1
|
Amiri A, Abedanzadeh S, Davaeil B, Shaabani A, Moosavi-Movahedi AA. Protein click chemistry and its potential for medical applications. Q Rev Biophys 2024; 57:e6. [PMID: 38619322 DOI: 10.1017/s0033583524000027] [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: 04/16/2024]
Abstract
A revolution in chemical biology occurred with the introduction of click chemistry. Click chemistry plays an important role in protein chemistry modifications, providing specific, sensitive, rapid, and easy-to-handle methods. Under physiological conditions, click chemistry often overlaps with bioorthogonal chemistry, defined as reactions that occur rapidly and selectively without interfering with biological processes. Click chemistry is used for the posttranslational modification of proteins based on covalent bond formations. With the contribution of click reactions, selective modification of proteins would be developed, representing an alternative to other technologies in preparing new proteins or enzymes for studying specific protein functions in different biological processes. Click-modified proteins have potential in diverse applications such as imaging, labeling, sensing, drug design, and enzyme technology. Due to the promising role of proteins in disease diagnosis and therapy, this review aims to highlight the growing applications of click strategies in protein chemistry over the last two decades, with a special emphasis on medicinal applications.
Collapse
Affiliation(s)
- Ahmad Amiri
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | | - Bagher Davaeil
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ahmad Shaabani
- Department of Chemistry, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
2
|
Abe K, Beer JC, Nguyen T, Ariyapala IS, Holmes TH, Feng W, Zhang B, Kuo D, Luo Y, Ma XJ, Maecker HT. Cross-Platform Comparison of Highly Sensitive Immunoassays for Inflammatory Markers in a COVID-19 Cohort. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1244-1253. [PMID: 38334457 PMCID: PMC10948291 DOI: 10.4049/jimmunol.2300729] [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] [Received: 10/31/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
A variety of commercial platforms are available for the simultaneous detection of multiple cytokines and associated proteins, often employing Ab pairs to capture and detect target proteins. In this study, we comprehensively evaluated the performance of three distinct platforms: the fluorescent bead-based Luminex assay, the proximity extension-based Olink assay, and a novel proximity ligation assay platform known as Alamar NULISAseq. These assessments were conducted on human serum samples from the National Institutes of Health IMPACC study, with a focus on three essential performance metrics: detectability, correlation, and differential expression. Our results reveal several key findings. First, the Alamar platform demonstrated the highest overall detectability, followed by Olink and then Luminex. Second, the correlation of protein measurements between the Alamar and Olink platforms tended to be stronger than the correlation of either of these platforms with Luminex. Third, we observed that detectability differences across the platforms often translated to differences in differential expression findings, although high detectability did not guarantee the ability to identify meaningful biological differences. Our study provides valuable insights into the comparative performance of these assays, enhancing our understanding of their strengths and limitations when assessing complex biological samples, as exemplified by the sera from this COVID-19 cohort.
Collapse
Affiliation(s)
- Koji Abe
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Tran Nguyen
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Tyson H. Holmes
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Wei Feng
- Alamar Biosciences, Inc., Fremont, CA 94538
| | | | - Dwight Kuo
- Alamar Biosciences, Inc., Fremont, CA 94538
| | - Yuling Luo
- Alamar Biosciences, Inc., Fremont, CA 94538
| | | | - Holden T. Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
3
|
Abe K, Beer JC, Nguyen T, Ariyapala IS, Holmes TH, Feng W, Zhang B, Kuo D, Luo Y, Ma XJ, Maecker HT. Cross-platform comparison of highly-sensitive immunoassays for inflammatory markers in a COVID-19 cohort 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563866. [PMID: 37961126 PMCID: PMC10634816 DOI: 10.1101/2023.10.24.563866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
A variety of commercial platforms are available for the simultaneous detection of multiple cytokines and associated proteins, often employing antibody pairs to capture and detect target proteins. In this study, we comprehensively evaluated the performance of three distinct platforms: the fluorescent bead-based Luminex assay, the proximity extension-based Olink assay, and a novel proximity ligation assay platform known as Alamar NULISAseq. These assessments were conducted on serum samples from the NIH IMPACC study, with a focus on three essential performance metrics: detectability, correlation, and differential expression. Our results reveal several key findings. Firstly, the Alamar platform demonstrated the highest overall detectability, followed by Olink and then Luminex. Secondly, the correlation of protein measurements between the Alamar and Olink platforms tended to be stronger than the correlation of either of these platforms with Luminex. Thirdly, we observed that detectability differences across the platforms often translated to differences in differential expression findings, although high detectability did not guarantee the ability to identify meaningful biological differences. Our study provides valuable insights into the comparative performance of these assays, enhancing our understanding of their strengths and limitations when assessing complex biological samples, as exemplified by the sera from this COVID-19 cohort.
Collapse
|
4
|
Krainer G, Saar KL, Arter WE, Welsh TJ, Czekalska MA, Jacquat RPB, Peter Q, Traberg WC, Pujari A, Jayaram AK, Challa P, Taylor CG, van der Linden LM, Franzmann T, Owens RM, Alberti S, Klenerman D, Knowles TPJ. Direct digital sensing of protein biomarkers in solution. Nat Commun 2023; 14:653. [PMID: 36746944 PMCID: PMC9902533 DOI: 10.1038/s41467-023-35792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/03/2023] [Indexed: 02/08/2023] Open
Abstract
The detection of proteins is of central importance to biomolecular analysis and diagnostics. Typical immunosensing assays rely on surface-capture of target molecules, but this constraint can limit specificity, sensitivity, and the ability to obtain information beyond simple concentration measurements. Here we present a surface-free, single-molecule microfluidic sensing platform for direct digital protein biomarker detection in solution, termed digital immunosensor assay (DigitISA). DigitISA is based on microchip electrophoretic separation combined with single-molecule detection and enables absolute number/concentration quantification of proteins in a single, solution-phase step. Applying DigitISA to a range of targets including amyloid aggregates, exosomes, and biomolecular condensates, we demonstrate that the assay provides information beyond stoichiometric interactions, and enables characterization of immunochemistry, binding affinity, and protein biomarker abundance. Taken together, our results suggest a experimental paradigm for the sensing of protein biomarkers, which enables analyses of targets that are challenging to address using conventional immunosensing approaches.
Collapse
Affiliation(s)
- Georg Krainer
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Kadi L Saar
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - William E Arter
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Timothy J Welsh
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Magdalena A Czekalska
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Fluidic Analytics Limited, Unit A The Paddocks Business Centre, Cherry Hinton Road, Cambridge, CB1 8DH, UK
| | - Raphaël P B Jacquat
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Quentin Peter
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Walther C Traberg
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Arvind Pujari
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Akhila K Jayaram
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Pavankumar Challa
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Christopher G Taylor
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Lize-Mari van der Linden
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47/49, Dresden, Germany
| | - Titus Franzmann
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47/49, Dresden, Germany
| | - Roisin M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Simon Alberti
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Tatzberg 47/49, Dresden, Germany
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. .,Cavendish Laboratory, Department of Physics, University of Cambridge, J J Thomson Ave, Cambridge, CB3 0HE, UK.
| |
Collapse
|
5
|
Makhneva E, Sklenárová D, Brandmeier JC, Hlaváček A, Gorris HH, Skládal P, Farka Z. Influence of Label and Solid Support on the Performance of Heterogeneous Immunoassays. Anal Chem 2022; 94:16376-16383. [DOI: 10.1021/acs.analchem.2c03543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Ekaterina Makhneva
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Dorota Sklenárová
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- CEITEC MU, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Julian C. Brandmeier
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93053 Regensburg, Germany
| | - Antonín Hlaváček
- Institute of Analytical Chemistry of the Czech Academy of Sciences, Veveří 967, 602 00 Brno, Czech Republic
| | - Hans H. Gorris
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Petr Skládal
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- CEITEC MU, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Zdeněk Farka
- Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
- CEITEC MU, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
6
|
Hofmann A, Saft B, Reich P, Grabmann M, Glaser G, Trubenbach M, Rolapp A, Reinhard M, Scholz F, Schafer E. Lock-In Pixel CMOS Image Sensor for Time-Resolved Fluorescence Readout of Lateral-Flow Assays. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2022; 16:535-544. [PMID: 35862324 DOI: 10.1109/tbcas.2022.3192926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
We present a CMOS image sensor (CIS) based time-resolved fluorescence (TRF) measurement system for filter-less, highly sensitive readout of lateral-flow assay (LFA) test strips. The CIS contains a 256 × 128 lock-in pixel (LIP) sensor array. Each pixel has a size of 10 μm × 10 μm and includes a photodiode acting as signal transducer. The LIP CIS was designed in a standard 0.18 μ m CMOS technology specifically for TRF applications. The LIP architecture blocks interfering light when fluorophores are excited and accumulates the emitted fluorescence light to be measured over multiple cycles after excitation. This allows to detect even small amounts of fluorescence light over a wide analyte concentration range. The LIP CIS based TRF reader was characterized in terms of reproducible and uniform signal intensities with use of appropriate Europium(III) [Eu 3+] chelate particles as fluorescence standards. We measured different concentrations of Eu-based nanoparticles (NP) on test strips with the TRF reader. The sensor system shows 5.1 orders of magnitude of detection dynamic range (DDR) with a limit of detection (LoD) of [Formula: see text]. In addition, using human C-reactive protein (hCRP) as a model analyte, we compared the developed TRF reader with a commercial colorimetric LFA reader. For the quantification of CRP, the LIP CIS based TRF reader demonstrates a DDR of 3.6 orders of magnitude with an excellent LoD of [Formula: see text], which is 14 times better than the LoD of the commercial LFA reader.
