1
|
Zhang Y, Hong Z, Fu X, Li J, Yao B, Ma G. Label-Free Impedance Imaging of Single Extracellular Vesicles Using Interferometric Electrochemical Microscopy. Anal Chem 2024; 96:20230-20238. [PMID: 39661744 DOI: 10.1021/acs.analchem.4c04244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Extracellular vesicles (EVs) are pivotal in various biological processes and diseases, yet their small size and heterogeneity pose challenges for single EV quantification. We introduce interferometric electrochemical microscopy (iECM), a sensitive label-free technique that combines interferometric scattering and electrochemical impedance imaging. This method enables the quantification of the impedance of single EVs, providing unique insights into their electrochemical properties, and allows for the simultaneous measurement of size and real-time monitoring of antibody binding. Notably, we discovered that the impedance spectra of EVs can serve as specific fingerprints to differentiate EVs from cancerous and healthy cells, establishing iECM as a unique platform for EV characterization and classification.
Collapse
Affiliation(s)
- Yuanxin Zhang
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Zichen Hong
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Xuan Fu
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Jiaying Li
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Bo Yao
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| | - Guangzhong Ma
- Department of Chemistry, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
2
|
Zhou X, Chieng A, Wang S. Label-Free Optical Imaging of Nanoscale Single Entities. ACS Sens 2024; 9:543-554. [PMID: 38346398 PMCID: PMC10990724 DOI: 10.1021/acssensors.3c02526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
The advancement of optical microscopy technologies has achieved imaging of nanoscale objects, including nanomaterials, virions, organelles, and biological molecules, at the single entity level. Recently developed plasmonic and scattering based optical microscopy technologies have enabled label-free imaging of single entities with high spatial and temporal resolutions. These label-free methods eliminate the complexity of sample labeling and minimize the perturbation of the analyte native state. Additionally, these imaging-based methods can noninvasively probe the dynamics and functions of single entities with sufficient throughput for heterogeneity analysis. This perspective will review label-free single entity imaging technologies and discuss their principles, applications, and key challenges.
Collapse
Affiliation(s)
- Xinyu Zhou
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, United States
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Andy Chieng
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, United States
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, Arizona 85287, United States
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
3
|
Xu J, Huang C, Li L, Zhao Y, Guo Z, Chen Y, Zhang P. Label-free analysis of membrane protein binding kinetics and cell adhesions using evanescent scattering microscopy. Analyst 2023; 148:5084-5093. [PMID: 37671903 DOI: 10.1039/d3an00977g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Measuring ligand interactions with membrane proteins in single live cells is critical for understanding many cellular processes and screening drugs. However, developing such a capability has been a difficult challenge. Here, we employ evanescent scattering microscopy (ESM) to show that ligand binding to membrane proteins can change the cell adhesion properties, which are intrinsic cell properties and independent of random cell micromotions and ligand mass, thus allowing the kinetics analyses of both proteins and small molecules binding to membrane proteins in both single fixed and live cells. In addition, utilizing the high spatiotemporal resolution of ESM, the positions of cell adhesion sites can be tracked in real-time to analyze the cell deformations and migrations, thus providing a potential approach for understanding the cell activity during the ligand binding process in detail. The presented method may pave the road for developing a versatile and easy-to-use label-free detection strategy for in situ analysis of molecular interaction dynamics in living biosystems with single-cell resolution.
Collapse
Affiliation(s)
- Jiying Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100049, China
| | - Caixin Huang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Liangju Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
| | - Ying Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang 453003, China
- Xinxiang Key Laboratory of Clinical psychopharmacology, Xinxiang 453003, China
| | - Zhenpeng Guo
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100049, China
- National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Pengfei Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
4
|
Mapar M, Sjöberg M, Zhdanov VP, Agnarsson B, Höök F. Label-free quantification of protein binding to lipid vesicles using transparent waveguide evanescent-field scattering microscopy with liquid control. BIOMEDICAL OPTICS EXPRESS 2023; 14:4003-4016. [PMID: 37799672 PMCID: PMC10549727 DOI: 10.1364/boe.490051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/16/2023] [Accepted: 06/18/2023] [Indexed: 10/07/2023]
Abstract
Recent innovations in microscopy techniques are paving the way for label-free studies of single nanoscopic biological entities such as viruses, lipid-nanoparticle drug carriers, and even proteins. One such technique is waveguide evanescent-field microscopy, which offers a relatively simple, yet sensitive, way of achieving label-free light scattering-based imaging of nanoparticles on surfaces. Herein, we extend the application of this technique by incorporating microfluidic liquid control and adapting the design for use with inverted microscopes by fabricating a waveguide on a transparent substrate. We furthermore formulate analytical models describing scattering and fluorescence intensities from single spherical and shell-like objects interacting with evanescent fields. The models are then applied to analyze scattering and fluorescence intensities from adsorbed polystyrene beads and to temporally resolve cholera-toxin B (CTB) binding to individual surface-immobilized glycosphingolipid GM1 containing vesicles. We also propose a self-consistent means to quantify the thickness of the CTB layer, revealing that protein-binding to individual vesicles can be characterized with sub-nm precision in a time-resolved manner.
Collapse
Affiliation(s)
- Mokhtar Mapar
- Division of Biological Physics, Department of Physics,
Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Mattias Sjöberg
- Division of Biological Physics, Department of Physics,
Chalmers University of Technology, SE-41296 Göteborg, Sweden
- Nanolyze AB, BioVentureHub, Pepparedsleden 1, SE-43183 Göteborg, Sweden
| | - Vladimir P. Zhdanov
- Division of Biological Physics, Department of Physics,
Chalmers University of Technology, SE-41296 Göteborg, Sweden
- Boreskov Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Björn Agnarsson
- Division of Biological Physics, Department of Physics,
Chalmers University of Technology, SE-41296 Göteborg, Sweden
- Nanolyze AB, BioVentureHub, Pepparedsleden 1, SE-43183 Göteborg, Sweden
| | - Fredrik Höök
- Division of Biological Physics, Department of Physics,
Chalmers University of Technology, SE-41296 Göteborg, Sweden
| |
Collapse
|
5
|
Wathudura P, Wamsley M, Wang A, Chen K, Nawalage S, Wang H, Zou S, Zhang D. Effects of Cascading Optical Processes: Part II: Impacts on Experimental Quantification of Sample Absorption and Scattering Properties. Anal Chem 2023; 95:4461-4469. [PMID: 36787490 DOI: 10.1021/acs.analchem.2c05055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
In Part I of the three companion articles, we reported the effects of light scattering on experimental quantification of scattering extinction, intensity, and depolarization in solutions that contain only scatterers with no significant absorption and photoluminescence activities. The present work (Part II) studies the effects of light scattering and absorption on a series of optical spectroscopic measurements done on samples that contain both absorbers and scatterers, but not emitters. The experimental UV-vis spectrum is the sum of the sample absorption and scattering extinction spectra. However, the upper limit of the experimental Beer's-law-abiding extinction can be limited prematurely by the interference of forward scattered light. Light absorption reduces not only the sample scattering intensity but also the scattering depolarization. The impact of scattering on sample light absorption is complicated, depending on whether the absorption of scattered light is taken into consideration. Scattering reduces light absorption along the optical path length from the excitation source to the UV-vis detector. However, the absorption of the scattered light can be adequate to compensate the reduced light absorption along such optical path, making the impacts of light scattering on the sample total light absorption negligibly small (<10%). The latter finding constitutes a critical validation of the integrating-sphere-assisted resonance synchronous spectroscopic method for experimental quantification of absorption and scattering contribution to the sample UV-vis extinction spectra. The techniques and general guidelines provided in this work should help improve the reliability of optical spectroscopic characterization of nanoscale or larger materials, many of which are simultaneous absorbers and scatterers. The insights from this work are foundational for Part III of this series of work, which is on the cascading optical processes on spectroscopic measurements of fluorescent samples.
