: 33101295 PMCID: PMC7546899 DOI: 10.3389/fimmu.2020.572634] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022] Open
Abstract
This study compared two 96-well multiplex immunoassay platforms for analytical performance in assessing cytokine concentrations in standards, quality controls and human plasma samples (n = 62), and evaluated assay time requirements. Assays included a bead-based fluorescence MILLIPLEX® assay/Luminex fluorescence platform (LMX) and three kits from Meso Scale Discovery (MSD) in planar electrochemiluminescence format. The LMX kit evaluated 21 cytokines and the MSD kits evaluated 10 cytokines each, with 16 overlapping cytokines between platforms. Both assays provided good reproducibility in standard curves for all analytes. Interassay CVs of shared analytes showed average kit quality control CVs ranging 1.9–18.2% for LMX and 2.4–13.9% for MSD. The MSD platform had lower LLoQs than LMX for 14/16 shared cytokines. For IL-17, the LLoQ was lower with LMX than MSD, and the LLoQs for IL-6 were similar. Although MSD calibration curves indicated lower LLoQs for most of those analytes, many more cytokines in human plasma samples were not detected by MSD than by LMX. The ULoQs were higher in LMX versus MSD assays for 13/16 shared analytes, lower than MSD for IL-17, and equivalent between assays for IL-6 and MIP-1α. Bland-Altman plots indicated that MSD classified 13/16 shared analytes as concentrations lower than by LMX. Time and motion analysis indicated that total mean assay times were 20 h 28 m and 21 h 33 m for LMX and MSD, respectively, including an overnight (17 h) incubation. The MSD assays employed a manufacturer-approved overnight incubation instead of the standard 2-h incubation, which kit instructions suggest might increase detection sensitivity. Hands-on labor time averaged 1 h 37 m for LMX and 2 h 33 m for MSD. In summary, assay selection factors should include selection of specific markers of interest, time and cost considerations, and anticipated cytokine concentrations in prospective samples.
Collapse
Affiliation(s)
- Anna Günther
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | - Matthias Becker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | - Jens Göpfert
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | - Thomas Joos
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
17
|
Kvas M, Teixeira AG, Chiang B, Frampton JP. Aqueous two-phase system antibody confinement enables cost-effective analysis of protein analytes by sandwich enzyme-linked immunosorbent assay with minimal optical crosstalk. Analyst 2020; 145:5458-5465. [PMID: 32578585 DOI: 10.1039/d0an00699h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
An aqueous two-phase system formed from polyethylene glycol and dextran was used to uniformly coat the bottom surfaces of the wells of standard 96-well assay plates with capture and detection antibodies to improve the performance and cost-effectiveness of sandwich enzyme-linked immunosorbent assay (ELISA). Using this approach, limits of detection and linear dynamic range values comparable to those obtained for conventional sandwich ELISA were obtained using considerably lower antibody quantities due to the much lower reagent volumes required when antibodies are applied in a dextran solution beneath a polyethylene glycol overlay. Confinement of the antibody reagents to the bottom surfaces of the wells within the dextran phase also dramatically decreased the optical crosstalk present between neighboring wells when using transparent microplates. Adaptation of the conventional single sandwich ELISA for aqueous two-phase system antibody confinement was demonstrated by analysis of standard curves for C-reactive protein, transforming growth factor beta 1, and the chemokine CXCL10.
Collapse
Affiliation(s)
- Maia Kvas
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada.
| | | | | | | |
Collapse
|
18
|
Tongdee M, Yamanishi C, Maeda M, Kojima T, Dishinger J, Chantiwas R, Takayama S. One-incubation one-hour multiplex ELISA enabled by aqueous two-phase systems. Analyst 2020; 145:3517-3527. [PMID: 32248215 PMCID: PMC7237315 DOI: 10.1039/d0an00383b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This work describes a convenient one-hour enzyme-linked immunosorbent assay (ELISA) formulated with conventional antibodies and horseradish peroxidase (HRP) reagents. The method utilizes aqueous two-phase system (ATPS) droplet formation based on poly(ethylene glycol) (PEG)-containing sample solution-triggered rehydration of dehydrated dextran (DEX) spots that contain all antibody reagents. Key advances in this paper include development of a formulation that allows a quick 1-hour overall incubation time and a procedure where inclusion of the HRP reagent in the PEG solution reduces the number of washing and incubation steps required to perform this assay. As an assay application, a 5-plex cytokine test compares cytokine secretion of differentially-treated human ThP-1 macrophages. Given the use of only readily available reagents and a common Western blot imaging system for the readout, this method is envisioned to be broadly applicable to a variety of multiplex immunoassays. To facilitate broader use, companion image processing software as an ImageJ plugin is also described and provided.
Collapse
Affiliation(s)
- Mintra Tongdee
- Department of Chemistry and Center of Excellence for Innovation in Chemistry and Flow Innovation-Research for Science and Technology Laboratories (FIRST Labs), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok 10400, Thailand and Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia, USA
| | - Cameron Yamanishi
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia, USA
| | - Midori Maeda
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia, USA
| | - Taisuke Kojima
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia, USA
| | | | - Rattikan Chantiwas
- Department of Chemistry and Center of Excellence for Innovation in Chemistry and Flow Innovation-Research for Science and Technology Laboratories (FIRST Labs), Faculty of Science, Mahidol University, Rama VI Rd., Bangkok 10400, Thailand
| | - Shuichi Takayama
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia, USA
| |
Collapse
|
19
|
Kim KH, Kim JY, Yoo JS. Mass spectrometry analysis of glycoprotein biomarkers in human blood of hepatocellular carcinoma. Expert Rev Proteomics 2019; 16:553-568. [PMID: 31145639 DOI: 10.1080/14789450.2019.1626235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kwang Hoe Kim
- Biomedical Omics Group, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jin Young Kim
- Biomedical Omics Group, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Jong Shin Yoo
- Biomedical Omics Group, Korea Basic Science Institute, Cheongju, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
20
|
Method Validation by CPTAC Guidelines for Multi-protein Marker Assays Using Multiple Reaction Monitoring-mass Spectrometry. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-018-0454-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Gagni P, Romanato A, Bergamaschi G, Bettotti P, Vanna R, Piotto C, Morasso CF, Chiari M, Cretich M, Gori A. A self-assembling peptide hydrogel for ultrarapid 3D bioassays. NANOSCALE ADVANCES 2019; 1:490-497. [PMID: 36132256 PMCID: PMC9473263 DOI: 10.1039/c8na00158h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/22/2018] [Indexed: 05/08/2023]
Abstract
Biosensing analytical platforms rely on the intimate structure-function relationship of immobilized probes. In this context, hydrogels are appealing semi-wet systems to locally confine biomolecules while preserving their structural integrity and function. Yet, limitations imposed by biomolecule diffusion rates or fabrication difficulties still hamper their broad application. Here, using a self-assembling peptide, a printable and self-adhesive hydrogel was obtained and applied to fabricate arrays of localized bio-functional 3D microenvironments on analytical interfaces. This soft matrix represents a robust and versatile material, allowing fast and selective tuning of analyte diffusion, which is exploited here to run in-gel immunoassays under solution-like conditions in an unprecedented (<10 min) time frame. The developed material overcomes major limitations associated with hydrogels for bioassays, widening the prospects for easy fabrication of multifunctional bio-interfaces for high-throughput, molecular recognition assays.