Collapse
|
7
|
Lee JE, Dan K, Kim HJ, Kim YM, Park KH. Plasma proteomic analysis to identify potential biomarkers of histologic chorioamnionitis in women with preterm premature rupture of membranes. PLoS One 2022; 17:e0270884. [PMID: 35797368 PMCID: PMC9262229 DOI: 10.1371/journal.pone.0270884] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 06/18/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction
To identify potential biomarkers in the plasma that could predict histologic chorioamnionitis (HCA) in women with preterm premature rupture of membranes (PPROM), using shotgun and targeted proteomic analyses.
Methods
This retrospective cohort study included 78 singleton pregnant women with PPROM (24–34 gestational weeks) who delivered within 96 h of blood sampling. Maternal plasma samples were analyzed by label-free liquid chromatography-tandem mass spectrometry for proteome profiling in a nested case-control study design (HCA cases vs. non-HCA controls [n = 9 each]). Differential expression of 12 candidate proteins was assessed by multiple reaction monitoring-mass spectrometry (MRM-MS) analysis in individual plasma samples from cases and controls matched by gestational age at sampling (n = 40, cohort 1). A validation study was further performed in an independent study group (n = 38, cohort 2) using ELISA and turbidimetric immunoassay for three differentially expressed proteins.
Results
Shotgun proteomics analyses yielded 18 proteins that were differentially expressed (P < 0.05) between HCA cases and non-HCA controls. MRM-MS analysis of 12 differentially expressed proteins further revealed that the CRP, C4A, and SAA4 levels were significantly increased in women with HCA. A multi-marker panel comprising plasma SAA4 and C4A showed enhanced potential for differentiating HCA from non-HCA women (area under the curve = 0.899). Additional validation of these findings by ELISA assays revealed that the CRP levels were significantly higher in women with HCA than in those without HCA, whereas the plasma levels of C4A and SAA4 did not significantly differ between the two groups.
Conclusions
Plasma C4A, SAA4, and CRP were identified as potential biomarkers for detecting HCA in women with PPROM, based on targeted and shotgun proteomic analyses, showing good accuracy when used as a combined dual-biomarker panel (C4A and SAA4). Nevertheless, ELISA validation of these proteins, except for CRP, may not yield clinically useful markers for predicting HCA.
Collapse
Affiliation(s)
- Ji Eun Lee
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea
| | - Kisoon Dan
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyeon Ji Kim
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Mi Kim
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyo Hoon Park
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail:
| |
Collapse
|
8
|
Stewart-McGuinness C, Platt CI, Ozols M, Goh B, Griffiths TW, Sherratt MJ. Defining the Protease and Protease Inhibitor (P/PI) Proteomes of Healthy and Diseased Human Skin by Modified Systematic Review. Biomolecules 2022; 12:475. [PMID: 35327667 PMCID: PMC8946613 DOI: 10.3390/biom12030475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/09/2022] [Accepted: 03/18/2022] [Indexed: 12/26/2022] Open
Abstract
Proteases and protease inhibitors (P/PIs) are involved in many biological processes in human skin, yet often only specific families or related groups of P/PIs are investigated. Proteomics approaches, such as mass spectrometry, can define proteome signatures (including P/PIs) in tissues; however, they struggle to detect low-abundance proteins. To overcome these issues, we aimed to produce a comprehensive proteome of all P/PIs present in normal and diseased human skin, in vivo, by carrying out a modified systematic review using a list of P/PIs from MEROPS and combining this with key search terms in Web of Science. Resulting articles were manually reviewed against inclusion/exclusion criteria and a dataset constructed. This study identified 111 proteases and 77 protease inhibitors in human skin, comprising the serine, metallo-, cysteine and aspartic acid catalytic families of proteases. P/PIs showing no evidence of catalytic activity or protease inhibition, were designated non-peptidase homologs (NPH), and no reported protease inhibitory activity (NRPIA), respectively. MMP9 and TIMP1 were the most frequently published P/PIs and were reported in normal skin and most skin disease groups. Normal skin and diseased skin showed significant overlap with respect to P/PI profile; however, MMP23 was identified in several skin disease groups, but was absent in normal skin. The catalytic profile of P/PIs in wounds, scars and solar elastosis was distinct from normal skin, suggesting that a different group of P/PIs is responsible for disease progression. In conclusion, this study uses a novel approach to provide a comprehensive inventory of P/PIs in normal and diseased human skin reported in our database. The database may be used to determine either which P/PIs are present in specific diseases or which diseases individual P/PIs may influence.
Collapse
Affiliation(s)
- Callum Stewart-McGuinness
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (C.S.-M.); (M.O.); (B.G.); (M.J.S.)
| | - Christopher I. Platt
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (C.S.-M.); (M.O.); (B.G.); (M.J.S.)
| | - Matiss Ozols
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (C.S.-M.); (M.O.); (B.G.); (M.J.S.)
- Department of Human Genetics, Wellcome Sanger Institute, Genome Campus, Hinxton CB10 1SA, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Brian Goh
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (C.S.-M.); (M.O.); (B.G.); (M.J.S.)
| | - Tamara W. Griffiths
- Centre for Dermatology Research, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PL, UK;
| | - Michael J. Sherratt
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK; (C.S.-M.); (M.O.); (B.G.); (M.J.S.)
| |
Collapse
|
9
|
Mc Ardle A, Kwasnik A, Szenpetery A, Hernandez B, Parnell A, de Jager W, de Roock S, FitzGerald O, Pennington SR. Identification and Evaluation of Serum Protein Biomarkers Which Differentiate Psoriatic from Rheumatoid Arthritis. Arthritis Rheumatol 2021; 74:81-91. [PMID: 34114357 DOI: 10.1002/art.41899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/08/2021] [Indexed: 11/10/2022]
Abstract
OBJECTIVES To identify serum protein biomarkers which might separate early inflammatory arthritis (EIA) patients with psoriatic arthritis (PsA) from those with rheumatoid arthritis (RA) and may be used to support appropriate early intervention. METHODS The serum proteome of patients with PsA and RA was interrogated using nano-flow liquid chromatography mass spectrometry (nLC-MS/MS) (n=64 patients), an aptamer-based assay (SOMAscan) targeting 1,129 proteins (n=36 patients) and a multiplexed antibody assay (Luminex) for 48 proteins (n=64 patients). Multiple reaction monitoring assays (MRM) were developed to evaluate the performance of putative markers using the discovery cohort (n=60) and subsequently an independent cohort of PsA and RA patients (n=167). RESULTS Multivariate machine learning analysis of the protein discovery data from the three platforms revealed that it was possible to discriminate PsA from RA patients with an area under the curve (AUC) of 0.94 for nLC-MS/MS, 0.69 for bead based immunoassay measurements and 0.73 for aptamer based analysis. Subsequently in the separate verification and evaluation studies, random forest models revealed that a subset of proteins measured by MRM could differentiate PsA and RA patients with AUCs of 0.79 and 0.85 respectively. CONCLUSION We report a serum protein biomarker panel which can separate EIA patients with PsA from those with RA. With continued evaluation and refinement using additional and larger patient cohorts including those with other arthropathies we suggest the panel identified here could contribute toward improved clinical decision making.
Collapse
Affiliation(s)
- Angela Mc Ardle
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Anna Kwasnik
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Agnes Szenpetery
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Belinda Hernandez
- School of Medical Gerontology, TILDA (The Irish Longitudinal Study on Aging), Trinity College Dublin, Ireland.,School of Mathematics and Statistics, University College Dublin, Ireland
| | - Andrew Parnell
- School of Mathematics and Statistics, University College Dublin, Ireland
| | - Wilco de Jager
- Department of Paediatric Immunology, Laboratory of Translation Immunology LTI, Wilhelmina Children Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.,Multiplex Core Facility, Laboratory of Translational Immunology LTI, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sytze de Roock
- Multiplex Core Facility, Laboratory of Translational Immunology LTI, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Oliver FitzGerald
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| | - Stephen R Pennington
- UCD Conway Institute of Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Ireland
| |
Collapse
|
10
|
Frutiger A, Tanno A, Hwu S, Tiefenauer RF, Vörös J, Nakatsuka N. Nonspecific Binding-Fundamental Concepts and Consequences for Biosensing Applications. Chem Rev 2021; 121:8095-8160. [PMID: 34105942 DOI: 10.1021/acs.chemrev.1c00044] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nature achieves differentiation of specific and nonspecific binding in molecular interactions through precise control of biomolecules in space and time. Artificial systems such as biosensors that rely on distinguishing specific molecular binding events in a sea of nonspecific interactions have struggled to overcome this issue. Despite the numerous technological advancements in biosensor technologies, nonspecific binding has remained a critical bottleneck due to the lack of a fundamental understanding of the phenomenon. To date, the identity, cause, and influence of nonspecific binding remain topics of debate within the scientific community. In this review, we discuss the evolution of the concept of nonspecific binding over the past five decades based upon the thermodynamic, intermolecular, and structural perspectives to provide classification frameworks for biomolecular interactions. Further, we introduce various theoretical models that predict the expected behavior of biosensors in physiologically relevant environments to calculate the theoretical detection limit and to optimize sensor performance. We conclude by discussing existing practical approaches to tackle the nonspecific binding challenge in vitro for biosensing platforms and how we can both address and harness nonspecific interactions for in vivo systems.