Collapse
Affiliation(s)
- Pathum Wathudura
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39759, United States
| | - Max Wamsley
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39759, United States
| | - Ankai Wang
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States
| | - Kexun Chen
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Samadhi Nawalage
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39759, United States
| | - Hui Wang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Shengli Zou
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States
| | - Dongmao Zhang
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39759, United States
| |
Collapse
|
6
|
Wan Z, Ma G, Zhang P, Wang S. Single-Protein Identification by Simultaneous Size and Charge Imaging Using Evanescent Scattering Microscopy. ACS Sens 2022; 7:2625-2633. [PMID: 36000947 PMCID: PMC9509435 DOI: 10.1021/acssensors.2c01008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Separation and identification of different proteins is one of the most fundamental tasks in biochemistry that is typically achieved by electrophoresis and Western blot techniques. Yet, it is challenging to perform such an analysis with a small sample size. Using a principle analogous to these conventional approaches, we present a label-free, single-molecule technique to identify different proteins based on the difference in their size, charge, and antibody binding. We tether single protein molecules to a sensor surface with a flexible polymer and drive them into oscillation by applying an alternating electric field. By tracking the nanometer-scale oscillation of each protein molecule via high-resolution scattering microscopy, the size and charge of each protein molecule can be determined simultaneously. Changes induced by varying the buffer pH and antibody binding are also investigated, which allows us to further expand the separation ability and identify two different proteins in a mixture. We anticipate our technique will contribute to single protein analysis and biosensing.
Collapse
Affiliation(s)
- Zijian Wan
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, Arizona 85287, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85287, USA
| | - Guangzhong Ma
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, Arizona 85287, USA
| | - Pengfei Zhang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, Arizona 85287, USA
| | - Shaopeng Wang
- Biodesign Center for Biosensors and Bioelectronics, Arizona State University, Tempe, Arizona 85287, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, USA
| |
Collapse
|
7
|
Jayakumar N, Dullo FT, Dubey V, Ahmad A, Ströhl F, Cauzzo J, Guerreiro EM, Snir O, Skalko-Basnet N, Agarwal K, Ahluwalia BS. Multi-moded high-index contrast optical waveguide for super-contrast high-resolution label-free microscopy. NANOPHOTONICS (BERLIN, GERMANY) 2022; 11:3421-3436. [PMID: 38144043 PMCID: PMC10741054 DOI: 10.1515/nanoph-2022-0100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/26/2023]
Abstract
The article elucidates the physical mechanism behind the generation of superior-contrast and high-resolution label-free images using an optical waveguide. Imaging is realized by employing a high index contrast multi-moded waveguide as a partially coherent light source. The modes provide near-field illumination of unlabeled samples, thereby repositioning the higher spatial frequencies of the sample into the far-field. These modes coherently scatter off the sample with different phases and are engineered to have random spatial distributions within the integration time of the camera. This mitigates the coherent speckle noise and enhances the contrast (2-10) × as opposed to other imaging techniques. Besides, the coherent scattering of the different modes gives rise to fluctuations in intensity. The technique demonstrated here is named chip-based Evanescent Light Scattering (cELS). The concepts introduced through this work are described mathematically and the high-contrast image generation process using a multi-moded waveguide as the light source is explained. The article then explores the feasibility of utilizing fluctuations in the captured images along with fluorescence-based techniques, like intensity-fluctuation algorithms, to mitigate poor-contrast and diffraction-limited resolution in the coherent imaging regime. Furthermore, a straight waveguide is demonstrated to have limited angular diversity between its multiple modes and therefore, for isotropic sample illumination, a multiple-arms waveguide geometry is used. The concepts introduced are validated experimentally via high-contrast label-free imaging of weakly scattering nanosized specimens such as extra-cellular vesicles (EVs), liposomes, nanobeads and biological cells such as fixed and live HeLa cells.
Collapse
Affiliation(s)
- Nikhil Jayakumar
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Firehun T. Dullo
- Department of Microsystems and Nanotechnology, SINTEF Digital, Gaustadalleen 23C, 0373Oslo, Norway
| | - Vishesh Dubey
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Azeem Ahmad
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Florian Ströhl
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Jennifer Cauzzo
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø9037, Norway
| | | | - Omri Snir
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Natasa Skalko-Basnet
- Department of Pharmacy, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Krishna Agarwal
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
| | - Balpreet Singh Ahluwalia
- Department of Physics and Technology, UiT The Arctic University of Norway, Tromsø9037, Norway
- Department of Clinical Science, Intervention and Technology, Karolinska Insitute, 17177Stockholm, Sweden
| |
Collapse
|
8
|
A review of optical methods for ultrasensitive detection and characterization of nanoparticles in liquid media with a focus on the wide field surface plasmon microscopy. Anal Chim Acta 2022; 1204:339633. [DOI: 10.1016/j.aca.2022.339633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/27/2022]
|
9
|
Altug H, Oh SH, Maier SA, Homola J. Advances and applications of nanophotonic biosensors. NATURE NANOTECHNOLOGY 2022; 17:5-16. [PMID: 35046571 DOI: 10.1038/s41565-021-01045-5] [Citation(s) in RCA: 245] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 05/14/2023]
Abstract
Nanophotonic devices, which control light in subwavelength volumes and enhance light-matter interactions, have opened up exciting prospects for biosensing. Numerous nanophotonic biosensors have emerged to address the limitations of the current bioanalytical methods in terms of sensitivity, throughput, ease-of-use and miniaturization. In this Review, we provide an overview of the recent developments of label-free nanophotonic biosensors using evanescent-field-based sensing with plasmon resonances in metals and Mie resonances in dielectrics. We highlight the prospects of achieving an improved sensor performance and added functionalities by leveraging nanostructures and on-chip and optoelectronic integration, as well as microfluidics, biochemistry and data science toolkits. We also discuss open challenges in nanophotonic biosensing, such as reducing the overall cost and handling of complex biological samples, and provide an outlook for future opportunities to improve these technologies and thereby increase their impact in terms of improving health and safety.