Collapse
Affiliation(s)
- Paola Gagni
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| | - Alessandro Romanato
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| | - Greta Bergamaschi
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| | - Paolo Bettotti
- Nanoscience Laboratory, Department of Physics, University of Trento Via Sommarive 14 38123 Povo Italy
| | - Renzo Vanna
- Istituti Clinici Scientifici Maugeri IRCCS Via Maugeri 4 27100 Pavia Italy
| | - Chiara Piotto
- Nanoscience Laboratory, Department of Physics, University of Trento Via Sommarive 14 38123 Povo Italy
| | - Carlo F Morasso
- Istituti Clinici Scientifici Maugeri IRCCS Via Maugeri 4 27100 Pavia Italy
| | - Marcella Chiari
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| | - Marina Cretich
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| | - Alessandro Gori
- National Research Council of Italy, Istituto di Chimica del Riconoscimento Molecolare (ICRM) Via Mario Bianco, 9 20131-Milano Italy
| |
Collapse
|
22
|
Núñez C. Blood-based protein biomarkers in breast cancer. Clin Chim Acta 2018; 490:113-127. [PMID: 30597138 DOI: 10.1016/j.cca.2018.12.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/26/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023]
Abstract
Breast cancer (BCa) is a significant healthcare problem on women worldwide. Thus, early detection is very important to reduce mortality. Furthermore, better BCa prognosis could improve selection of patients eligible for adjuvant therapy. New markers for early diagnosis, accurate prognosis and prediction of response to treatment are necessary to improve BCa care. The present review summarizes important aspects of the potential usefulness of modern technologies, strategies, and scientific findings in proteomic research for discovery of breast cancer-associated blood-based protein biomarkers in the clinic.
Collapse
Affiliation(s)
- Cristina Núñez
- Research Unit, Hospital Universitario Lucus Augusti (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain.
| |
Collapse
|
23
|
Dagher M, Kleinman M, Ng A, Juncker D. Ensemble multicolour FRET model enables barcoding at extreme FRET levels. NATURE NANOTECHNOLOGY 2018; 13:925-932. [PMID: 30061659 DOI: 10.1038/s41565-018-0205-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 06/17/2018] [Indexed: 05/02/2023]
Abstract
Quantitative models of Förster resonance energy transfer (FRET)-pioneered by Förster-define our understanding of FRET and underpin its widespread use. However, multicolour FRET (mFRET), which arises between multiple, stochastically distributed fluorophores, lacks a mechanistic model and remains intractable. mFRET notably arises in fluorescently barcoded microparticles, resulting in a complex, non-orthogonal fluorescence response that impedes their encoding and decoding. Here, we introduce an ensemble mFRET (emFRET) model, and apply it to guide barcoding into regimes with extreme FRET. We further introduce a facile, proportional multicolour labelling method using oligonucleotides as homogeneous linkers. A total of 580 barcodes were rapidly designed and validated using four dyes-with FRET efficiencies reaching 76%-and used for multiplexed immunoassays with cytometric readout and fully automated decoding. The emFRET model helps to expand the barcoding capacity of barcoded microparticles using common organic dyes and will benefit other applications subject to stochastic mFRET.
Collapse
Affiliation(s)
- Milad Dagher
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - Michael Kleinman
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - Andy Ng
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada
| | - David Juncker
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada.
- McGill University and Genome Quebec Innovation Center, Montreal, Quebec, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
24
|
Krishnan VV, Selvan SR, Parameswaran N, Venkateswaran N, Luciw PA, Venkateswaran KS. Proteomic profiles by multiplex microsphere suspension array. J Immunol Methods 2018; 461:1-14. [PMID: 30003895 DOI: 10.1016/j.jim.2018.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/03/2018] [Accepted: 07/05/2018] [Indexed: 02/08/2023]
Abstract
Advances in high-throughput proteomic approaches have provided substantial momentum to novel disease-biomarker discovery research and have augmented the quality of clinical studies. Applications based on multiplexed microsphere suspension array technology are making strong in-roads into the clinical diagnostic/prognostic practice. Conventional proteomic approaches are designed to discover a broad set of proteins that are associated with a specific medical condition. In comparison, multiplex microsphere immunoassays use quantitative measurements of selected set(s) of specific/particular molecular markers such as cytokines, chemokines, pathway signaling or disease-specific markers for detection, metabolic disorders, cancer, and infectious agents causing human, plant and animal diseases. This article provides a foundation to the multiplexed microsphere suspension array technology, with an emphasis on the improvements in the technology, data analysis approaches, and applications to translational and clinical research with implications for personalized and precision medicine.
Collapse
Affiliation(s)
- Viswanathan V Krishnan
- Department of Chemistry, California State University, Fresno, CA 93750, United States; Department of Medical Pathology and Laboratory Medicine, University of California School of Medicine, Sacramento, CA 95817, United States.
| | | | | | | | - Paul A Luciw
- Center for Comparative Medicine, University of California Davis, Davis, CA 95616, United States; Department of Medical Pathology and Laboratory Medicine, University of California School of Medicine, Sacramento, CA 95817, United States
| | | |
Collapse
|
25
|
Sanchis A, Salvador JP, Campbell K, Elliott CT, Shelver WL, Li QX, Marco MP. Fluorescent microarray for multiplexed quantification of environmental contaminants in seawater samples. Talanta 2018; 184:499-506. [PMID: 29674074 DOI: 10.1016/j.talanta.2018.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 01/10/2023]
Abstract
The development of a fluorescent multiplexed microarray platform able to detect and quantify a wide variety of pollutants in seawater is reported. The microarray platform has been manufactured by spotting 6 different bioconjugate competitors and it uses a cocktail of 6 monoclonal or polyclonal antibodies raised against important families of chemical pollutants such as triazine biocide (i.e. Irgarol 1051®), sulfonamide and chloramphenicol antibiotics, polybrominated diphenyl ether flame-retardant (PBDE, i.e. BDE-47), hormone (17β-estradiol), and algae toxin (domoic acid). These contaminants were selected as model analytes, however, the platform developed has the potential to detect a broader group of compounds based on the cross-reactivity of the immunoreagents used. The microarray chip is able to simultaneously determine these families of contaminants directly in seawater samples reaching limits of detection close to the levels found in contaminated areas (Irgarol 1051®, 0.19 ± 0,06 µg L-1; sulfapyridine, 0.17 ± 0.07 µg L-1; chloramphenicol, 0.11 ± 0.03 µg L-1; BDE-47, 2.71 ± 1.13 µg L-1; 17β-estradiol, 0.94 ± 0.30 µg L-1 and domoic acid, 1.71 ± 0.30 µg L-1). Performance of the multiplexed microarray chip was assessed by measuring 38 blind spiked seawater samples containing either one of these contaminants or mixtures of them. The accuracy found was very good and the coefficient of variation was < 20% in all the cases. No sample pre-treatment was necessary, and the results could be obtained in just 1 h 30 min. The microarray shows high sample throughput capabilities, being able to measure simultaneously more than 68 samples and screen them for a significant number of chemical contaminants of interest in environmental screening programs.