Collapse
Affiliation(s)
- Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Alexander Tanno
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Stephanie Hwu
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Raphael F Tiefenauer
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| | - Nako Nakatsuka
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, Zürich CH-8092, Switzerland
| |
Collapse
|
11
|
Landegren U, Hammond M. Cancer diagnostics based on plasma protein biomarkers: hard times but great expectations. Mol Oncol 2021; 15:1715-1726. [PMID: 33012111 PMCID: PMC8169444 DOI: 10.1002/1878-0261.12809] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer diagnostics based on the detection of protein biomarkers in blood has promising potential for early detection and continuous monitoring of disease. However, the currently available protein biomarkers and assay formats largely fail to live up to expectations, mainly due to insufficient diagnostic specificity. Here, we discuss what kinds of plasma proteins might prove useful as biomarkers of malignant processes in specific organs. We consider the need to search for biomarkers deep down in the lowest reaches of the proteome, below current detection levels. In this regard, we comment on the poor molecular detection sensitivity of current protein assays compared to nucleic acid detection reactions, and we discuss requirements for achieving detection of vanishingly small amounts of proteins, to ensure detection of early stages of malignant growth through liquid biopsy.
Collapse
Affiliation(s)
- Ulf Landegren
- Department of Immunology, Genetics and PathologyUppsala University and SciLifeLabUppsalaSweden
| | - Maria Hammond
- Department of Immunology, Genetics and PathologyUppsala University and SciLifeLabUppsalaSweden
| |
Collapse
|
12
|
Hofmann A, Meister M, Rolapp A, Reich P, Scholz F, Schafer E. Light Absorption Measurement With a CMOS Biochip for Quantitative Immunoassay Based Point-of-Care Applications. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2021; 15:369-379. [PMID: 34033547 DOI: 10.1109/tbcas.2021.3083359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We present a CMOS biochip-based photometer for quantitative immunoassay diagnostics. The photometer quantifies the concentration of antigens based on light absorption, which allows for a low-cost implementation without expensive optical components. We propose a light controller to lower the start-up and settling time of the light source to 30 seconds, to facilitate fast measurement starts, and to decrease the overall measurement times. The application-specific integrated circuit (ASIC) contains a 6 x 7-sensor array with 100 μm x 100 μm photodiodes that serve as signal transducers. The ASIC was developed in a normal 0.35- μm CMOS technology, avoiding the need for expensive post-CMOS processes. We present our strategy for the assembly of the ASIC and the immobilization of antibodies. For its first time, we demonstrate the quantification of prostate specific antigen (PSA) with an optoelectronic CMOS biochip using this approach. A PSA immunoassay is performed on the top surface of the CMOS sensor array, enzyme kinetics and PSA concentration are measured within 6 minutes with a limit of detection (LoD) of 0.5 ng/ml, which meets clinical testing requirements. We achieve an overall coefficient of variation (CV) of 7%, which is good compared to other point-of-care (PoC) systems.
Collapse
|
13
|
David P, Hansen FJ, Bhat A, Weber GF. An overview of proteomic methods for the study of 'cytokine storms'. Expert Rev Proteomics 2021; 18:83-91. [PMID: 33849358 DOI: 10.1080/14789450.2021.1911652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: The cytokine storm is a form of excessive systemic inflammatory reaction triggered by a myriad of factors that may lead to multi-organ failure, and finally to death. The cytokine storm can occur in a number of infectious and noninfectious diseases including COVID-19, sepsis, ebola, avian influenza, and graft versus host disease, or during the severe inflammatory response syndrome.Area covered: This review mainly focuses on the most common and well-known methods of protein studies (PAGE, SDS-PAGE, and high- performance liquid chromatography). It also discusses other modern technologies in proteomics like mass spectrometry, soft ionization techniques, cytometric bead assays, and the next generation of microarrays that have been used to get an in-depth understanding of the pathomechanisms involved during the cytokine storm.Expert opinion: Overactivation of leukocytes drives the production and secretion of inflammatory cytokines fueling the cytokine storm. These events lead to a systemic hyper-inflammation, circulatory collapse and shock, and finally to multiorgan failure. Therefore, monitoring the patient's systemic cytokine levels with proteomic technologies that are redundant, economical, and require minimal sample volume for real-time assessment might help in a better clinical evaluation and management of critically ill patients.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Frederik J Hansen
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Adil Bhat
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Georg F Weber
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
14
|
Reichmuth AM, Kübrich K, Blickenstorfer Y, Frutiger A, Momotenko D, Gatterdam V, Treindl F, Fattinger C, Vörös J. Investigating Complex Samples with Molograms of Low-Affinity Binders. ACS Sens 2021; 6:1067-1076. [PMID: 33629586 DOI: 10.1021/acssensors.0c02346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In vitro diagnostics relies on the quantification of minute amounts of a specific biomolecule, called biomarker, from a biological sample. The majority of clinically relevant biomarkers for conditions beyond infectious diseases are detected by means of binding assays, where target biomarkers bind to a solid phase and are detected by biochemical or physical means. Nonspecifically bound biomolecules, the main source of variation in such assays, need to be washed away in a laborious process, restricting the development of widespread point-of-care diagnostics. Here, we show that a diffractometric assay provides a new, label-free possibility to investigate complex samples, such as blood plasma. A coherently arranged sub-micron pattern, that is, a peptide mologram, is created to demonstrate the insensitivity of this diffractometric assay to the unwanted masking effect of nonspecific interactions. In addition, using an array of low-affinity binders, we also demonstrate the feasibility of molecular profiling of blood plasma in real time and show that individual patients can be differentiated based on the binding kinetics of circulating proteins.
Collapse
Affiliation(s)
- Andreas M. Reichmuth
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Katharina Kübrich
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Yves Blickenstorfer
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Dmitry Momotenko
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Volker Gatterdam
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Fridolin Treindl
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Christof Fattinger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
15
|
Frutiger A, Fattinger C, Vörös J. Ultra-Stable Molecular Sensors by Sub-Micron Referencing and Why They Should Be Interrogated by Optical Diffraction-Part I. The Concept of a Spatial Affinity Lock-in Amplifier. SENSORS (BASEL, SWITZERLAND) 2021; 21:E469. [PMID: 33440783 PMCID: PMC7827303 DOI: 10.3390/s21020469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/30/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
Label-free optical biosensors, such as surface plasmon resonance, are sensitive and well-established for the characterization of molecular interactions. Yet, these sensors require stabilization and constant conditions even with the use of reference channels. In this paper, we use tools from signal processing to show why these sensors are so cross-sensitive and how to overcome their drawbacks. In particular, we conceptualize the spatial affinity lock-in as a universal design principle for sensitive molecular sensors even in the complete absence of stabilization. The spatial affinity lock-in is analogous to the well-established time-domain lock-in. Instead of a time-domain signal, it modulates the binding signal at a high spatial frequency to separate it from the low spatial frequency environmental noise in Fourier space. In addition, direct sampling of the locked-in sensor's response in Fourier space enabled by diffraction has advantages over sampling in real space as done by surface plasmon resonance sensors using the distributed reference principle. This paper and part II hint at the potential of spatially locked-in diffractometric biosensors to surpass state-of-the-art temperature-stabilized refractometric biosensors. Even simple, miniaturized and non-stabilized sensors might achieve the performance of bulky lab instruments. This may enable new applications in label-free analysis of molecular binding and point-of-care diagnostics.