Collapse
Affiliation(s)
- Hatice Altug
- Laboratory of Bionanophotonic Systems, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, USA.
| | - Stefan A Maier
- Chair in Hybrid Nanosystems, Nanoinstitut Munich, Faculty of Physics, Ludwig-Maximilians Universität München, Munich, Germany.
- Department of Physics, Imperial College London, London, UK.
| | - Jiří Homola
- Institute of Photonics and Electronics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
10
|
Priest L, Peters JS, Kukura P. Scattering-based Light Microscopy: From Metal Nanoparticles to Single Proteins. Chem Rev 2021; 121:11937-11970. [PMID: 34587448 PMCID: PMC8517954 DOI: 10.1021/acs.chemrev.1c00271] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Indexed: 02/02/2023]
Abstract
Our ability to detect, image, and quantify nanoscopic objects and molecules with visible light has undergone dramatic improvements over the past few decades. While fluorescence has historically been the go-to contrast mechanism for ultrasensitive light microscopy due to its superior background suppression and specificity, recent developments based on light scattering have reached single-molecule sensitivity. They also have the advantages of universal applicability and the ability to obtain information about the species of interest beyond its presence and location. Many of the recent advances are driven by novel approaches to illumination, detection, and background suppression, all aimed at isolating and maximizing the signal of interest. Here, we review these developments grouped according to the basic principles used, namely darkfield imaging, interferometric detection, and surface plasmon resonance microscopy.
Collapse
Affiliation(s)
| | | | - Philipp Kukura
- Physical and Theoretical
Chemistry Laboratory, Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QZ, United Kingdom
| |
Collapse
|
11
|
Ettabib MA, Marti A, Liu Z, Bowden BM, Zervas MN, Bartlett PN, Wilkinson JS. Waveguide Enhanced Raman Spectroscopy for Biosensing: A Review. ACS Sens 2021; 6:2025-2045. [PMID: 34114813 DOI: 10.1021/acssensors.1c00366] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Waveguide enhanced Raman spectroscopy (WERS) utilizes simple, robust, high-index contrast dielectric waveguides to generate a strong evanescent field, through which laser light interacts with analytes residing on the surface of the waveguide. It offers a powerful tool for the direct identification and reproducible quantification of biochemical species and an alternative to surface enhanced Raman spectroscopy (SERS) without reliance on fragile noble metal nanostructures. The advent of low-cost laser diodes, compact spectrometers, and recent progress in material engineering, nanofabrication techniques, and software modeling tools have made realizing portable and cheap WERS Raman systems with high sensitivity a realistic possibility. This review highlights the latest progress in WERS technology and summarizes recent demonstrations and applications. Following an introduction to the fundamentals of WERS, the theoretical framework that underpins the WERS principles is presented. The main WERS design considerations are then discussed, and a review of the available approaches for the modification of waveguide surfaces for the attachment of different biorecognition elements is provided. The review concludes by discussing and contrasting the performance of recent WERS implementations, thereby providing a future roadmap of WERS technology where the key opportunities and challenges are highlighted.
Collapse
Affiliation(s)
- Mohamed A. Ettabib
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Almudena Marti
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Zhen Liu
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Bethany M. Bowden
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Michalis N. Zervas
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Philip N. Bartlett
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - James S. Wilkinson
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
12
|
Sjöberg M, Mapar M, Armanious A, Zhdanov VP, Agnarsson B, Höök F. Time-Resolved and Label-Free Evanescent Light-Scattering Microscopy for Mass Quantification of Protein Binding to Single Lipid Vesicles. NANO LETTERS 2021; 21:4622-4628. [PMID: 34003003 PMCID: PMC8289281 DOI: 10.1021/acs.nanolett.1c00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/05/2021] [Indexed: 06/12/2023]
Abstract
In-depth understanding of the intricate interactions between biomolecules and nanoparticles is hampered by a lack of analytical methods providing quantitative information about binding kinetics. Herein, we demonstrate how label-free evanescent light-scattering microscopy can be used to temporally resolve specific protein binding to individual surface-bound (∼100 nm) lipid vesicles. A theoretical model is proposed that translates protein-induced changes in light-scattering intensity into bound mass. Since the analysis is centered on individual lipid vesicles, the signal from nonspecific protein binding to the surrounding surface is completely avoided, offering a key advantage over conventional surface-based techniques. Further, by averaging the intensities from less than 2000 lipid vesicles, the sensitivity is shown to increase by orders of magnitude. Taken together, these features provide a new avenue in studies of protein-nanoparticle interaction, in general, and specifically in the context of nanoparticles in medical diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mattias Sjöberg
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Mokhtar Mapar
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Antonius Armanious
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Vladimir P. Zhdanov
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
- Boreskov
Institute of Catalysis, Russian Academy
of Sciences, Novosibirsk 630090, Russia
| | - Björn Agnarsson
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| | - Fredrik Höök
- Division
of Nano and Biophysics, Department of Physics, Chalmers University of Technology, Gothenburg 41296, Sweden
| |
Collapse
|
13
|
Single-molecule studies of amyloid proteins: from biophysical properties to diagnostic perspectives. Q Rev Biophys 2020; 53:e12. [PMID: 33148356 DOI: 10.1017/s0033583520000086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In neurodegenerative diseases, a wide range of amyloid proteins or peptides such as amyloid-beta and α-synuclein fail to keep native functional conformations, followed by misfolding and self-assembling into a diverse array of aggregates. The aggregates further exert toxicity leading to the dysfunction, degeneration and loss of cells in the affected organs. Due to the disordered structure of the amyloid proteins, endogenous molecules, such as lipids, are prone to interact with amyloid proteins at a low concentration and influence amyloid cytotoxicity. The heterogeneity of amyloid proteinscomplicates the understanding of the amyloid cytotoxicity when relying only on conventional bulk and ensemble techniques. As complementary tools, single-molecule techniques (SMTs) provide novel insights into the different subpopulations of a heterogeneous amyloid mixture as well as the cytotoxicity, in particular as involved in lipid membranes. This review focuses on the recent advances of a series of SMTs, including single-molecule fluorescence imaging, single-molecule force spectroscopy and single-nanopore electrical recording, for the understanding of the amyloid molecular mechanism. The working principles, benefits and limitations of each technique are discussed and compared in amyloid protein related studies.. We also discuss why SMTs show great potential and are worthy of further investigation with feasibility studies as diagnostic tools of neurodegenerative diseases and which limitations are to be addressed.