Collapse
Affiliation(s)
- Ana Sanchis
- Nanobiotechnology for diagnostics (Nb4D), Department of Chemical and Biomolecular Nanotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - J-Pablo Salvador
- Nanobiotechnology for diagnostics (Nb4D), Department of Chemical and Biomolecular Nanotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Katrina Campbell
- Institute of Agri-Food and Land Use (IAFLU), School of Biological Sciences, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, Northern Ireland, United Kingdom
| | - Christopher T Elliott
- Institute of Agri-Food and Land Use (IAFLU), School of Biological Sciences, Queen's University Belfast, David Keir Building, Stranmillis Road, Belfast BT9 5AG, Northern Ireland, United Kingdom
| | - Weilin L Shelver
- USDA-ARS, Red River Valley Agricultural Research Center, Biosciences Research Laboratory, Fargo, ND, USA
| | - Qing X Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, USA
| | - M-Pilar Marco
- Nanobiotechnology for diagnostics (Nb4D), Department of Chemical and Biomolecular Nanotechnology, Institute for Advanced Chemistry of Catalonia (IQAC) of the Spanish Council for Scientific Research (CSIC), Jordi Girona 18-26, 08034 Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
26
|
Chen Z, Dodig-Crnković T, Schwenk JM, Tao SC. Current applications of antibody microarrays. Clin Proteomics 2018; 15:7. [PMID: 29507545 PMCID: PMC5830343 DOI: 10.1186/s12014-018-9184-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/19/2018] [Indexed: 12/14/2022] Open
Abstract
The concept of antibody microarrays is one of the most versatile approaches within multiplexed immunoassay technologies. These types of arrays have increasingly become an attractive tool for the exploratory detection and study of protein abundance, function, pathways, and potential drug targets. Due to the properties of the antibody microarrays and their potential use in basic research and clinical analytics, various types of antibody microarrays have already been developed. In spite of the growing number of studies utilizing this technique, few reviews about antibody microarray technology have been presented to reflect the quality and future uses of the generated data. In this review, we provide a summary of the recent applications of antibody microarray techniques in basic biology and clinical studies, providing insights into the current trends and future of protein analysis.
Collapse
Affiliation(s)
- Ziqing Chen
- Key Laboratory of Systems Biomedicine, (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Tea Dodig-Crnković
- Affinity Proteomics, SciLifeLab, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Jochen M. Schwenk
- Affinity Proteomics, SciLifeLab, KTH - Royal Institute of Technology, 171 65 Solna, Sweden
| | - Sheng-ce Tao
- Key Laboratory of Systems Biomedicine, (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240 China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240 China
| |
Collapse
|
27
|
Li H, Bergeron S, Larkin H, Juncker D. Snap Chip for Cross-reactivity-free and Spotter-free Multiplexed Sandwich Immunoassays. J Vis Exp 2017. [PMID: 29155743 DOI: 10.3791/56230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Multiplexed protein analysis has shown superior diagnostic sensitivity and accuracy compared to single proteins. Antibody microarrays allow for thousands of micro-scale immunoassays performed simultaneously on a single chip. Sandwich assay format improves assay specificity by detecting each target with two antibodies, but suffers from cross-reactivity between reagents thus limiting their multiplexing capabilities. Antibody colocalization microarray (ACM) has been developed for cross-reactivity-free multiplexed protein detection, but requires an expensive spotter on-site for microarray fabrication during assays. In this work, we demonstrate a snap chip technology that transfers reagent from microarray-to-microarray by simply snapping two chips together, thus no spotter is needed during the sample incubation and subsequent application of detection antibodies (dAbs) upon storage of pre-spotted slides, dissociating the slide preparation from assay execution. Both single and double transfer methods are presented to achieve accurate alignment between the two microarrays and the slide fabrication for both methods are described. Results show that <40 μm alignment has been achieved with double transfer, reaching an array density of 625 spots/cm2. A 50-plexed immunoassay has been conducted to demonstrate the usability of the snap chip in multiplexed protein analysis. Limits of detection of 35 proteins are in the range of pg/mL.
Collapse
Affiliation(s)
- Huiyan Li
- McGill University and Génome Québec Innovation Centre; Biomedical Engineering Department, McGill University
| | | | - Heidi Larkin
- McGill University and Génome Québec Innovation Centre; Biomedical Engineering Department, McGill University; Parallex BioAssays Inc
| | - David Juncker
- McGill University and Génome Québec Innovation Centre; Biomedical Engineering Department, McGill University;
| |
Collapse
|
28
|
Optomechanical devices for deep plasma cancer proteomics. Semin Cancer Biol 2017; 52:26-38. [PMID: 28867489 DOI: 10.1016/j.semcancer.2017.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/22/2017] [Accepted: 08/30/2017] [Indexed: 12/27/2022]
Abstract
Most of the cancer deaths could be avoided by early detection of the tumor when it is confined to its primary site and it has not metastasized. To this aim, one of the most promising strategies is the discovery and detection of protein biomarkers shed by the young tumor to the bloodstream. Proteomic technologies, mainly mass spectrometry and multiplexed immunoassays, have rapidly developed during last years with improved limits of detection and multiplexing capability. Unfortunately, these developments together major investments and large international efforts have not resulted into new useful protein biomarkers. Here, we analyze the potential and limitations of current proteomic technologies for detecting protein biomarkers released into circulation by the tumor. We find that these technologies can hardly probe the deepest region of the plasma proteome, at concentrations below the pg/mL level, where protein biomarkers for early cancer detection may exist. This clearly indicates the need of incorporating novel ultrasensitive techniques to the proteomic tool-box that can cover the inaccessible regions of the plasma proteome. We here propose biological detectors based on nanomechanical systems for discovery and detection of cancer protein biomarkers in plasma. We review the modes of operation of these devices, putting our focus on recent developments on nanomechanical sandwich immunoassays and nanomechanical spectrometry. The first technique enables reproducible immunodetection of proteins at concentrations well below the pg/mL level, with a limit of detection on the verge of 10 ag/mL. This technology can potentially detect low abundance tumor-associated proteins in plasma at the very early stages of the tumor. The second technique enables the identification of individual intact proteins by two physical coordinates, the mass and stiffness, instead of the mass-to-charge ratio of the protein constituents. This technology enormously simplifies the identification of proteins and it can provide useful information on interactions and posttranslational modifications, that otherwise is lost in mass spectrometry.