Collapse
Affiliation(s)
- Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zürich, 8092 Zürich, Switzerland;
| | - Christof Fattinger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute of Biomedical Engineering, University and ETH Zürich, 8092 Zürich, Switzerland;
| |
Collapse
|
16
|
Chern M, Garden PM, Baer RC, Galagan JE, Dennis AM. Transcription Factor Based Small‐Molecule Sensing with a Rapid Cell Phone Enabled Fluorescent Bead Assay. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Margaret Chern
- Division of Materials Science and Engineering Boston University Boston MA USA
| | - Padric M. Garden
- Department of Biomedical Engineering Boston University Boston MA USA
| | - R C. Baer
- Department of Microbiology Boston University Boston MA USA
| | - James E. Galagan
- Department of Biomedical Engineering Boston University Boston MA USA
- Department of Microbiology Boston University Boston MA USA
- National Emerging Infectious Diseases Laboratories Boston University Boston MA USA
| | - Allison M. Dennis
- Division of Materials Science and Engineering Boston University Boston MA USA
- Department of Biomedical Engineering Boston University Boston MA USA
| |
Collapse
|
17
|
Chern M, Garden PM, Baer RC, Galagan JE, Dennis AM. Transcription Factor Based Small-Molecule Sensing with a Rapid Cell Phone Enabled Fluorescent Bead Assay. Angew Chem Int Ed Engl 2020; 59:21597-21602. [PMID: 32945589 DOI: 10.1002/anie.202007575] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 12/26/2022]
Abstract
Recently, allosteric transcription factors (TFs) were identified as a novel class of biorecognition elements for in vitro sensing, whereby an indicator of the differential binding affinity between a TF and its cognate DNA exhibits dose-dependent responsivity to an analyte. Described is a modular bead-based biosensor design that can be applied to such TF-DNA-analyte systems. DNA-functionalized beads enable efficient mixing and spatial separation, while TF-labeled semiconductor quantum dots serve as bright fluorescent indicators of the TF-DNA bound (on bead) and unbound states. The prototype sensor for derivatives of the antibiotic tetracycline exhibits nanomolar sensitivity with visual detection of bead fluorescence. Facile changes to the sensor enable sensor response tuning without necessitating changes to the biomolecular affinities. Assay components self-assemble, and readout by eye or digital camera is possible within 5 minutes of analyte addition, making sensor use facile, rapid, and instrument-free.
Collapse
Affiliation(s)
- Margaret Chern
- Division of Materials Science and Engineering, Boston University, Boston, MA, USA
| | - Padric M Garden
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - R C Baer
- Department of Microbiology, Boston University, Boston, MA, USA
| | - James E Galagan
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.,Department of Microbiology, Boston University, Boston, MA, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Allison M Dennis
- Division of Materials Science and Engineering, Boston University, Boston, MA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA
| |
Collapse
|
18
|
Luyten LJ, Dieu M, Demazy C, Fransolet M, Nawrot TS, Renard P, Debacq-Chainiaux F. Optimization of label-free nano LC-MS/MS analysis of the placental proteome. Placenta 2020; 101:159-162. [PMID: 32992125 DOI: 10.1016/j.placenta.2020.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/31/2020] [Accepted: 09/11/2020] [Indexed: 10/23/2022]
Abstract
The placenta can be regarded as a mirror of the events to which the fetus is exposed during development. The placental proteome has been studied with several methodologies differing in sample handling, protein extraction, and processing. We optimized a protocol to analyze the placental proteome by means of label-free nano-LC-MS/MS mass spectrometry with regard to sample treatment, protein extraction, and protein digestion, in order to obtain a high protein concentration for identification of a specific protein signature according to the conditions studied. We recommend mechanical tissue disruption, blood removal prior to protein extraction, and FASP-based or in-gel digestion.
Collapse
Affiliation(s)
- Leen J Luyten
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium; Centre for Environmental Sciences, Hasselt University (UHasselt), Diepenbeek, Belgium
| | - Marc Dieu
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium; MaSUN, Mass Spectrometry Facility, University of Namur (UNamur), Namur, Belgium
| | - Catherine Demazy
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium; MaSUN, Mass Spectrometry Facility, University of Namur (UNamur), Namur, Belgium
| | - Maude Fransolet
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium; MaSUN, Mass Spectrometry Facility, University of Namur (UNamur), Namur, Belgium
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University (UHasselt), Diepenbeek, Belgium; Department of Public Health & Primary Care, Occupational and Environmental Medicine, Leuven University (KULeuven), Leuven, Belgium
| | - Patricia Renard
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium; MaSUN, Mass Spectrometry Facility, University of Namur (UNamur), Namur, Belgium
| | - Florence Debacq-Chainiaux
- Unité de Recherche en Biologie Cellulaire (URBC) - Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), Namur, Belgium.
| |
Collapse
|
19
|
Migisha Ntwali P, Heo CE, Han JY, Chae SY, Kim M, Vu HM, Kim MS, Kim HI. Mass spectrometry-based proteomics of single cells and organoids: The new generation of cancer research. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.116005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Sun J, Ning X, Cui L, Ling M, Xu X, He S. Assembly of "carrier free" enzymatic nano-reporters for improved ELISA. Analyst 2020; 145:6541-6548. [PMID: 32776037 DOI: 10.1039/d0an00585a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Enzyme-linked immunosorbent assay (ELISA) is an economic and easy operation technique that has been widely used for the detection of protein in industry. However, the low loading capacity of the enzyme reporter has contributed to the low sensitivity of traditional ELISA, and the cross-linking procedures of the enzyme-labeled antibody in ELISA methods can lead to the inactivation of the enzyme, which will further decrease the sensitivity. To address this issue, herein we fabricated "carrier-free" nanoparticles to obtain a horseradish peroxidase (HRP) labelled reporter with a high HRP loading capacity. A disulphide-containing bis-N-hydroxy succinimide (NHS) crosslinker (NHS-SS-NHS) was used to control the link and release of traceless HRPs, thus without reduction of its enzymatic activity. The HRP nanoparticle (NanoHRP) was successfully applied for dot blotting and ELISA. When carcinoembryonic antigen (CEA) was used as a target, the detection limit of the NanoHRP-based ELISA was 0.005 ng mL-1, which was about 400 times more sensitive than traditional ELISA. A good correlation between the CEA concentrations and the response values measured by NanoHRP ELISA was obtained in the range of 0.005 to 1 ng mL-1. This concept could be exploited to improve ELISA tests, especially those requiring a high accuracy, to facilitate physicians in deciding the appropriate medical treatment.
Collapse
Affiliation(s)
- Jian Sun
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530021, China.
| | | | | | | | | | | |
Collapse
|
21
|
Parry S, Leite R, Esplin MS, Bukowski R, Zhang H, Varner M, Andrews WW, Saade GR, Ilekis J, Reddy UM, Huang H, Sadovsky Y, Blair IA, Biggio J. Cervicovaginal fluid proteomic analysis to identify potential biomarkers for preterm birth. Am J Obstet Gynecol 2020; 222:493.e1-493.e13. [PMID: 31758918 PMCID: PMC7196033 DOI: 10.1016/j.ajog.2019.11.1252] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Spontaneous preterm birth is a leading cause of neonatal morbidity and mortality. Early identification of at-risk women by reliable screening tests could reduce the spontaneous preterm birth rate, but conventional methods such as obstetrical history and maternal cervical length screening identify only a minority of spontaneous preterm birth cases. Cervicovaginal fluid might prove to be a useful, readily available biological fluid for identifying spontaneous preterm birth biomarkers. OBJECTIVE The objective of the study was to identify cervicovaginal fluid biomarkers of early spontaneous preterm birth in a high-risk cohort of pregnant women with a history of spontaneous preterm birth using targeted and shotgun proteomic analyses. STUDY DESIGN A nested case control study (cases were spontaneous preterm birth <34 weeks in the current pregnancy; controls were spontaneous labor and delivery at 39-41 weeks) was performed using cervicovaginal fluid samples collected at 3 study visits (100/7 to 186/7 weeks, 190/7 to 236/7 weeks, and 280/7 to 316/7 weeks). All participants had a history of at least 1 prior spontaneous preterm birth. Targeted proteomic analysis was performed using a stable isotope-labeled proteome derived from endocervical and vaginal mucosal cells. This served as a standard to quantitate candidate protein levels in individual cervicovaginal fluid samples from the second and third study visits using liquid chromatography-multiple reaction monitoring mass spectrometry. The ratio of endogenous peptide area/stable isotope-labeled proteome-derived peptide area was used to measure levels of 42 peptides in 22 proteins. To maximize biomarker discovery in the cervicovaginal fluid samples, shotgun proteomic analysis also was performed utilizing liquid chromatography and ion trap mass spectrometry. A validation study was performed in second-trimester cervicovaginal fluid samples from an independent study group (12 spontaneous preterm birth cases, 19 term delivery controls) using enzyme-linked immunosorbent assay for 5 proteins expressed at higher levels in spontaneous preterm birth cases compared with controls in targeted or shotgun proteomic analyses. RESULTS For targeted proteomics, cervicovaginal fluid samples from 33 cases and 32 controls at 190/7 to 236/7 weeks and 16 cases and 14 controls at 280/7 to 316/7 weeks from the same pregnancies were analyzed. When samples were compared between cases and controls, the relative abundance of 5 proteins was greater (P = .02-.05) in cases at both visits, while the relative abundance of 1 protein was lower (P = .03) in cases at both visits. For shotgun proteomics analyses, cervicovaginal fluid samples were pooled for 9 spontaneous preterm birth cases and 9 term delivery controls at each study visit. Shotgun proteomics yielded 28 proteins that were detected at levels >2 times higher and 1 protein that was detected at a level <0.5 times lower in spontaneous preterm birth cases compared with controls at all 3 study visits. Validation enzyme-linked immunosorbent assay for 5 proteins that were detected at higher levels in cervicovaginal fluid samples from spontaneous preterm birth cases compared with term delivery controls in proteomics analyses did not demonstrate statistically significant differences between spontaneous preterm birth cases and controls. CONCLUSIONS Potential biomarkers of spontaneous preterm birth were identified by targeted and shotgun proteomics analyses in cervicovaginal fluid samples from high-risk, asymptomatic women. Many of the proteins detected at higher levels in cervicovaginal fluid samples from spontaneous preterm birth cases are extracellular matrix proteins and/or regulate cell membrane physiology. These proteins have substantial biological interest, but validation enzyme-linked immunosorbent assay for 5 of these proteins did not yield clinically useful biomarkers for spontaneous preterm birth.