Collapse
|
14
|
Plasmonic scattering imaging of single proteins and binding kinetics. Nat Methods 2020; 17:1010-1017. [PMID: 32958922 PMCID: PMC7541716 DOI: 10.1038/s41592-020-0947-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 08/10/2020] [Indexed: 11/08/2022]
Abstract
Measuring the binding kinetics of single proteins represents one of the most important and challenging tasks in protein analysis. Here we show that this is possible using a surface plasmon resonance (SPR) scattering technique. SPR is a popular label-free detection technology because of its extraordinary sensitivity, but it has never been used for imaging single proteins. We overcome this limitation by imaging scattering of surface plasmonic waves by proteins. This allows us to image single proteins, measure their sizes and identify them based on their specific binding to antibodies. We further show that it is possible to quantify protein binding kinetics by counting the binding of individual molecules, providing a digital method to measure binding kinetics and analyze heterogeneity of protein behavior. We anticipate that this imaging method will become an important tool for single protein analysis, especially for low volume samples, such as single cells.
Collapse
|
15
|
Single-vesicle imaging reveals lipid-selective and stepwise membrane disruption by monomeric α-synuclein. Proc Natl Acad Sci U S A 2020; 117:14178-14186. [PMID: 32513706 PMCID: PMC7322013 DOI: 10.1073/pnas.1914670117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative diseases are increasing among the world's population, but there are no cures. These disorders all involve proteins that assemble into amyloid fibers which results in brain cell death. Evidence suggests that association of these proteins with lipid membranes is crucial for both functional and pathological roles. In Parkinson's disease, the involved protein, α-synuclein, is thought to function in trafficking of lipid vesicles in the brain. In search of mechanistic origins, increasing focus is put on identifying neurotoxic reactions induced by membrane interactions. To contribute new clues to this question, we here employed a new surface-sensitive scattering microscopy technique. With this approach, we discovered that α-synuclein perturbs vesicles in a stepwise and lipid-dependent fashion already at very low protein coverage. The interaction of the neuronal protein α-synuclein with lipid membranes appears crucial in the context of Parkinson’s disease, but the underlying mechanistic details, including the roles of different lipids in pathogenic protein aggregation and membrane disruption, remain elusive. Here, we used single-vesicle resolution fluorescence and label-free scattering microscopy to investigate the interaction kinetics of monomeric α-synuclein with surface-tethered vesicles composed of different negatively charged lipids. Supported by a theoretical model to account for structural changes in scattering properties of surface-tethered lipid vesicles, the data demonstrate stepwise vesicle disruption and asymmetric membrane deformation upon α-synuclein binding to phosphatidylglycerol vesicles at protein concentrations down to 10 nM (∼100 proteins per vesicle). In contrast, phosphatidylserine vesicles were only marginally affected. These insights into structural consequences of α-synuclein interaction with lipid vesicles highlight the contrasting roles of different anionic lipids, which may be of mechanistic relevance for both normal protein function (e.g., synaptic vesicle binding) and dysfunction (e.g., mitochondrial membrane interaction).
Collapse
|
16
|
Midtvedt D, Eklund F, Olsén E, Midtvedt B, Swenson J, Höök F. Size and Refractive Index Determination of Subwavelength Particles and Air Bubbles by Holographic Nanoparticle Tracking Analysis. Anal Chem 2019; 92:1908-1915. [DOI: 10.1021/acs.analchem.9b04101] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Daniel Midtvedt
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Fredrik Eklund
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Erik Olsén
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Benjamin Midtvedt
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Jan Swenson
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Fredrik Höök
- Division of Biological Physics, Department of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| |
Collapse
|
17
|
Quantitative accounting of dye leakage and photobleaching in single lipid vesicle measurements: Implications for biomacromolecular interaction analysis. Colloids Surf B Biointerfaces 2019; 182:110338. [DOI: 10.1016/j.colsurfb.2019.06.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/08/2019] [Accepted: 06/28/2019] [Indexed: 02/05/2023]
|
18
|
Abstract
A microfluidic device as a pivotal research tool in chemistry and life science is now widely recognized. Indeed, microfluidic techniques have made significant advancements in fundamental research, such as the inherent heterogeneity of single-cells studies in cell populations, which would be helpful in understanding cellular molecular mechanisms and clinical diagnosis of major diseases. Single-cell analyses on microdevices have shown great potential for precise fluid control, cell manipulation, and signal output with rapid and high throughput. Moreover, miniaturized devices also have open functions such as integrating with traditional detection methods, for example, optical, electrochemical or mass spectrometry for single-cell analysis. In this review, we summarized recent advances of single-cell analysis based on various microfluidic approaches from different dimensions, such as in vitro, ex vivo, and in vivo analysis of single cells.
Collapse
Affiliation(s)
- Xiaowen Ou
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology
| |
Collapse
|
19
|
Park S, Jackman JA, Xu X, Weiss PS, Cho NJ. Micropatterned Viral Membrane Clusters for Antiviral Drug Evaluation. ACS APPLIED MATERIALS & INTERFACES 2019; 11:13984-13990. [PMID: 30855935 DOI: 10.1021/acsami.9b01724] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The function of biological nanoparticles, such as membrane-enveloped viral particles, is often enhanced when the particles form higher-order supramolecular assemblies. While there is intense interest in developing biomimetic platforms that recapitulate these collective properties, existing platforms are limited to mimicking individual virus particles. Here, we present a micropatterning strategy to print linker molecules selectively onto bioinert surfaces, thereby enabling controlled tethering of biomimetic viral particle clusters across defined geometric patterns. By controlling the linker concentration, it is possible to tune the density of tethered particles within clusters while enhancing the signal intensity of encapsulated fluorescent markers. Time-resolved tracking of pore formation and membrane lysis revealed that an antiviral peptide can disturb clusters of the membrane-enclosed particles akin to the targeting of individual viral particles. This platform is broadly useful for evaluating the performance of membrane-active antiviral drug candidates, whereas the micropatterning strategy can be applied to a wide range of biological nanoparticles and other macromolecular entities.