Collapse
|
29
|
Kim H, Yu SJ, Yeo I, Cho YY, Lee DH, Cho Y, Cho EJ, Lee JH, Kim YJ, Lee S, Jun J, Park T, Yoon JH, Kim Y. Prediction of Response to Sorafenib in Hepatocellular Carcinoma: A Putative Marker Panel by Multiple Reaction Monitoring-Mass Spectrometry (MRM-MS). Mol Cell Proteomics 2017; 16:1312-1323. [PMID: 28550167 DOI: 10.1074/mcp.m116.066704] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
Sorafenib is the only standard treatment for unresectable hepatocellular carcinoma (HCC), but it provides modest survival benefits over placebo, necessitating predictive biomarkers of the response to sorafenib. Serum samples were obtained from 115 consecutive patients with HCC before sorafenib treatment and analyzed by multiple reaction monitoring-mass spectrometry (MRM-MS) and ELISA to quantify candidate biomarkers. We verified a triple-marker panel to be predictive of the response to sorafenib by MRM-MS, comprising CD5 antigen-like (CD5L), immunoglobulin J (IGJ), and galectin-3-binding protein (LGALS3BP), in HCC patients. This panel was a significant predictor (AUROC > 0.950) of the response to sorafenib treatment, having the best cut-off value (0.4) by multivariate analysis. In the training set, patients who exceeded this cut-off value had significantly better overall survival (median, 21.4 months) than those with lower values (median, 8.6 months; p = 0.001). Further, a value that was lower than this cutoff was an independent predictor of poor overall survival [hazard ratio (HR), 2.728; 95% confidence interval (CI), 1.312-5.672; p = 0.007] and remained an independent predictive factor of rapid progression (HR, 2.631; 95% CI, 1.448-4.780; p = 0.002). When applied to the independent validation set, levels of the cut-off value for triple-marker panel maintained their prognostic value for poor clinical outcomes. On the contrast, the triple-marker panel was not a prognostic factor for patients who were treated with transarterial chemoembolization (TACE). The discriminatory signature of a triple-marker panel provides new insights into targeted proteomic biomarkers for individualized sorafenib therapy.
Collapse
Affiliation(s)
- Hyunsoo Kim
- From the ‡Department of Biomedical Engineering.,§Institute of Medical and Biological Engineering, Medical Research Center, and
| | - Su Jong Yu
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Injun Yeo
- From the ‡Department of Biomedical Engineering
| | - Young Youn Cho
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Dong Hyeon Lee
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Yuri Cho
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Eun Ju Cho
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Jeong-Hoon Lee
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | - Yoon Jun Kim
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea
| | | | - Jongsoo Jun
- **Department of Statistics, Seoul National University, Daehak-dong, Seoul 151-742 Korea
| | - Taesung Park
- ‖Interdisciplinary program in Bioinformatics and.,**Department of Statistics, Seoul National University, Daehak-dong, Seoul 151-742 Korea
| | - Jung-Hwan Yoon
- ¶Department of Internal Medicine and Liver Research Institute, Yongon-Dong, Seoul 110-799 Korea;
| | - Youngsoo Kim
- From the ‡Department of Biomedical Engineering; .,§Institute of Medical and Biological Engineering, Medical Research Center, and
| |
Collapse
|
30
|
Li Z, Li Z, Jiang J, Xu D. Simultaneous detection of various contaminants in milk based on visualized microarray. Food Control 2017. [DOI: 10.1016/j.foodcont.2016.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
31
|
Laforte V, Lo PS, Li H, Juncker D. Antibody Colocalization Microarray for Cross-Reactivity-Free Multiplexed Protein Analysis. Methods Mol Biol 2017; 1619:239-261. [PMID: 28674891 DOI: 10.1007/978-1-4939-7057-5_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Measuring many proteins at once is of great importance to the idea of personalized medicine, in order to get a snapshot of a person's health status. We describe the antibody colocalization microarray (ACM), a variant of antibody microarrays which avoids reagent-induced cross-reactivity by printing individual detection antibodies atop their corresponding capture antibodies. We discuss experimental parameters that are critical for the success of ACM experiments, namely, the printing positional accuracy needed for the two printing rounds and the need for protecting dried spots during the second printing round. Using small sample volumes (less than 30 μL) and small quantities of reagents, up to 108 different targets can be measured in hundreds of samples with great specificity and sensitivity.
Collapse
Affiliation(s)
- Véronique Laforte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, Canada, H3A 2B4
- Department of Biomedical Engineering, McGill University, 3775 University St., Montreal, QC, Canada, H3A 2B4
- McGill University and Genome Quebec Innovation Center, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, Canada, H3A 0G1
| | - Pik-Shan Lo
- Department of Biomedical Engineering, McGill University, 3775 University St., Montreal, QC, Canada, H3A 2B4
- McGill University and Genome Quebec Innovation Center, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, Canada, H3A 0G1
| | - Huiyan Li
- Department of Biomedical Engineering, McGill University, 3775 University St., Montreal, QC, Canada, H3A 2B4
- McGill University and Genome Quebec Innovation Center, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, Canada, H3A 0G1
| | - David Juncker
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, Canada, H3A 2B4.
- Department of Biomedical Engineering, McGill University, 3775 University St., Montreal, QC, Canada, H3A 2B4.
- McGill University and Genome Quebec Innovation Center, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, Canada, H3A 0G1.
- Micro and Nanobioengineering Laboratory, Department of Biomedical Engineering, McGill University, 740 Dr. Penfield #6206, Montreal, QC, Canada, H3A 0G1.
| |
Collapse
|
32
|
Eiden L, Yamanishi C, Takayama S, Dishinger JF. Aqueous Two-Phase System Rehydration of Antibody-Polymer Microarrays Enables Convenient Compartmentalized Multiplex Immunoassays. Anal Chem 2016; 88:11328-11334. [PMID: 27808499 DOI: 10.1021/acs.analchem.6b02960] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiplex immunoassays are rapidly increasing in popularity due to the offered advantages of increased throughput and decreased sample volume requirements. However, a major weakness inherent to multiplex enzyme-linked immunosorbent assays (ELISA) is generation of false signals through reagent-driven cross-talk. Typically, multiplex platforms necessitate bath application of antibody cocktails, increasing probability of nonspecific antibody binding, especially when multiplexing large numbers of analytes. Aqueous two-phase systems (ATPS) exploiting the phase-separating polymers poly(ethylene) glycol (PEG) and dextran (DEX) have been used to compartmentalize antibodies and prevent cross-talk in multliplex, plate-based ELISA. However, the resulting protocol is tedious and lengthy, and requires too many user steps to be practical for widespread use. Here, we report an improved, user-friendly, cross-talk-free multiplex ELISA method in which dehydrated arrays of colocalized capture and detection antibodies in DEX are prepared on multiwell plates. Addition of a PEG-based sample buffer rehydrates antibody/DEX droplets for analysis. In this report, we demonstrate rehydrated ATPS components for multiplex ELISA retain the ability to compartmentalize antibodies and prevent cross-talk, while analytes in sample buffer partition into rehydrated DEX droplets for analysis. Utility of this method was demonstrated through successful quantitative analysis of five inflammatory cytokines in lipopolysaccharide-stimulated ThP-1 cell culture supernatant.