Collapse
Affiliation(s)
- Samuel Parry
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA.
| | - Rita Leite
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Radek Bukowski
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - Heping Zhang
- Collaborative Center for Statistics in Science, Yale University School of Public Health, New Haven, CT
| | - Michael Varner
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT
| | - William W Andrews
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - George R Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - John Ilekis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Uma M Reddy
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Hao Huang
- Collaborative Center for Statistics in Science, Yale University School of Public Health, New Haven, CT
| | - Yoel Sadovsky
- Magee Women's Research Institute, University of Pittsburgh, Pittsburgh, PA
| | - Ian A Blair
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Joseph Biggio
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
22
|
Kiio TM, Park S. Nano-scientific Application of Atomic Force Microscopy in Pathology: from Molecules to Tissues. Int J Med Sci 2020; 17:844-858. [PMID: 32308537 PMCID: PMC7163363 DOI: 10.7150/ijms.41805] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/26/2020] [Indexed: 12/28/2022] Open
Abstract
The advantages of atomic force microscopy (AFM) in biological research are its high imaging resolution, sensitivity, and ability to operate in physiological conditions. Over the past decades, rigorous studies have been performed to determine the potential applications of AFM techniques in disease diagnosis and prognosis. Many pathological conditions are accompanied by alterations in the morphology, adhesion properties, mechanical compliances, and molecular composition of cells and tissues. The accurate determination of such alterations can be utilized as a diagnostic and prognostic marker. Alteration in cell morphology represents changes in cell structure and membrane proteins induced by pathologic progression of diseases. Mechanical compliances are also modulated by the active rearrangements of cytoskeleton or extracellular matrix triggered by disease pathogenesis. In addition, adhesion is a critical step in the progression of many diseases including infectious and neurodegenerative diseases. Recent advances in AFM techniques have demonstrated their ability to obtain molecular composition as well as topographic information. The quantitative characterization of molecular alteration in biological specimens in terms of disease progression provides a new avenue to understand the underlying mechanisms of disease onset and progression. In this review, we have highlighted the application of diverse AFM techniques in pathological investigations.
Collapse
Affiliation(s)
| | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Daegu 42601, Republic of Korea
| |
Collapse
|
23
|
Silbereisen A, Alassiri S, Bao K, Grossmann J, Nanni P, Fernandez C, Tervahartiala T, Nascimento GG, Belibasakis GN, Heikkinen A, Lopez R, Sorsa T, Bostanci N. Label‐Free Quantitative Proteomics versus Antibody‐Based Assays to Measure Neutrophil‐Derived Enzymes in Saliva. Proteomics Clin Appl 2020; 14:e1900050. [DOI: 10.1002/prca.201900050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/20/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Angelika Silbereisen
- Section of Periodontology and Dental Prevention Division of Oral Diseases Department of Dental Medicine Karolinska Institutet Alfred Nobels allé 8 14152 Huddinge Stockholm Sweden
| | - Saeed Alassiri
- Department of Oral and Maxillofacial Diseases University of Helsinki Helsinki University Central Hospital P.O. Box 41 (Mannerheimintie 172) 00014 Helsinki Finland
| | - Kai Bao
- Section of Periodontology and Dental Prevention Division of Oral Diseases Department of Dental Medicine Karolinska Institutet Alfred Nobels allé 8 14152 Huddinge Stockholm Sweden
| | - Jonas Grossmann
- Functional Genomics Center Zurich ETH Zurich and University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Paolo Nanni
- Functional Genomics Center Zurich ETH Zurich and University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Claudia Fernandez
- Functional Genomics Center Zurich ETH Zurich and University of Zurich Winterthurerstrasse 190 8057 Zurich Switzerland
| | - Taina Tervahartiala
- Department of Oral and Maxillofacial Diseases University of Helsinki Helsinki University Central Hospital P.O. Box 41 (Mannerheimintie 172) 00014 Helsinki Finland
| | - Gustavo G. Nascimento
- Section of Periodontology Department of Dentistry and Oral Health Aarhus University Vennelyst Boulevard 9 8000 Aarhus C Denmark
| | - Georgios N. Belibasakis
- Section of Periodontology and Dental Prevention Division of Oral Diseases Department of Dental Medicine Karolinska Institutet Alfred Nobels allé 8 14152 Huddinge Stockholm Sweden
| | - Anna‐Maria Heikkinen
- Department of Oral and Maxillofacial Diseases University of Helsinki Helsinki University Central Hospital P.O. Box 41 (Mannerheimintie 172) 00014 Helsinki Finland
| | - Rodrigo Lopez
- Section of Periodontology Department of Dentistry and Oral Health Aarhus University Vennelyst Boulevard 9 8000 Aarhus C Denmark
| | - Timo Sorsa
- Section of Periodontology and Dental Prevention Division of Oral Diseases Department of Dental Medicine Karolinska Institutet Alfred Nobels allé 8 14152 Huddinge Stockholm Sweden
- Department of Oral and Maxillofacial Diseases University of Helsinki Helsinki University Central Hospital P.O. Box 41 (Mannerheimintie 172) 00014 Helsinki Finland
| | - Nagihan Bostanci
- Section of Periodontology and Dental Prevention Division of Oral Diseases Department of Dental Medicine Karolinska Institutet Alfred Nobels allé 8 14152 Huddinge Stockholm Sweden
| |
Collapse
|
24
|
Broecker F, Wegner E, Seco BMS, Kaplonek P, Bräutigam M, Ensser A, Pfister F, Daniel C, Martin CE, Mattner J, Seeberger PH. Synthetic Oligosaccharide-Based Vaccines Protect Mice from Clostridioides difficile Infections. ACS Chem Biol 2019; 14:2720-2728. [PMID: 31692324 PMCID: PMC6929054 DOI: 10.1021/acschembio.9b00642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Infections with Clostridioides difficile (formerly Clostridium difficile) have increased
in incidence, morbidity,
and mortality over the past decade. Preventing infections is becoming
increasingly important, as frontline antibiotics become less effective
and frequently induce recurrence by disrupting intestinal microbiota.
The clinically most advanced vaccine approaches prevent symptoms once C. difficile infection is established by inducing immunity
to secreted clostridial cytotoxins. However, they do not inhibit bacterial
colonization and thereby favor asymptomatic carriage. Synthetic oligosaccharides
resembling the C. difficile surface glycans PS-I,
PS-II, and PS-III are immunogenic and serve as basis for colonization-preventing
vaccines. Here, we demonstrate that glycoconjugate vaccine candidates
based on synthetic oligosaccharides protected mice from infections
with two different C. difficile strains. Four synthetic
antigens, ranging in size from disaccharides to hexasaccharides, were
conjugated to CRM197, which is a carrier protein used in
commercial vaccines. The vaccine candidates induced glycan-specific
antibodies in mice and substantially limited C. difficile colonization and colitis after experimental infection. The glycoconjugates
ameliorated intestinal pathology more substantially than a toxin-targeting
vaccine. Colonization of the gut by C. difficile was
selectively inhibited while intestinal microbiota remained preserved.
Passive transfer experiments with anti-PS-I serum revealed that protection
is mediated by specific antiglycan antibodies; however, cell-mediated
immunity likely also contributed to protection in vivo. Thus, glycoconjugate vaccines against C. difficile are a complementary approach to toxin-targeting strategies and are
advancing through preclinical work.
Collapse
Affiliation(s)
- Felix Broecker
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Erik Wegner
- Mikrobiologisches Institut−Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Bruna M. S. Seco
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Paulina Kaplonek
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Maria Bräutigam
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Armin Ensser
- Virologisches Institut, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Frederick Pfister
- Department of Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christopher E. Martin
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut−Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Peter H. Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| |
Collapse
|
25
|
Halloran MW, Lumb JP. Recent Applications of Diazirines in Chemical Proteomics. Chemistry 2019; 25:4885-4898. [PMID: 30444029 DOI: 10.1002/chem.201805004] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/12/2018] [Indexed: 02/06/2023]
Abstract
The elucidation of substrate-protein interactions is an important component of the drug development process. Due to the complexity of native cellular environments, elucidating these fundamental biochemical interactions remains challenging. Photoaffinity labeling (PAL) is a versatile technique that can provide insight into ligand-target interactions. By judicious modification of substrates with a photoreactive group, PAL creates a covalent crosslink between a substrate and its biological target following UV-irradiation. Among the commonly employed photoreactive groups, diazirines have emerged as the gold standard. In this Minireview, recent developments in the field of diazirine-based photoaffinity labeling will be discussed, with emphasis being placed on their applications in chemical proteomic studies.