Collapse
Affiliation(s)
- Soohyun Park
- School of Materials Science and Engineering , Nanyang Technological University , 50 Nanyang Drive , Singapore 637553 , Singapore
| | | | - Xiaobin Xu
- California NanoSystems Institute, Department of Chemistry and Biochemistry, and Department of Materials Science and Engineering , University of California, Los Angeles , Los Angeles , California 90095-7227 , United States
- School of Materials Science and Engineering , Tongji University , 1239 Siping Road , Shanghai 200092 , China
| | - Paul S Weiss
- California NanoSystems Institute, Department of Chemistry and Biochemistry, and Department of Materials Science and Engineering , University of California, Los Angeles , Los Angeles , California 90095-7227 , United States
| | - Nam-Joon Cho
- School of Materials Science and Engineering , Nanyang Technological University , 50 Nanyang Drive , Singapore 637553 , Singapore
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 62 Nanyang Drive , Singapore 637459 , Singapore
| |
Collapse
|
20
|
Watts KE, Blackburn TJ, Pemberton JE. Optical Spectroscopy of Surfaces, Interfaces, and Thin Films: A Status Report. Anal Chem 2019; 91:4235-4265. [PMID: 30790520 DOI: 10.1021/acs.analchem.9b00735] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Kristen E Watts
- Department of Chemistry and Biochemistry University of Arizona 1306 East University Boulevard , Tucson , Arizona 85721 , United States
| | - Thomas J Blackburn
- Department of Chemistry and Biochemistry University of Arizona 1306 East University Boulevard , Tucson , Arizona 85721 , United States
| | - Jeanne E Pemberton
- Department of Chemistry and Biochemistry University of Arizona 1306 East University Boulevard , Tucson , Arizona 85721 , United States
| |
Collapse
|
21
|
Yang CT, Xu Y, Pourhassan-Moghaddam M, Tran DP, Wu L, Zhou X, Thierry B. Surface Plasmon Enhanced Light Scattering Biosensing: Size Dependence on the Gold Nanoparticle Tag. SENSORS (BASEL, SWITZERLAND) 2019; 19:E323. [PMID: 30650578 PMCID: PMC6359480 DOI: 10.3390/s19020323] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 12/31/2022]
Abstract
Surface plasmon enhanced light scattering (SP-LS) is a powerful new sensing SPR modality that yields excellent sensitivity in sandwich immunoassay using spherical gold nanoparticle (AuNP) tags. Towards further improving the performance of SP-LS, we systematically investigated the AuNP size effect. Simulation results indicated an AuNP size-dependent scattered power, and predicted the optimized AuNPs sizes (i.e., 100 and 130 nm) that afford extremely high signal enhancement in SP-LS. The maximum scattered power from a 130 nm AuNP is about 1700-fold higher than that obtained from a 17 nm AuNP. Experimentally, a bio-conjugation protocol was developed by coating the AuNPs with mixture of low and high molecular weight PEG molecules. Optimal IgG antibody bioconjugation conditions were identified using physicochemical characterization and a model dot-blot assay. Aggregation prevented the use of the larger AuNPs in SP-LS experiments. As predicted by simulation, AuNPs with diameters of 50 and 64 nm yielded significantly higher SP-LS signal enhancement in comparison to the smaller particles. Finally, we demonstrated the feasibility of a two-step SP-LS protocol based on a gold enhancement step, aimed at enlarging 36 nm AuNPs tags. This study provides a blue-print for the further development of SP-LS biosensing and its translation in the bioanalytical field.
Collapse
Affiliation(s)
- Chih-Tsung Yang
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio and Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Mawson Lakes 5095, Australia.
| | - Yi Xu
- Electronics and Photonics Department, Institute of High Performance Computing, Agency for Science, Technology, and Research (A*STAR), Singapore 138632, Singapore.
- SUTD-MIT International Design Center & Science and Math Cluster, Singapore University of Technology and Design, Singapore 487372, Singapore.
| | - Mohammad Pourhassan-Moghaddam
- School of Biomedical Engineering, University of Technology Sydney, Sydney 2007, Australia.
- Institute for Biomedical Materials and Devices (IBMD), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia.
| | - Duy Phu Tran
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio and Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Mawson Lakes 5095, Australia.
| | - Lin Wu
- Electronics and Photonics Department, Institute of High Performance Computing, Agency for Science, Technology, and Research (A*STAR), Singapore 138632, Singapore.
| | - Xin Zhou
- Institute of Comparative Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio and Nano Science and Technology, University of South Australia, Mawson Lakes Campus, Mawson Lakes 5095, Australia.
| |
Collapse
|
22
|
Abstract
Advances in microfluidic techniques have prompted researchers to study the inherent heterogeneity of single cells in cell populations.
Collapse
Affiliation(s)
- Qiushi Huang
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Sifeng Mao
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Mashooq Khan
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Jin-Ming Lin
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| |
Collapse
|
23
|
Dubey V, Ahmad A, Singh R, Wolfson DL, Basnet P, Acharya G, Mehta DS, Ahluwalia BS. Multi-modal chip-based fluorescence and quantitative phase microscopy for studying inflammation in macrophages. OPTICS EXPRESS 2018; 26:19864-19876. [PMID: 30119307 DOI: 10.1364/oe.26.019864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/14/2018] [Indexed: 05/22/2023]
Abstract
Total internal reflection fluorescence (TIRF) microscopy benefits from high-sensitivity, low background noise, low photo-toxicity and high-contrast imaging of sub-cellular structures close to the membrane surface. Although, TIRF microscopy provides high-contrast imaging it does not provide quantitative information about morphological features of the biological cells. Here, we propose an integrated waveguide chip-based TIRF microscopy and label-free quantitative phase imaging (QPI). The evanescent field present on top of a waveguide surface is used to excite the fluorescence and an upright microscope is used to collect the signal. The upright microscope is converted into a Linnik-type interferometer to sequentially extract both the quantitative phase information and TIRF images of the cells. Waveguide chip-based TIRF microscopy benefits from decoupling of illumination and collection light path, large field of view imaging and pre-aligned configuration for multi-color TIRF imaging. The proposed multi-modal microscopy is used to study inflammation caused by lipopolysaccharide (LPS) on rat macrophages. The TIRF microscopy showed that LPS inflammatory molecule disrupts the cell membrane and causes cells to significantly expand across a substrate. While, QPI module quantified changes in the sub-cellular content of the LPS challenged macrophages, showing a net decrease in its maximum phase values.