Collapse
Affiliation(s)
- Lisa Eiden
- PHASIQ, Inc. , Ann Arbor, Michigan 48109, United States
| | - Cameron Yamanishi
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
- Department of Macromolecular Science and Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | | |
Collapse
|
33
|
Pan L, Aguilar HA, Wang L, Iliuk A, Tao WA. Three-Dimensionally Functionalized Reverse Phase Glycoprotein Array for Cancer Biomarker Discovery and Validation. J Am Chem Soc 2016; 138:15311-15314. [PMID: 27933927 DOI: 10.1021/jacs.6b10239] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glycoproteins have vast structural diversity that plays an important role in many biological processes and have great potential as disease biomarkers. Here, we report a novel functionalized reverse phase protein array (RPPA), termed polymer-based reverse phase glycoprotein array (polyGPA), to capture and profile glycoproteomes specifically, and validate glycoproteins. Nitrocellulose membrane functionalized with globular hydroxyaminodendrimers was used to covalently capture preoxidized glycans on glycoproteins from complex protein samples such as biofluids. The captured glycoproteins were subsequently detected using the same validated antibodies as in RPPA. We demonstrated the outstanding specificity, sensitivity, and quantitative capabilities of polyGPA by capturing and detecting purified as well as endogenous α-1-acid glycoprotein (AGP) in human plasma. We further applied quantitative N-glycoproteomics and the strategy to validate a panel of glycoproteins identified as potential biomarkers for bladder cancer by analyzing urine glycoproteins from bladder cancer patients or matched healthy individuals.
Collapse
Affiliation(s)
| | | | | | - Anton Iliuk
- Tymora Analytical Operations , West Lafayette, Indiana 47906, United States
| | - W Andy Tao
- Tymora Analytical Operations , West Lafayette, Indiana 47906, United States.,Center for Cancer Research, Purdue University , West Lafayette, Indiana 47907, United States
| |
Collapse
|
34
|
Zou C, Wang P, Xu Y. Bulked sample analysis in genetics, genomics and crop improvement. PLANT BIOTECHNOLOGY JOURNAL 2016; 14:1941-55. [PMID: 26990124 PMCID: PMC5043468 DOI: 10.1111/pbi.12559] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/09/2016] [Accepted: 03/12/2016] [Indexed: 05/18/2023]
Abstract
Biological assay has been based on analysis of all individuals collected from sample populations. Bulked sample analysis (BSA), which works with selected and pooled individuals, has been extensively used in gene mapping through bulked segregant analysis with biparental populations, mapping by sequencing with major gene mutants and pooled genomewide association study using extreme variants. Compared to conventional entire population analysis, BSA significantly reduces the scale and cost by simplifying the procedure. The bulks can be built by selection of extremes or representative samples from any populations and all types of segregants and variants that represent wide ranges of phenotypic variation for the target trait. Methods and procedures for sampling, bulking and multiplexing are described. The samples can be analysed using individual markers, microarrays and high-throughput sequencing at all levels of DNA, RNA and protein. The power of BSA is affected by population size, selection of extreme individuals, sequencing strategies, genetic architecture of the trait and marker density. BSA will facilitate plant breeding through development of diagnostic and constitutive markers, agronomic genomics, marker-assisted selection and selective phenotyping. Applications of BSA in genetics, genomics and crop improvement are discussed with their future perspectives.
Collapse
Affiliation(s)
- Cheng Zou
- Institute of Crop Science, National Key Facility of Crop Gene Resources and Genetic Improvement, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Pingxi Wang
- Institute of Crop Science, National Key Facility of Crop Gene Resources and Genetic Improvement, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yunbi Xu
- Institute of Crop Science, National Key Facility of Crop Gene Resources and Genetic Improvement, Chinese Academy of Agricultural Sciences, Beijing, China.
- International Maize and Wheat Improvement Center (CIMMYT), Texcoco, Mexico.
| |
Collapse
|
35
|
Ostromohov N, Bercovici M, Kaigala GV. Delivery of minimally dispersed liquid interfaces for sequential surface chemistry. LAB ON A CHIP 2016; 16:3015-23. [PMID: 27354032 DOI: 10.1039/c6lc00473c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
We present a method for sequential delivery of reagents to a reaction site with minimal dispersion of their interfaces. Using segmented flow to encapsulate the reagents as droplets, the dispersion between reagent plugs remains confined in a limited volume, while being transmitted to the reaction surface. In close proximity to the target surface, we use a passive array of microstructures for removal of the oil phase such that the original reagent sequence is reconstructed, and only the aqueous phase reaches the reaction surface. We provide a detailed analysis of the conditions under which the method can be applied and demonstrate maintaining a transition time of 560 ms between reagents transported to a reaction site over a distance of 60 cm. We implemented the method using a vertical microfluidic probe on an open surface, allowing contact-free interaction with biological samples, and demonstrated two examples of assays implemented using the method: measurements of receptor-ligand reaction kinetics and of the fluorescence response of immobilized GFP to local variations in pH. We believe that the method can be useful for studying the dynamic response of cells and proteins to various stimuli, as well as for highly automated multi-step assays.
Collapse
Affiliation(s)
- N Ostromohov
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel. and IBM Research-Zurich, Saeumerstrasse 4, CH-8803 Rueschlikon, Switzerland.
| | - M Bercovici
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| | - G V Kaigala
- IBM Research-Zurich, Saeumerstrasse 4, CH-8803 Rueschlikon, Switzerland.
| |
Collapse
|
36
|
Cayer DM, Nazor KL, Schork NJ. Mission critical: the need for proteomics in the era of next-generation sequencing and precision medicine. Hum Mol Genet 2016; 25:R182-R189. [DOI: 10.1093/hmg/ddw214] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022] Open
|
37
|
Pla-Roca M, Altay G, Giralt X, Casals A, Samitier J. Design and development of a microarray processing station (MPS) for automated miniaturized immunoassays. Biomed Microdevices 2016; 18:64. [PMID: 27405464 DOI: 10.1007/s10544-016-0087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Here we describe the design and evaluation of a fluidic device for the automatic processing of microarrays, called microarray processing station or MPS. The microarray processing station once installed on a commercial microarrayer allows automating the washing, and drying steps, which are often performed manually. The substrate where the assay occurs remains on place during the microarray printing, incubation and processing steps, therefore the addressing of nL volumes of the distinct immunoassay reagents such as capture and detection antibodies and samples can be performed on the same coordinate of the substrate with a perfect alignment without requiring any additional mechanical or optical re-alignment methods. This allows the performance of independent immunoassays in a single microarray spot.
Collapse
Affiliation(s)
- Mateu Pla-Roca
- Nanobioengineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac, 10-12, 08028, Barcelona, Spain.
| | - Gizem Altay
- Nanobioengineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac, 10-12, 08028, Barcelona, Spain
| | - Xavier Giralt
- Robotics Laboratory, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac, 10-12, 08028, Barcelona, Spain
| | - Alícia Casals
- Robotics Laboratory, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac, 10-12, 08028, Barcelona, Spain.,Center of Research in Biomedical Engineering, Universitat Politècnica de Catalunya, Jordi Girona, 1-3, 08034, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac, 10-12, 08028, Barcelona, Spain.,The Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Maria de Luna, 11, 50018, Zaragoza, Spain.,Department of Engineering: Electronics, University of Barcelona (UB), Martí i Franquès, 1, 08028, Barcelona, Spain
| |
Collapse
|
38
|
Dias JT, Lama L, Gantelius J, Andersson-Svahn H. Minimizing antibody cross-reactivity in multiplex detection of biomarkers in paper-based point-of-care assays. NANOSCALE 2016; 8:8195-8201. [PMID: 27030365 DOI: 10.1039/c5nr09207h] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Highly multiplexed immunoassays could allow convenient screening of hundreds or thousands of protein biomarkers simultaneously in a clinical sample such as serum or plasma, potentially allowing improved diagnostic accuracy and clinical management of many conditions such as autoimmune disorders, infections, and several cancers. Currently, antibody microarray-based tests are limited in part due to cross reactivity from detection antibody reagents. Here we present a strategy that reduces the cross-reactivity between nanoparticle-bound reporter antibodies through the application of ultrasound energy. By this concept, it was possible to achieve a sensitivity 10(3)-fold (5 pg mL(-1)) lower than when no ultrasound was applied (50 ng mL(-1)) for the simultaneous detection of three different antigens. The detection limits and variability achieved with this technique rival those obtained with other types of multiplex sandwich assays.