Collapse
Affiliation(s)
- Matthew W Halloran
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, H3A 0B8, Canada
| | - Jean-Philip Lumb
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, H3A 0B8, Canada
| |
Collapse
|
26
|
Kalra P, Dhiman A, Cho WC, Bruno JG, Sharma TK. Simple Methods and Rational Design for Enhancing Aptamer Sensitivity and Specificity. Front Mol Biosci 2018; 5:41. [PMID: 29868605 PMCID: PMC5966647 DOI: 10.3389/fmolb.2018.00041] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/13/2018] [Indexed: 12/27/2022] Open
Abstract
Aptamers are structured nucleic acid molecules that can bind to their targets with high affinity and specificity. However, conventional SELEX (Systematic Evolution of Ligands by EXponential enrichment) methods may not necessarily produce aptamers of desired affinity and specificity. Thus, to address these questions, this perspective is intended to suggest some approaches and tips along with novel selection methods to enhance evolution of aptamers. This perspective covers latest novel innovations as well as a broad range of well-established approaches to improve the individual binding parameters (aptamer affinity, avidity, specificity and/or selectivity) of aptamers during and/or post-SELEX. The advantages and limitations of individual aptamer selection methods and post-SELEX optimizations, along with rational approaches to overcome these limitations are elucidated in each case. Further the impact of chosen selection milieus, linker-systems, aptamer cocktails and detection modules utilized in conjunction with target-specific aptamers, on the overall assay performance are discussed in detail, each with its own advantages and limitations. The simple variations suggested are easily available for facile implementation during and/or post-SELEX to develop ultrasensitive and specific assays. Finally, success studies of established aptamer-based assays are discussed, highlighting how they utilized some of the suggested methodologies to develop commercially successful point-of-care diagnostic assays.
Collapse
Affiliation(s)
- Priya Kalra
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Abhijeet Dhiman
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India.,Faculty of Pharmacy, Uttarakhand Technical University, Dehradun, India
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - John G Bruno
- Operational Technologies Corporation, San Antonio, TX, United States
| | - Tarun K Sharma
- Center for Biodesign and Diagnostics, Translational Health Science and Technology Institute, Faridabad, India.,AptaBharat Innovation Private Limited, Translational Health Science and Technology Institute Incubator, Faridabad, India
| |
Collapse
|
27
|
Proteomic-genomic adjustments and their confluence for elucidation of pathways and networks during liver fibrosis. Int J Biol Macromol 2018; 111:379-392. [DOI: 10.1016/j.ijbiomac.2017.12.168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/28/2017] [Accepted: 12/31/2017] [Indexed: 12/31/2022]
|
28
|
Ertürk G, Hedström M, Mattiasson B, Ruzgas T, Lood R. Highly sensitive detection and quantification of the secreted bacterial benevolence factor RoxP using a capacitive biosensor: A possible early detection system for oxidative skin diseases. PLoS One 2018; 13:e0193754. [PMID: 29494704 PMCID: PMC5833275 DOI: 10.1371/journal.pone.0193754] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/16/2018] [Indexed: 01/20/2023] Open
Abstract
The impact of the microbiota on our health is rapidly gaining interest. While several bacteria have been associated with disease, and others being indicated as having a probiotic effect, the individual biomolecules behind these alterations are often not known. A major problem in the study of these factors in vivo is their low abundance in complex environments. We recently identified the first secreted bacterial antioxidant protein, RoxP, from the skin commensal Propionibacterium acnes, suggesting its relevance for maintaining the redox homeostasis on the skin. In order to study the effect, and prevalence, of RoxP in vivo, a capacitive biosensor with a recognition surface based on molecular imprinting was used to detect RoxP on skin in vivo. In vitro analyses demonstrated the ability to detect and quantify RoxP in a concentration range of 1 x 10−13 M to 1 x 10−8 M from human skin swabs; with a limit of detection of 2.5 x 10−19 M in buffer systems. Further, the biosensor was highly selective, not responding to any other secreted protein from P. acnes. Thus, it was possible to demonstrate the presence, and quantity, of RoxP on human skin. Therefore, the developed biosensor is a very promising tool for the detection of RoxP from clinical samples, offering a rapid, cost-effective and sensitive means of detecting low-abundant bacterial proteins in vivo in complex milieus.
Collapse
Affiliation(s)
- Gizem Ertürk
- Department of Clinical Sciences Lund, Division of Infection Medicine, Biomedical Center B14, Lund University, Lund, Sweden
| | - Martin Hedström
- Department of Biotechnology, Lund University, Lund, Sweden
- CapSenze Biosystems AB, Lund, Sweden
| | - Bo Mattiasson
- Department of Biotechnology, Lund University, Lund, Sweden
- CapSenze Biosystems AB, Lund, Sweden
| | - Tautgirdas Ruzgas
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Rolf Lood
- Department of Clinical Sciences Lund, Division of Infection Medicine, Biomedical Center B14, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
29
|
Kachuk C, Doucette AA. The benefits (and misfortunes) of SDS in top-down proteomics. J Proteomics 2018; 175:75-86. [DOI: 10.1016/j.jprot.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/19/2017] [Accepted: 03/03/2017] [Indexed: 12/18/2022]
|
30
|
Yang CT, Ghosh D, Beaudry F. Detection of gelatin adulteration using bio-informatics, proteomics and high-resolution mass spectrometry. Food Addit Contam Part A Chem Anal Control Expo Risk Assess 2018; 35:599-608. [DOI: 10.1080/19440049.2017.1416680] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Charles T. Yang
- Department of Environment & Food Safety, Thermo Fisher Scientific, San Jose, U.S.A
| | - Dipankar Ghosh
- Department of Environment & Food Safety, Thermo Fisher Scientific, San Jose, U.S.A
| | - Francis Beaudry
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| |
Collapse
|
31
|
Sepehri S, Eriksson E, Kalaboukhov A, Zardán Gómez de la Torre T, Kustanovich K, Jesorka A, Schneiderman JF, Blomgren J, Johansson C, Strømme M, Winkler D. Volume-amplified magnetic bioassay integrated with microfluidic sample handling and high- Tc SQUID magnetic readout. APL Bioeng 2017; 2:016102. [PMID: 31069287 PMCID: PMC6481700 DOI: 10.1063/1.4999713] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/26/2017] [Indexed: 11/14/2022] Open
Abstract
A bioassay based on a high-Tc superconducting quantum interference device (SQUID) reading out functionalized magnetic nanoparticles (fMNPs) in a prototype microfluidic platform is presented. The target molecule recognition is based on volume amplification using padlock-probe-ligation followed by rolling circle amplification (RCA). The MNPs are functionalized with single-stranded oligonucleotides, which give a specific binding of the MNPs to the large RCA coil product, resulting in a large change in the amplitude of the imaginary part of the ac magnetic susceptibility. The RCA products from amplification of synthetic Vibrio cholera target DNA were investigated using our SQUID ac susceptibility system in microfluidic channel with an equivalent sample volume of 3 μl. From extrapolation of the linear dependence of the SQUID signal versus concentration of the RCA coils, it is found that the projected limit of detection for our system is about 1.0 × 105 RCA coils (0.2 × 10−18 mol), which is equivalent to 66 fM in the 3 μl sample volume. This ultra-high magnetic sensitivity and integration with microfluidic sample handling are critical steps towards magnetic bioassays for rapid detection of DNA and RNA targets at the point of care.
Collapse
Affiliation(s)
- Sobhan Sepehri
- Department of Microtechnology and Nanoscience-MC2, Chalmers University of Technology, Göteborg 412 96, Sweden
| | | | - Alexei Kalaboukhov
- Department of Microtechnology and Nanoscience-MC2, Chalmers University of Technology, Göteborg 412 96, Sweden
| | | | - Kiryl Kustanovich
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg 412 96, Sweden
| | - Aldo Jesorka
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg 412 96, Sweden
| | | | | | | | - Maria Strømme
- Department of Engineering Sciences, Uppsala University, The Ångström Laboratory, Box 534, SE-751 21 Uppsala, Sweden
| | - Dag Winkler
- Department of Microtechnology and Nanoscience-MC2, Chalmers University of Technology, Göteborg 412 96, Sweden
| |
Collapse
|
32
|
Gatterdam V, Frutiger A, Stengele KP, Heindl D, Lübbers T, Vörös J, Fattinger C. Focal molography is a new method for the in situ analysis of molecular interactions in biological samples. NATURE NANOTECHNOLOGY 2017; 12:1089-1095. [PMID: 28945239 DOI: 10.1038/nnano.2017.168] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/11/2017] [Indexed: 05/09/2023]
Abstract
Focal molography is a next-generation biosensor that visualizes specific biomolecular interactions in real time. It transduces affinity modulation on the sensor surface into refractive index modulation caused by target molecules that are bound to a precisely assembled nanopattern of molecular recognition sites, termed the 'mologram'. The mologram is designed so that laser light is scattered at specifically bound molecules, generating a strong signal in the focus of the mologram via constructive interference, while scattering at nonspecifically bound molecules does not contribute to the effect. We present the realization of molograms on a chip by submicrometre near-field reactive immersion lithography on a light-sensitive monolithic graft copolymer layer. We demonstrate the selective and sensitive detection of biomolecules, which bind to the recognition sites of the mologram in various complex biological samples. This allows the label-free analysis of non-covalent interactions in complex biological samples, without a need for extensive sample preparation, and enables novel time- and cost-saving ways of performing and developing immunoassays for diagnostic tests.