Collapse
|
24
|
Bioinspired, nanoscale approaches in contemporary bioanalytics (Review). Biointerphases 2018; 13:040801. [DOI: 10.1116/1.5037582] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
25
|
Rupert DLM, Mapar M, Shelke GV, Norling K, Elmeskog M, Lötvall JO, Block S, Bally M, Agnarsson B, Höök F. Effective Refractive Index and Lipid Content of Extracellular Vesicles Revealed Using Optical Waveguide Scattering and Fluorescence Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:8522-8531. [PMID: 29923735 DOI: 10.1021/acs.langmuir.7b04214] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Extracellular vesicles (EVs) are generating a growing interest because of the key roles they play in various biological processes and because of their potential use as biomarkers in clinical diagnostics and as efficient carriers in drug-delivery and gene-therapy applications. Their full exploitation, however, depends critically on the possibility to classify them into different subpopulations, a task that in turn relies on efficient means to identify their unique biomolecular and physical signatures. Because of the large heterogeneity of EV samples, such information remains rather elusive, and there is accordingly a need for new and complementary characterization schemes that can help expand the library of distinct EV features. In this work, we used surface-sensitive waveguide scattering microscopy with single EV resolution to characterize two subsets of similarly sized EVs that were preseparated based on their difference in buoyant density. Unexpectedly, the scattering intensity distribution revealed that the scattering intensity of the high-density (HD) population was on an average a factor of three lower than that of the low-density (LD) population. By further labeling the EV samples with a self-inserting lipid-membrane dye, the scattering and fluorescence intensities from EVs could be simultaneously measured and correlated at the single-particle level. The labeled HD sample exhibited not only lower fluorescence and scattering intensities but also lower effective refractive index ( n ≈ 1.35) compared with the LD EVs ( n ≈ 1.38), indicating that both the lipid and protein contents were indeed lower in the HD EVs. Although separation in density gradients of similarly sized EVs is usually linked to differences in biomolecular content, we suggest based on these observations that the separation rather reflects the ability of the solute of the gradient to penetrate the lipid membrane enclosing the EVs, that is, the two gradient bands are more likely because of the differences in membrane permeability than to differences in biomolecular content of the EVs.
Collapse
Affiliation(s)
- Déborah L M Rupert
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Mokhtar Mapar
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Ganesh Vilas Shelke
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition , University of Gothenburg , 40530 Gothenburg , Sweden
| | - Karin Norling
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Mathias Elmeskog
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Jan O Lötvall
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition , University of Gothenburg , 40530 Gothenburg , Sweden
| | - Stephan Block
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Marta Bally
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
- Institut Curie, Centre de Recherche, CNRS, UMR168, Physico-Chimie Curie , Paris 75016 , France
| | - Björn Agnarsson
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| | - Fredrik Höök
- Department of Physics , Chalmers University of Technology , 41296 Gothenburg , Sweden
| |
Collapse
|
26
|
Wiklander OPB, Bostancioglu RB, Welsh JA, Zickler AM, Murke F, Corso G, Felldin U, Hagey DW, Evertsson B, Liang XM, Gustafsson MO, Mohammad DK, Wiek C, Hanenberg H, Bremer M, Gupta D, Björnstedt M, Giebel B, Nordin JZ, Jones JC, El Andaloussi S, Görgens A. Systematic Methodological Evaluation of a Multiplex Bead-Based Flow Cytometry Assay for Detection of Extracellular Vesicle Surface Signatures. Front Immunol 2018; 9:1326. [PMID: 29951064 PMCID: PMC6008374 DOI: 10.3389/fimmu.2018.01326] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/28/2018] [Indexed: 01/07/2023] Open
Abstract
Extracellular vesicles (EVs) can be harvested from cell culture supernatants and from all body fluids. EVs can be conceptually classified based on their size and biogenesis as exosomes and microvesicles. Nowadays, it is however commonly accepted in the field that there is a much higher degree of heterogeneity within these two subgroups than previously thought. For instance, the surface marker profile of EVs is likely dependent on the cell source, the cell’s activation status, and multiple other parameters. Within recent years, several new methods and assays to study EV heterogeneity in terms of surface markers have been described; most of them are being based on flow cytometry. Unfortunately, such methods generally require dedicated instrumentation, are time-consuming and demand extensive operator expertise for sample preparation, acquisition, and data analysis. In this study, we have systematically evaluated and explored the use of a multiplex bead-based flow cytometric assay which is compatible with most standard flow cytometers and facilitates a robust semi-quantitative detection of 37 different potential EV surface markers in one sample simultaneously. First, assay variability, sample stability over time, and dynamic range were assessed together with the limitations of this assay in terms of EV input quantity required for detection of differently abundant surface markers. Next, the potential effects of EV origin, sample preparation, and quality of the EV sample on the assay were evaluated. The findings indicate that this multiplex bead-based assay is generally suitable to detect, quantify, and compare EV surface signatures in various sample types, including unprocessed cell culture supernatants, cell culture-derived EVs isolated by different methods, and biological fluids. Furthermore, the use and limitations of this assay to assess heterogeneities in EV surface signatures was explored by combining different sets of detection antibodies in EV samples derived from different cell lines and subsets of rare cells. Taken together, this validated multiplex bead-based flow cytometric assay allows robust, sensitive, and reproducible detection of EV surface marker expression in various sample types in a semi-quantitative way and will be highly valuable for many researchers in the EV field in different experimental contexts.
Collapse
Affiliation(s)
- Oscar P B Wiklander
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Evox Therapeutics Limited, Oxford, United Kingdom
| | - R Beklem Bostancioglu
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joshua A Welsh
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Antje M Zickler
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Division of Pathology F56, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Florian Murke
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Giulia Corso
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Felldin
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel W Hagey
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Björn Evertsson
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Xiu-Ming Liang
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Manuela O Gustafsson
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dara K Mohammad
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Constanze Wiek
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.,Department of Pediatrics III, University Children's Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Michel Bremer
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dhanu Gupta
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Björnstedt
- Division of Pathology F56, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Joel Z Nordin
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Evox Therapeutics Limited, Oxford, United Kingdom
| | - Jennifer C Jones
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Samir El Andaloussi
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Evox Therapeutics Limited, Oxford, United Kingdom.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - André Görgens
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Evox Therapeutics Limited, Oxford, United Kingdom.,Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
27
|
Lundgren A, Fast BJ, Block S, Agnarsson B, Reimhult E, Gunnarsson A, Höök F. Affinity Purification and Single-Molecule Analysis of Integral Membrane Proteins from Crude Cell-Membrane Preparations. NANO LETTERS 2018; 18:381-385. [PMID: 29231738 DOI: 10.1021/acs.nanolett.7b04227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The function of integral membrane proteins is critically dependent on their naturally surrounding lipid membrane. Detergent-solubilized and purified membrane proteins are therefore often reconstituted into cell-membrane mimics and analyzed for their function with single-molecule microscopy. Expansion of this approach toward a broad range of pharmaceutically interesting drug targets and biomarkers however remains hampered by the fact that these proteins have low expression levels, and that detergent solubilization and reconstitution often cause protein conformational changes and loss of membrane-specific cofactors, which may impair protein function. To overcome this limitation, we here demonstrate how antibody-modified nanoparticles can be used to achieve affinity purification and enrichment of selected integral membrane proteins directly from cell membrane preparations. Nanoparticles were first bound to the ectodomain of β-secretase 1 (BACE1) contained in cell-derived membrane vesicles. In a subsequent step, these were merged into a continuous supported membrane in a microfluidic channel. Through the extended nanoparticle tag, a weak (∼fN) hydrodynamic force could be applied, inducing directed in-membrane movement of targeted BACE1 exclusively. This enabled selective thousand-fold enrichment of the targeted membrane protein while preserving a natural lipid environment. In addition, nanoparticle-targeting also enabled simultaneous tracking analysis of each individual manipulated protein, revealing how their mobility changed when moved from one lipid environment to another. We therefore believe this approach will be particularly useful for separation in-line with single-molecule analysis, eventually opening up for membrane-protein sorting devices analogous to fluorescence-activated cell sorting.