Collapse
Affiliation(s)
- J T Dias
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH Royal Institute of Technology, Sweden.
| | - L Lama
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH Royal Institute of Technology, Sweden.
| | - J Gantelius
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH Royal Institute of Technology, Sweden.
| | - H Andersson-Svahn
- Division of Proteomics and Nanobiotechnology, Science for Life Laboratory, KTH Royal Institute of Technology, Sweden.
| |
Collapse
|
39
|
Granger JH, Schlotter NE, Crawford AC, Porter MD. Prospects for point-of-care pathogen diagnostics using surface-enhanced Raman scattering (SERS). Chem Soc Rev 2016; 45:3865-82. [DOI: 10.1039/c5cs00828j] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review highlights recent advances in the application of surface-enhanced Raman scattering (SERS) in pathogen detection and discusses many of the challenges in moving this technology to the point-of-care (POC) arena.
Collapse
Affiliation(s)
| | | | | | - Marc D. Porter
- Nano Institute of Utah
- University of Utah
- Salt Lake City
- USA
- Department of Chemistry
| |
Collapse
|
40
|
Antibody-Based Assays for Phenotyping of Extracellular Vesicles. BIOMED RESEARCH INTERNATIONAL 2015; 2015:524817. [PMID: 26770974 PMCID: PMC4681819 DOI: 10.1155/2015/524817] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/22/2015] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of membrane-enclosed vesicles. EVs are recognized as important players in cell-to-cell communication and are described to be involved in numerous biological and pathological processes. The fact that EVs are involved in the development and progression of several diseases has formed the basis for the use of EV analysis in a clinical setting. As the interest in EVs has increased immensely, multiple techniques have been developed aiming at characterizing these vesicles. These techniques characterize different features of EVs, like the size distribution, enumeration, protein composition, and the intravesicular cargo (e.g., RNA). This review focuses on techniques that exploit the specificity and sensitivity associated with antibody-based assays to characterize the protein phenotype of EVs. The protein phenotype of EVs can provide information on the functionality of the vesicles and may be used for identification of disease-related biomarkers. Thus, protein profiling of EVs holds great diagnostic and prognostic potential.
Collapse
|
41
|
Fredolini C, Byström S, Pin E, Edfors F, Tamburro D, Iglesias MJ, Häggmark A, Hong MG, Uhlen M, Nilsson P, Schwenk JM. Immunocapture strategies in translational proteomics. Expert Rev Proteomics 2015; 13:83-98. [PMID: 26558424 PMCID: PMC4732419 DOI: 10.1586/14789450.2016.1111141] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aiming at clinical studies of human diseases, antibody-assisted assays have been applied to biomarker discovery and toward a streamlined translation from patient profiling to assays supporting personalized treatments. In recent years, integrated strategies to couple and combine antibodies with mass spectrometry-based proteomic efforts have emerged, allowing for novel possibilities in basic and clinical research. Described in this review are some of the field's current and emerging immunocapture approaches from an affinity proteomics perspective. Discussed are some of their advantages, pitfalls and opportunities for the next phase in clinical and translational proteomics.
Collapse
Affiliation(s)
- Claudia Fredolini
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Sanna Byström
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Elisa Pin
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Fredrik Edfors
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Davide Tamburro
- Department of Oncology-Pathology, Clinical Proteomics Mass Spectrometry, SciLifeLab, Karolinska Institutet, Solna, Sweden
| | - Maria Jesus Iglesias
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Anna Häggmark
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Mun-Gwan Hong
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Mathias Uhlen
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Peter Nilsson
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| | - Jochen M Schwenk
- Affinity Proteomics, SciLifeLab, School of Biotechnology, KTH - Royal Institute of Technology, Solna, Sweden
| |
Collapse
|
42
|
Bergeron S, Laforte V, Lo PS, Li H, Juncker D. Evaluating mixtures of 14 hygroscopic additives to improve antibody microarray performance. Anal Bioanal Chem 2015; 407:8451-62. [PMID: 26345442 DOI: 10.1007/s00216-015-8992-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 08/17/2015] [Accepted: 08/19/2015] [Indexed: 11/27/2022]
Abstract
Microarrays allow the miniaturization and multiplexing of biological assays while only requiring minute amounts of samples. As a consequence of the small volumes used for spotting and the assays, evaporation often deteriorates the quality, reproducibility of spots, and the overall assay performance. Glycerol is commonly added to antibody microarray printing buffers to decrease evaporation; however, it often decreases the binding of antibodies to the surface, thereby negatively affecting assay sensitivity. Here, combinations of 14 hygroscopic chemicals were used as additives to printing buffers for contact-printed antibody microarrays on four different surface chemistries. The ability of the additives to suppress evaporation was quantified by measuring the residual buffer volume in open quill pins over time. The seven best additives were then printed either individually or as a 1:1 mixture of two additives, and the homogeneity, intensity, and reproducibility of both the spotted protein and of a fluorescently labeled analyte in an assay were quantified. Among the 28 combinations on the four slides, many were found to outperform glycerol, and the best additive mixtures were further evaluated by changing the ratio of the two additives. We observed that the optimal additive mixture was dependent on the slide chemistry, and that it was possible to increase the binding of antibodies to the surface threefold compared to 50 % glycerol, while decreasing whole-slide coefficient of variation to 5.9 %. For the two best slides, improvements were made for both the limit of detection (1.6× and 5.9×, respectively) and the quantification range (1.2× and 2.1×, respectively). The additive mixtures identified here thus help improve assay reproducibility and performance, and might be beneficial to all types of microarrays that suffer from evaporation of the printing buffers.
Collapse
Affiliation(s)
- Sébastien Bergeron
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec, Canada, H3A 2B4
| | - Veronique Laforte
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec, Canada, H3A 2B4
- Neurology and Neurosurgery Department, McGill University, 3801 University Street, Montreal, Quebec, Canada, H3A 2B4
| | - Pik-Shan Lo
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec, Canada, H3A 2B4
| | - Huiyan Li
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec, Canada, H3A 2B4
| | - David Juncker
- McGill University & Genome Quebec Innovation Centre, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1.
- Biomedical Engineering Department, McGill University, 3775 University Street, Montreal, Quebec, Canada, H3A 2B4.