Collapse
Affiliation(s)
- Volker Gatterdam
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Andreas Frutiger
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | | | | | - Thomas Lübbers
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Janos Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zurich, 8092 Zurich, Switzerland
| | - Christof Fattinger
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| |
Collapse
|
33
|
de Lange V, Habegger M, Schmidt M, Vörös J. Improving FoRe: A New Inlet Design for Filtering Samples through Individual Microarray Spots. ACS Sens 2017; 2:339-345. [PMID: 28723211 DOI: 10.1021/acssensors.6b00271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this publication we present an improvement to our previously introduced vertical flow microarray, the FoRe array, which capitalizes on the fusion of immunofiltration and densely packed micron test sites. Filtering samples through individual microarray spots allows us to rapidly analyze dilute samples with high-throughput and high signal-to-noise. Unlike other flowthrough microarrays, in the FoRe design samples are injected into micron channels and sequentially exposed to different targets. This arrangement makes it possible to increase the sensitivity of the microarray by simply increasing the sample volume or to rapidly reconcentrate samples after preprocessing steps dilute the analyte. Here we present a new inlet system which allows us to increase the analyzed sample volume without compromising the micron spot size and dense layout. We combined this with a model assay to demonstrate that the device is sensitive to the amount of antigen, and as a result, sample volume directly correlates to sensitivity. We introduced a simple technique for analysis of blood, which previously clogged the nanometer-sized pores, requiring only microliter volumes expected from an infant heel prick. A drop of blood is mixed with buffer to separate the plasma before reconcentrating the sample on the microarray spot. We demonstrated the success of this procedure by spiking TNF-α into blood and achieved a limit of detection of 18 pM. Compared to traditional protein microarrays, the FoRe array is still inexpensive, customizable, and simple to use, and thanks to these improvements has a broad range of applications from small animal studies to environmental monitoring.
Collapse
Affiliation(s)
- Victoria de Lange
- Laboratory of Biosensors
and Bioelectronics, Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Marco Habegger
- Laboratory of Biosensors
and Bioelectronics, Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - Marco Schmidt
- Laboratory of Biosensors
and Bioelectronics, Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| | - János Vörös
- Laboratory of Biosensors
and Bioelectronics, Institute for Biomedical Engineering, University and ETH Zurich, Gloriastrasse 35, 8092 Zurich, Switzerland
| |
Collapse
|
34
|
Tong QH, Tao T, Xie LQ, Lu HJ. ELISA–PLA: A novel hybrid platform for the rapid, highly sensitive and specific quantification of proteins and post-translational modifications. Biosens Bioelectron 2016; 80:385-391. [DOI: 10.1016/j.bios.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/20/2016] [Accepted: 02/02/2016] [Indexed: 12/01/2022]
|
35
|
Spengler M, Adler M, Niemeyer CM. Highly sensitive ligand-binding assays in pre-clinical and clinical applications: immuno-PCR and other emerging techniques. Analyst 2016. [PMID: 26196036 DOI: 10.1039/c5an00822k] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recombinant DNA technology and corresponding innovations in molecular biology, chemistry and medicine have led to novel therapeutic biomacromolecules as lead candidates in the pharmaceutical drug development pipelines. While monoclonal antibodies and other proteins provide therapeutic potential beyond the possibilities of small molecule drugs, the concomitant demand for supportive bioanalytical sample testing creates multiple novel challenges. For example, intact macromolecules can usually not be quantified by mass-spectrometry without enzymatic digestion and isotopically labeled internal standards are costly and/or difficult to prepare. Classical ELISA-type immunoassays, on the other hand, often lack the sensitivity required to obtain pharmacokinetics of low dosed drugs or pharmacodynamics of suitable biomarkers. Here we summarize emerging state-of-the-art ligand-binding assay technologies for pharmaceutical sample testing, which reveal enhanced analytical sensitivity over classical ELISA formats. We focus on immuno-PCR, which combines antibody specificity with the extremely sensitive detection of a tethered DNA marker by quantitative PCR, and alternative nucleic acid-based technologies as well as methods based on electrochemiluminescence or single-molecule counting. Using case studies, we discuss advantages and drawbacks of these methods for preclinical and clinical sample testing.
Collapse
Affiliation(s)
- Mark Spengler
- Chimera Biotec GmbH, Emil-Figge-Str. 76 A, D-44227 Dortmund, Germany.
| | | | | |
Collapse
|
36
|
Rajan NK, Rajauria S, Ray T, Pennathur S, Cleland AN. A simple microfluidic aggregation analyzer for the specific, sensitive and multiplexed quantification of proteins in a serum environment. Biosens Bioelectron 2016; 77:1062-9. [DOI: 10.1016/j.bios.2015.10.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 10/10/2015] [Accepted: 10/31/2015] [Indexed: 11/26/2022]
|
37
|
Percy AJ, Yang J, Chambers AG, Mohammed Y, Miliotis T, Borchers CH. Protocol for Standardizing High-to-Moderate Abundance Protein Biomarker Assessments Through an MRM-with-Standard-Peptides Quantitative Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 919:515-530. [DOI: 10.1007/978-3-319-41448-5_24] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
38
|
McArdle A, Qasim Butt A, Szentpetery A, de Jager W, de Roock S, FitzGerald O, Pennington SR. Developing clinically relevant biomarkers in inflammatory arthritis: A multiplatform approach for serum candidate protein discovery. Proteomics Clin Appl 2015; 10:691-8. [PMID: 26332844 DOI: 10.1002/prca.201500046] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/17/2015] [Accepted: 08/27/2015] [Indexed: 12/23/2022]
Abstract
PURPOSE To identify candidate biomarkers that have the potential to distinguish between patients with psoriatic arthritis (PsA) or rheumatoid arthritis (RA) and explore the value of combining different protein discovery platforms for the development of a multiplexed protein biomarker panel. EXPERIMENTAL DESIGN Serum samples from 32 patients (PsA; n = 16 and RA; n = 16) defined as active, early onset, and treatment naïve were analyzed using unbiased label-free LC-MS/MS, a microsphere bead-based immunoassay (Luminex xMAP) and an aptamer-based assay (SOMAscan). RESULTS LC-MS/MS was used to quantify 324 proteins, while the Luminex xMAP targeted 48 proteins and SOMAscan supported the measurement of 1129 proteins. The combined data from these techniques gave reproducible quantification of 1501 proteins in total. Of these, 42 (LC-MS/MS), 3 (Luminex xMAP), and 127 (SOMAscan) proteins were found to be differentially expressed between PsA and RA (p < 0.05). CONCLUSION AND CLINICAL RELEVANCE Using three different and potentially complementary proteomic platforms we identified a total of 172 proteins that are differentially expressed in patients with PsA compared to RA. These proteins collectively represent candidates for inclusion in a protein signature that could be developed as a diagnostic test to discriminate patients with PsA from RA and therefore be of clinical utility.
Collapse
Affiliation(s)
- Angela McArdle
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Aisha Qasim Butt
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Agnes Szentpetery
- Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Wilco de Jager
- Department of Pediatric Immunology, Laboratory of Translational Immunology LTI, Wilhelmia Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.,Multiplex Core Facility, Laboratory of Translational Immunology LTI, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Sytze de Roock
- Department of Pediatric Immunology, Laboratory of Translational Immunology LTI, Wilhelmia Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Oliver FitzGerald
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland.,Department of Rheumatology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Stephen R Pennington
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
39
|
da Costa JP, Carvalhais V, Ferreira R, Amado F, Vilanova M, Cerca N, Vitorino R. Proteome signatures—how are they obtained and what do they teach us? Appl Microbiol Biotechnol 2015. [DOI: 10.1007/s00253-015-6795-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
40
|
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: 0.9] [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
|
41
|
Feist P, Hummon AB. Proteomic challenges: sample preparation techniques for microgram-quantity protein analysis from biological samples. Int J Mol Sci 2015; 16:3537-63. [PMID: 25664860 PMCID: PMC4346912 DOI: 10.3390/ijms16023537] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/29/2015] [Indexed: 12/22/2022] Open
Abstract
Proteins regulate many cellular functions and analyzing the presence and abundance of proteins in biological samples are central focuses in proteomics. The discovery and validation of biomarkers, pathways, and drug targets for various diseases can be accomplished using mass spectrometry-based proteomics. However, with mass-limited samples like tumor biopsies, it can be challenging to obtain sufficient amounts of proteins to generate high-quality mass spectrometric data. Techniques developed for macroscale quantities recover sufficient amounts of protein from milligram quantities of starting material, but sample losses become crippling with these techniques when only microgram amounts of material are available. To combat this challenge, proteomicists have developed micro-scale techniques that are compatible with decreased sample size (100 μg or lower) and still enable excellent proteome coverage. Extraction, contaminant removal, protein quantitation, and sample handling techniques for the microgram protein range are reviewed here, with an emphasis on liquid chromatography and bottom-up mass spectrometry-compatible techniques. Also, a range of biological specimens, including mammalian tissues and model cell culture systems, are discussed.