Collapse
Affiliation(s)
- Anders Lundgren
- Department of Physics, Chalmers University of Technology , 41296 Göteborg, Sweden
- Department of Nanobiotechnology, University of Natural Resources and Life Sciences , 1190 Vienna, Austria
| | - Björn Johansson Fast
- Department of Physics, Chalmers University of Technology , 41296 Göteborg, Sweden
| | - Stephan Block
- Department of Physics, Chalmers University of Technology , 41296 Göteborg, Sweden
| | - Björn Agnarsson
- Department of Physics, Chalmers University of Technology , 41296 Göteborg, Sweden
| | - Erik Reimhult
- Department of Nanobiotechnology, University of Natural Resources and Life Sciences , 1190 Vienna, Austria
| | - Anders Gunnarsson
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca , 43183 Mölndal, Sweden
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology , 41296 Göteborg, Sweden
| |
Collapse
|
28
|
Pandey V, Gupta S, Elangovan R. Compact 3D printed module for fluorescence and label-free imaging using evanescent excitation. Methods Appl Fluoresc 2017; 6:015007. [PMID: 29076809 DOI: 10.1088/2050-6120/aa967a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Total internal reflection fluorescence (TIRF) microscopy is widely used for selective excitation and high-resolution imaging of fluorophores, and more recently label-free nanosized objects, with high vertical confinement near a liquid-solid interface. Traditionally, high numerical aperture objectives (>1.4) are used to simultaneously generate evanescent waves and collect fluorescence emission signals which limits their use to small area imaging (<0.1 mm2). Objective-based TIRFs are also expensive as they require dichroic mirrors and efficient notch filters to prevent specular reflection within the objective lenses. We have developed a compact 3D module called cTIRF that can generate evanescent waves in microscope glass slides via a planar waveguide illumination. The module can be attached as a fixture to any existing optical microscope, converting it into a TIRF and enabling high signal-to-noise ratio (SNR) fluorescence imaging using any magnification objective. As the incidence optics is perpendicular to the detector, label-free evanescent scattering-based imaging of submicron objects can also be performed without using emission filters. SNR is significantly enhanced in this case as compared to cTIRF alone, as seen through our model experiments performed on latex beads and mammalian cells. Extreme flexibility and the low cost of our approach makes it scalable for limited resource settings.
Collapse
Affiliation(s)
- Vikas Pandey
- Department of Chemical Engineering, Indian Institute of Technology, Delhi, 110016, India
| | | | | |
Collapse
|
29
|
Tinguely JC, Helle ØI, Ahluwalia BS. Silicon nitride waveguide platform for fluorescence microscopy of living cells. OPTICS EXPRESS 2017; 25:27678-27690. [PMID: 29092239 DOI: 10.1364/oe.25.027678] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/29/2017] [Indexed: 05/28/2023]
Abstract
Waveguide chip-based microscopy reduces the complexity of total internal reflection fluorescence (TIRF) microscopy, and adds features like large field of view illumination, decoupling of illumination and collection path and easy multimodal imaging. However, for the technique to become widespread there is a need of low-loss and affordable waveguides made of high-refractive index material. Here, we develop and report a low-loss silicon nitride (Si3N4) waveguide platform for multi-color TIRF microscopy. Single mode conditions at visible wavelengths (488-660 nm) were achieved using shallow rib geometry. To generate uniform excitation over appropriate dimensions waveguide bends were used to filter-out higher modes followed by adiabatic tapering. Si3N4 material is finally shown to be biocompatible for growing and imaging living cells.
Collapse
|
30
|
Yang CT, Pourhassan-Moghaddam M, Wu L, Bai P, Thierry B. Ultrasensitive Detection of Cancer Prognostic miRNA Biomarkers Based on Surface Plasmon Enhanced Light Scattering. ACS Sens 2017; 2:635-640. [PMID: 28723162 DOI: 10.1021/acssensors.6b00776] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of simple yet ultrasensitive biosensing approaches for the detection of cancer prognostic microRNA is an important step toward their successful clinical implementation. We demonstrate the relevance for the detection of circulating miRNA of a novel signal amplification scheme based on surface plasmon resonance enhanced light scattering (SP-LS). In addition to experimental optimization carried out using gold nanoparticle (AuNP) tags conjugated with a monoclonal antibody with high affinity for RNA*DNA hybrid duplexes, simulation modeling was conducted to obtain insights about SP-LS biosensing. SP-LS enabled the detection of miRNA-122 at subpicomolar concentrations within 30 min, and a limit of detection of 2 attomoles (60 fM, 50 μL) was determined. MiRNA-122 could also be reliably detected in a high concentration background of nontarget miRNA. The proposed SP-LS miRNA detection approach could be readily applied to other miRNA targets of diagnostic importance and further developed to allow for multiplex measurements of miRNA panels. The promising results obtained in this study and advantageous features of SP-LS warrant further development and its application to clinical samples.
Collapse
Affiliation(s)
| | - Mohammad Pourhassan-Moghaddam
- Department
of Medical Biotechnology and Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Lin Wu
- Electronics
and Photonics Department, Institute of High Performance Computing,
Agency for Science, Technology, and Research (A*STAR), Singapore 138632
| | - Ping Bai
- Electronics
and Photonics Department, Institute of High Performance Computing,
Agency for Science, Technology, and Research (A*STAR), Singapore 138632
| | | |
Collapse
|
31
|
Wang J, Huang X, Liu H, Dong C, Ren J. Fluorescence and Scattering Light Cross Correlation Spectroscopy and Its Applications in Homogeneous Immunoassay. Anal Chem 2017; 89:5230-5237. [PMID: 28436659 DOI: 10.1021/acs.analchem.6b04547] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In this work, we propose fluorescence and scattering light cross-correlation spectroscopy (FSCCS) based on laser confocal configuration using silver nanoparticle (SNPs) and Alexa Fluor 488 (Alexa) as probe pairs. FSCCS is a single molecule (particle) method, and its principle is similar to that of fluorescence cross-correlation spectroscopy (FCCS). We established the setup of FSCCS using single wavelength laser and developed an immunoassay model of FSCCS. The reliability and adaptability of FSCCS method were evaluated by homogeneous sandwich immunoassay mode. In the study, liver cancer biomarker alpha-fetoprotein (AFP) was used as an assay model, two different antibodies were labeled with SNPs and fluorophore Alexa Fluor 488, respectively. In the optimal conditions, the linear range of AFP covers 5 pM to 580 pM and the detection limit is 3.1 pM. This method was successfully applied for direct determination of AFP levels in human serum samples, and the obtained results were in good agreement with data obtained via ELISAs. The advantage of this method lies in its simplicity, attractive SNPs probes, high sensitivity and selectivity and high efficiency. We believe that FSCCS method exhibits promising potential applications in homogeneous bioassays and study on the molecular interaction and nanoparticle-molecule interaction.