- Neurology and Neurosurgery Department, McGill University, 3801 University Street, Montreal, Quebec, Canada, H3A 2B4.
| |
Collapse
|
43
|
Li H, Munzar JD, Ng A, Juncker D. A versatile snap chip for high-density sub-nanoliter chip-to-chip reagent transfer. Sci Rep 2015; 5:11688. [PMID: 26148566 PMCID: PMC4493572 DOI: 10.1038/srep11688] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/05/2015] [Indexed: 01/15/2023] Open
Abstract
The coordinated delivery of minute amounts of different reagents is important for microfluidics and microarrays, but is dependent on advanced equipment such as microarrayers. Previously, we developed the snap chip for the direct transfer of reagents, thus realizing fluidic operations by only manipulating microscope slides. However, owing to the misalignment between arrays spotted on different slides, millimeter spacing was needed between spots and the array density was limited. In this work, we have developed a novel double transfer method and have transferred 625 spots cm(-2), corresponding to >10000 spots for a standard microscope slide. A user-friendly snapping system was manufactured to make liquid handling straightforward. Misalignment, which for direct transfer ranged from 150-250 μm, was reduced to <40 μm for double transfer. The snap chip was used to quantify 50 proteins in 16 samples simultaneously, yielding limits of detection in the pg/mL range for 35 proteins. The versatility of the snap chip is illustrated with a 4-plex homogenous enzyme inhibition assay analyzing 128 conditions with precise timing. The versatility and high density of the snap chip with double transfer allows for the development of high throughput reagent transfer protocols compatible with a variety of applications.
Collapse
Affiliation(s)
- Huiyan Li
- Biomedical Engineering Department, McGill University, Montréal, QC, H3A 0G1, Canada
- McGill University and Genome Quebec Innovation Centre, McGill University, Montréal, QC, H3A 0G1, Canada
| | - Jeffrey D. Munzar
- Biomedical Engineering Department, McGill University, Montréal, QC, H3A 0G1, Canada
- McGill University and Genome Quebec Innovation Centre, McGill University, Montréal, QC, H3A 0G1, Canada
| | - Andy Ng
- Biomedical Engineering Department, McGill University, Montréal, QC, H3A 0G1, Canada
- McGill University and Genome Quebec Innovation Centre, McGill University, Montréal, QC, H3A 0G1, Canada
| | - David Juncker
- Biomedical Engineering Department, McGill University, Montréal, QC, H3A 0G1, Canada
- McGill University and Genome Quebec Innovation Centre, McGill University, Montréal, QC, H3A 0G1, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, H3A 0G1, Canada
| |
Collapse
|
44
|
Yeat NC, Lin C, Sager M, Lin J. Cancer proteomics: developments in technology, clinical use and commercialization. Expert Rev Proteomics 2015; 12:391-405. [PMID: 26145529 DOI: 10.1586/14789450.2015.1051969] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last two decades, advances in genomic, transcriptomic and proteomic methods have enabled us to identify and classify cancers by their molecular profiles. Many anticipate that a molecular taxonomy of cancer will not only lead to more effective subtyping of cancers but also earlier diagnoses, more informative prognoses and more targeted treatments. This article reviews recent technological developments in the field of proteomics, recent discoveries in proteomic cancer biomarker research and trends in clinical use. Readers are also informed of examples of successful commercialization, and the future of proteomics in cancer diagnostics.
Collapse
Affiliation(s)
- Nai Chien Yeat
- Rare Genomics Institute, 4100 Forest Park, St. Louis, MO 63108, USA
| | | | | | | |
Collapse
|
45
|
Tighe PJ, Ryder RR, Todd I, Fairclough LC. ELISA in the multiplex era: potentials and pitfalls. Proteomics Clin Appl 2015; 9:406-22. [PMID: 25644123 PMCID: PMC6680274 DOI: 10.1002/prca.201400130] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/08/2014] [Accepted: 01/19/2015] [Indexed: 12/20/2022]
Abstract
Multiplex immunoassays confer several advantages over widely adopted singleplex immunoassays including increased efficiency at a reduced expense, greater output per sample volume ratios and higher throughput predicating more resolute, detailed diagnostics and facilitating personalised medicine. Nonetheless, to date, relatively few protein multiplex immunoassays have been validated for in vitro diagnostics in clinical/point-of-care settings. This review article will outline the challenges, which must be ameliorated prior to the widespread integration of multiplex immunoassays in clinical settings: (i) biomarker validation; (ii) standardisation of immunoassay design and quality control (calibration and quantification); (iii) availability, stability, specificity and cross-reactivity of reagents; (iv) assay automation and the use of validated algorithms for transformation of raw data into diagnostic results. A compendium of multiplex immunoassays applicable to in vitro diagnostics and a summary of the diagnostic products currently available commercially are included, along with an analysis of the relative states of development for each format (namely planar slide based, suspension and planar/microtitre plate based) with respect to the aforementioned issues.
Collapse
Affiliation(s)
- Patrick J Tighe
- School of Life Sciences, The University of Nottingham, Nottingham, UK
| | | | | | | |
Collapse
|
46
|
Zhou G, Bergeron S, Juncker D. High-Performance Low-Cost Antibody Microarrays Using Enzyme-Mediated Silver Amplification. J Proteome Res 2015; 14:1872-9. [DOI: 10.1021/pr501259e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Gina Zhou
- Department
of Biomedical Engineering, McGill University, Room 316, Duff Medical Building,
3775, rue University, Montréal, QC H3A 2B4, Canada
- McGill University Genome Quebec Innovation Centre, 740 Dr. Penfield Avenue, Room 7104, Montreal, Québec H3A 0G1, Canada
| | - Sebastien Bergeron
- Department
of Biomedical Engineering, McGill University, Room 316, Duff Medical Building,
3775, rue University, Montréal, QC H3A 2B4, Canada
- McGill University Genome Quebec Innovation Centre, 740 Dr. Penfield Avenue, Room 7104, Montreal, Québec H3A 0G1, Canada
| | - David Juncker
- Department
of Biomedical Engineering, McGill University, Room 316, Duff Medical Building,
3775, rue University, Montréal, QC H3A 2B4, Canada
- McGill University Genome Quebec Innovation Centre, 740 Dr. Penfield Avenue, Room 7104, Montreal, Québec H3A 0G1, Canada
- Department
of Neurology and Neurosurgery, McGill University, 3801 University Street, Montreal, Québec H3A 2B4, Canada
| |
Collapse
|
47
|
Li H, Bergeron S, Annis MG, Siegel PM, Juncker D. Serial analysis of 38 proteins during the progression of human breast tumor in mice using an antibody colocalization microarray. Mol Cell Proteomics 2015; 14:1024-37. [PMID: 25680959 PMCID: PMC4390249 DOI: 10.1074/mcp.m114.046516] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Indexed: 01/20/2023] Open
Abstract
Proteins in serum or plasma hold great potential for use in disease diagnosis and monitoring. However, the correlation between tumor burden and protein biomarker concentration has not been established. Here, using an antibody colocalization microarray, the protein concentration in serum was measured and compared with the size of mammary xenograft tumors in 11 individual mice from the time of injection; seven blood samples were collected from each tumor-bearing mouse as well as control mice on a weekly basis. The profiles of 38 proteins detected in sera from these animals were analyzed by clustering, and we identified 10 proteins with the greatest relative increase in serum concentration that correlated with growth of the primary mammary tumor. To evaluate the diagnosis of cancer based on these proteins using either an absolute threshold (i.e. a concentration cutoff) or self-referenced differential threshold based on the increase in concentration before cell injection, receiver operating characteristic curves were produced for 10 proteins with increased concentration, and the area under curve was calculated for each time point based on a single protein or on a panel of proteins, in each case showing a rapid increase of the area under curve. Next, the sensitivity and specificity of individual and optimal protein panels were calculated, showing high accuracy as early as week 2. These results provide a foundation for studies of tumor growth through measuring serial changes of protein concentration in animal models.