Collapse
Affiliation(s)
- Peter Feist
- Department of Chemistry and Biochemistry, Integrated Biomedical Sciences Program, and the Harper Cancer Research Institute, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry, Integrated Biomedical Sciences Program, and the Harper Cancer Research Institute, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
42
|
Bladergroen MR, van der Burgt YEM. Solid-phase extraction strategies to surmount body fluid sample complexity in high-throughput mass spectrometry-based proteomics. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2015; 2015:250131. [PMID: 25692071 PMCID: PMC4322654 DOI: 10.1155/2015/250131] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/08/2015] [Accepted: 01/08/2015] [Indexed: 05/08/2023]
Abstract
For large-scale and standardized applications in mass spectrometry- (MS-) based proteomics automation of each step is essential. Here we present high-throughput sample preparation solutions for balancing the speed of current MS-acquisitions and the time needed for analytical workup of body fluids. The discussed workflows reduce body fluid sample complexity and apply for both bottom-up proteomics experiments and top-down protein characterization approaches. Various sample preparation methods that involve solid-phase extraction (SPE) including affinity enrichment strategies have been automated. Obtained peptide and protein fractions can be mass analyzed by direct infusion into an electrospray ionization (ESI) source or by means of matrix-assisted laser desorption ionization (MALDI) without further need of time-consuming liquid chromatography (LC) separations.
Collapse
Affiliation(s)
- Marco R. Bladergroen
- Leiden University Medical Center (LUMC), Center for Proteomics and Metabolomics, P.O. Box 9600, 2300 RC Leiden, Netherlands
| | - Yuri E. M. van der Burgt
- Leiden University Medical Center (LUMC), Center for Proteomics and Metabolomics, P.O. Box 9600, 2300 RC Leiden, Netherlands
- *Yuri E. M. van der Burgt:
| |
Collapse
|
43
|
Solier C, Langen H. Antibody-based proteomics and biomarker research - current status and limitations. Proteomics 2014; 14:774-83. [PMID: 24520068 DOI: 10.1002/pmic.201300334] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 11/08/2013] [Accepted: 12/16/2013] [Indexed: 11/09/2022]
Abstract
Antibody-based proteomics play a very important role in biomarker discovery and validation, facilitating the high-throughput evaluation of candidate markers. Most proteomics-driven discovery is nowadays based on the use of MS. MS has many advantages, including its suitability for hypothesis-free biomarker discovery, since information on protein content of a sample is not required prior to analysis. However, MS presents one main caveat which is the limited sensitivity in complex samples, especially for body fluids, where protein expression covers a huge dynamic range. Antibody-based technologies remain the main solution to address this challenge since they reach higher sensitivity. In this article, we review the benefits and limitations of antibody-based proteomics in preclinical and clinical biomarker research for discovery and validation in body fluids and tissue. The combination of antibodies and MS, utilizing the best of both worlds, opens new avenues in biomarker research.
Collapse
Affiliation(s)
- Corinne Solier
- Translational Technologies and Bioinformatics, Pharma Research and Early Development, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | |
Collapse
|
44
|
Twist on Protein Microarrays: Layering Wax-Patterned Nitrocellulose to Create Customizable and Separable Arrays of Multiplexed Affinity Columns. Anal Chem 2014; 86:4209-16. [DOI: 10.1021/ac501211m] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
45
|
Acosta-Martin AE, Lane L. Combining bioinformatics and MS-based proteomics: clinical implications. Expert Rev Proteomics 2014; 11:269-84. [PMID: 24720436 DOI: 10.1586/14789450.2014.900446] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clinical proteomics research aims at i) discovery of protein biomarkers for screening, diagnosis and prognosis of disease, ii) discovery of protein therapeutic targets for improvement of disease prevention, treatment and follow-up, and iii) development of mass spectrometry (MS)-based assays that could be implemented in clinical chemistry, microbiology or hematology laboratories. MS has been increasingly applied in clinical proteomics studies for the identification and quantification of proteins. Bioinformatics plays a key role in the exploitation of MS data in several aspects such as the generation and curation of protein sequence databases, the development of appropriate software for MS data treatment and integration with other omics data and the establishment of adequate standard files for data sharing. In this article, we discuss the main MS approaches and bioinformatics solutions that are currently applied to accomplish the objectives of clinical proteomic research.
Collapse
|
46
|
Abstract
Shotgun proteomics (liquid chromatography-electrospray ionization-mass spectrometry, LC-ESI-MS/MS) has dominated the strategies for global protein expression in subcells, cells, tissues, and whole organisms with several types of approaches, as isobaric tags for relative and absolute quantification (iTRAQ), isotope-coded affinity tags (ICAT), or stable isotope labeling using amino acids in cell culture (SILAC) and non-labeling (label free) methods. Shotgun proteomics practically replaced the classical 2D gel electrophoresis. Selected reaction monitoring (SRM), also denominated multiple reaction monitoring (MRM), is a targeted quantitative technology that uses a complex mixture of tryptic peptides that can be selectively detected by liquid chromatography coupled to electrospray triple-quadrupole mass spectrometer; this system can select precursor ions in combination with their correspondent product ions during collision-induced dissociation to produce specific detection related to a particular protein. Here we describe protocols that are efficient to produce a complete enzymatic trypsin digestion from complex biological matrices and concomitant material to be used for LC-SRM-MS and LC-ESI-MS/MS (labeled or label free).
Collapse
|
47
|
Senapati S, Manna S, Lindsay S, Zhang P. Application of catalyst-free click reactions in attaching affinity molecules to tips of atomic force microscopy for detection of protein biomarkers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:14622-30. [PMID: 24180289 PMCID: PMC3886287 DOI: 10.1021/la4039667] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Atomic force microscopy (AFM) has been extensively used in studies of biological interactions. Particularly, AFM based force spectroscopy and recognition imaging can sense biomolecules on a single molecule level, having great potential to become a tool for molecular diagnostics in clinics. These techniques, however, require affinity molecules to be attached to AFM tips in order to specifically detect their targets. The attachment chemistry currently used on silicon tips involves multiple steps of reactions and moisture sensitive chemicals, such as (3-aminopropyl)triethoxysilane (APTES) and N-hydroxysuccinimide (NHS) ester, making the process difficult to operate in aqueous solutions. In the present study, we have developed a user-friendly protocol to functionalize the AFM tips with affinity molecules. A key feature of it is that all reactions are carried out in aqueous solutions. In summary, we first synthesized a molecular anchor composed of cyclooctyne and silatrane for introduction of a chemically reactive function to AFM tips and a bifunctional polyethylene glycol linker that harnesses two orthogonal click reactions, copper free alkyne-azide cycloaddition and thiol-vinylsulfone Michael addition, for attaching affinity molecules to AFM tips. The attachment chemistry was then validated by attaching antithrombin DNA aptamers and cyclo-RGD peptides to silicon nitride (SiN) tips, respectively, and measuring forces of unbinding these affinity molecules from their protein cognates human α-thrombin and human α5β1-integrin immobilized on mica surfaces. In turn, we used the same attachment chemistry to functionalize silicon tips with the same affinity molecules for AFM based recognition imaging, showing that the disease-relevant biomarkers such as α-thrombin and α5β1-integrin can be detected with high sensitivity and specificity by the single molecule technique. These studies demonstrate the feasibility of our attachment chemistry for the use in functionalization of AFM tips with affinity molecules.
Collapse
Affiliation(s)
- Subhadip Senapati
- Center for Single Molecule Biophysics of the Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA
| | - Saikat Manna
- Center for Single Molecule Biophysics of the Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA
| | - Stuart Lindsay
- Center for Single Molecule Biophysics of the Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA
| | - Peiming Zhang
- Center for Single Molecule Biophysics of the Biodesign Institute, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
48
|
Li J, Yuan J, Cheng KCC, Inglese J, Su XZ. Chemical genomics for studying parasite gene function and interaction. Trends Parasitol 2013; 29:603-11. [PMID: 24215777 DOI: 10.1016/j.pt.2013.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/08/2013] [Accepted: 10/08/2013] [Indexed: 12/20/2022]
Abstract
With the development of new technologies in genome sequencing, gene expression profiling, genotyping, and high-throughput screening of chemical compound libraries, small molecules are playing increasingly important roles in studying gene expression regulation, gene-gene interaction, and gene function. Here we briefly review and discuss some recent advancements in drug target identification and phenotype characterization using combinations of high-throughput screening of small-molecule libraries and various genome-wide methods such as whole-genome sequencing, genome-wide association studies (GWAS), and genome-wide expression analysis. These approaches can be used to search for new drugs against parasite infections, to identify drug targets or drug resistance genes, and to infer gene function.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | | | | | | | | |
Collapse
|