Collapse
Affiliation(s)
- Huiyan Li
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre
| | - Sébastien Bergeron
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre
| | | | - Peter M Siegel
- ‖Rosalind and Morris Goodman Cancer Research Centre, and
| | - David Juncker
- From the ‡Biomedical Engineering Department, §McGill University and Genome Quebec Innovation Centre, **Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec H3A 0G1, Canada
| |
Collapse
|
48
|
Volpetti F, Garcia-Cordero J, Maerkl SJ. A microfluidic platform for high-throughput multiplexed protein quantitation. PLoS One 2015; 10:e0117744. [PMID: 25680117 PMCID: PMC4334502 DOI: 10.1371/journal.pone.0117744] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 12/31/2014] [Indexed: 11/18/2022] Open
Abstract
We present a high-throughput microfluidic platform capable of quantitating up to 384 biomarkers in 4 distinct samples by immunoassay. The microfluidic device contains 384 unit cells, which can be individually programmed with pairs of capture and detection antibody. Samples are quantitated in each unit cell by four independent MITOMI detection areas, allowing four samples to be analyzed in parallel for a total of 1,536 assays per device. We show that the device can be pre-assembled and stored for weeks at elevated temperature and we performed proof-of-concept experiments simultaneously quantitating IL-6, IL-1β, TNF-α, PSA, and GFP. Finally, we show that the platform can be used to identify functional antibody combinations by screening 64 antibody combinations requiring up to 384 unique assays per device.
Collapse
Affiliation(s)
- Francesca Volpetti
- Institute of Bioengineering, School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Jose Garcia-Cordero
- Institute of Bioengineering, School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Sebastian J. Maerkl
- Institute of Bioengineering, School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Al-Ameen MA, Li J, Beer DG, Ghosh G. Sensitive, quantitative, and high-throughput detection of angiogenic markers using shape-coded hydrogel microparticles. Analyst 2015; 140:4530-9. [DOI: 10.1039/c5an00358j] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Demonstration of the application of shape coded hydrogel microparticles for multiplexed detection of angiogenic molecules. Utilization of single fluorophore eliminates the spectral overlap associated with microparticle based multiplexed analysis.
Collapse
Affiliation(s)
- Mohammad Ali Al-Ameen
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| | - Ji Li
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| | - David G. Beer
- Thoracic Surgery
- Department of Surgery
- University of Michigan Comprehensive Cancer Center
- Ann Arbor MI 48109
- USA
| | - Gargi Ghosh
- Bioengineering Program
- Department of Mechanical Engineering
- University of Michigan
- Dearborn
- USA
| |
Collapse
|
50
|
Blanco Y, Rivas LA, García-Moyano A, Aguirre J, Cruz-Gil P, Palacín A, van Heerden E, Parro V. Deciphering the prokaryotic community and metabolisms in South African deep-mine biofilms through antibody microarrays and graph theory. PLoS One 2014; 9:e114180. [PMID: 25531640 PMCID: PMC4273990 DOI: 10.1371/journal.pone.0114180] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/05/2014] [Indexed: 11/18/2022] Open
Abstract
In the South African deep mines, a variety of biofilms growing in mine corridor walls as water seeps from intersections or from fractures represents excellent proxies for deep-subsurface environments. However, they may be greatly affected by the oxygen inputs through the galleries of mining activities. As a consequence, the interaction between the anaerobic water coming out from the walls with the oxygen inputs creates new conditions that support rich microbial communities. The inherent difficulties for sampling these delicate habitats, together with transport and storage conditions may alter the community features and composition. Therefore, the development of in situ monitoring methods would be desirable for quick evaluation of the microbial community. In this work, we report the usefulness of an antibody-microarray (EMChip66) immunoassay for a quick check of the microbial diversity of biofilms located at 1.3 km below surface within the Beatrix deep gold mine (South Africa). In addition, a deconvolution method, previously described and used for environmental monitoring, based on graph theory and applied on antibody cross-reactivity was used to interpret the immunoassay results. The results were corroborated and further expanded by 16S rRNA gene sequencing analysis. Both culture-independent techniques coincided in detecting features related to aerobic sulfur-oxidizers, aerobic chemoorganotrophic Alphaproteobacteria and metanotrophic Gammaproteobacteria. 16S rRNA gene sequencing detected phylotypes related to nitrate-reducers and anaerobic sulfur-oxidizers, whereas the EMChip66 detected immunological features from methanogens and sulfate-reducers. The results reveal a diverse microbial community with syntrophic metabolisms both anaerobic (fermentation, methanogenesis, sulphate and nitrate reduction) and aerobic (methanotrophy, sulphur oxidation). The presence of oxygen-scavenging microbes might indicate that the system is modified by the artificial oxygen inputs from the mine galleries.
Collapse
Affiliation(s)
- Yolanda Blanco
- Department of Molecular Evolution, Centro de Astrobiología (INTA-CSIC), Carretera de Ajalvir, km 4, Torrejón de Ardoz, 28850, Madrid, Spain
| | - Luis A. Rivas
- Department of Molecular Evolution, Centro de Astrobiología (INTA-CSIC), Carretera de Ajalvir, km 4, Torrejón de Ardoz, 28850, Madrid, Spain
| | - Antonio García-Moyano
- TIA/UFS Metagenomics Platform, Department of Biotechnology, University of the Free State, P. O. Box 339, Bloemfontein, 9300, South Africa
| | - Jacobo Aguirre
- Department of Molecular Evolution, Centro de Astrobiología (INTA-CSIC), Carretera de Ajalvir, km 4, Torrejón de Ardoz, 28850, Madrid, Spain
| | - Patricia Cruz-Gil
- Department of Molecular Evolution, Centro de Astrobiología (INTA-CSIC), Carretera de Ajalvir, km 4, Torrejón de Ardoz, 28850, Madrid, Spain
| | - Arantxa Palacín
- Centro de Biotecnología y Genómica de Plantas, Campus de Montegancedo, Autopista M40, km 38, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Esta van Heerden
- TIA/UFS Metagenomics Platform, Department of Biotechnology, University of the Free State, P. O. Box 339, Bloemfontein, 9300, South Africa
| | - Víctor Parro
- Department of Molecular Evolution, Centro de Astrobiología (INTA-CSIC), Carretera de Ajalvir, km 4, Torrejón de Ardoz, 28850, Madrid, Spain
- * E-mail:
| |
Collapse
|