Collapse
Affiliation(s)
- Jinjie Wang
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China.,College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science , 333 Longteng Road, Shanghai, 201620, P.R. China
| | - Xiangyi Huang
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Heng Liu
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Chaoqing Dong
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jicun Ren
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
32
|
Trueb J, Avci O, Sevenler D, Connor JH, Ünlü MS. Robust Visualization and Discrimination of Nanoparticles by Interferometric Imaging. IEEE JOURNAL OF SELECTED TOPICS IN QUANTUM ELECTRONICS : A PUBLICATION OF THE IEEE LASERS AND ELECTRO-OPTICS SOCIETY 2017; 23:6900610. [PMID: 28989270 PMCID: PMC5627618 DOI: 10.1109/jstqe.2016.2639824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Single-molecule and single-nanoparticle biosensors are a growing frontier in diagnostics. Digital biosensors are those which enumerate all specifically immobilized biomolecules or biological nanoparticles, and thereby achieve limits of detection usually beyond the reach of ensemble measurements. Here we review modern optical techniques for single nanoparticle detection and describe the single-particle interferometric reflectance imaging sensor (SP-IRIS). We present challenges associated with reliably detecting faint nanoparticles with SP-IRIS, and describe image acquisition processes and software modifications to address them. Specifically, we describe a image acquisition processing method for the discrimination and accurate counting of nanoparticles that greatly reduces both the number of false positives and false negatives. These engineering improvements are critical steps in the translation of SP-IRIS towards applications in medical diagnostics.
Collapse
Affiliation(s)
- Jacob Trueb
- Department of Mechanical Engineering Boston University, 8 St. Mary's Street, Boston, MA 02215
| | - Oguzhan Avci
- Department of Electrical and Computer Engineering, Boston University, 8 St. Mary's Street, Boston, MA 02215, USA (617-353-5067)
| | - Derin Sevenler
- Department of Biomedical Engineering at Boston University, 44 Cummington Way, Boston MA 02215
| | - John H Connor
- Microbiology Department, Boston University School of Medicine, Boston, MA 02118, USA
| | - M Selim Ünlü
- Department of Electrical and Computer Engineering, Boston University, 8 St. Mary's Street, Boston, MA 02215, USA (617-353-5067)
| |
Collapse
|
33
|
Friedrich R, Block S, Alizadehheidari M, Heider S, Fritzsche J, Esbjörner EK, Westerlund F, Bally M. A nano flow cytometer for single lipid vesicle analysis. LAB ON A CHIP 2017; 17:830-841. [PMID: 28128381 DOI: 10.1039/c6lc01302c] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We present a nanofluidic device for fluorescence-based detection and characterization of small lipid vesicles on a single particle basis. The device works like a nano flow cytometer where individual vesicles are visualized by fluorescence microscopy while passing through parallel nanochannels in a pressure-driven flow. An experiment requires less than 20 μl sample volume to quantify both the vesicle content and the fluorescence signals emitted by individual vesicles. We show that the device can be used to accurately count the number of fluorescent synthetic lipid vesicles down to a vesicle concentration of 170 fM. We also show that the size-distribution of the vesicles can be resolved from their fluorescence intensity distribution after calibration. We demonstrate the applicability of the assay in two different examples. In the first, we use the nanofluidic device to determine the particle concentration in a sample containing cell-derived extracellular vesicles labelled with a lipophilic dye. In the second, we demonstrate that dual-color detection can be used to probe peptide binding to synthetic lipid vesicles; we identify a positive membrane-curvature sensing behavior of an arginine enriched version of the Antennapedia homeodomain peptide penetratin. Altogether, these results illustrate the potential of this nanofluidic-based methodology for characterization and quantification of small biological vesicles and their interactors without ensemble averaging. The device is therefore likely to find use as a quantitative analytical tool in a variety of fields ranging from diagnostics to fundamental biology research. Moreover, our results have potential to facilitate further development of automated lab-on-a-chip devices for vesicle analysis.
Collapse
Affiliation(s)
- Remo Friedrich
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden.
| | - Stephan Block
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden.
| | | | - Susanne Heider
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden. and Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Joachim Fritzsche
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden.
| | - Elin K Esbjörner
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Fredrik Westerlund
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| | - Marta Bally
- Department of Physics, Chalmers University of Technology, Gothenburg, Sweden. and Institut Curie, Centre de Recherche, CNRS, UMR168, Physico-Chimie Curie, Paris, France
| |
Collapse
|
34
|
Affiliation(s)
- Lucas Armbrecht
- Department of Biosystems Science and Engineering, ETH Zurich, CH-8093 Zurich, Switzerland
| | | |
Collapse
|
35
|
Shahin H, Gupta M, Janowska-Wieczorek A, Rozmus W, Tsui YY. Physical characterization of hematopoietic stem cells using multidirectional label-free light scatterings. OPTICS EXPRESS 2016; 24:28877-28888. [PMID: 27958553 DOI: 10.1364/oe.24.028877] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
An experimental setup capable of measuring simultaneous 2D scattered light angular distribution from two directions to study cell morphology without the use of bio-labels was developed. Experiments with hematopoietic stem cells (CD34+ cells) show good agreement with detailed numerical simulations of light scattering. Numerical simulations and computer models of cells are used to identify physical features of cells with the largest scattering cross sections. This allows for determination of size, geometry of the nucleus and distribution of mitochondria in hematopoietic stem cells by means of our label-free method.
Collapse
|
36
|
Agnarsson B, Wayment-Steele HK, Höök F, Kunze A. Monitoring of single and double lipid membrane formation with high spatiotemporal resolution using evanescent light scattering microscopy. NANOSCALE 2016; 8:19219-19223. [PMID: 27845799 DOI: 10.1039/c6nr06726c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Formation and quality of single solid supported lipid membranes and double lipid membranes were investigated with single vesicle resolution using label-free evanescence light scattering microscopy (EvSM). For the formation of double lipid membranes we made use of electrostatic interaction between charged lipids and oppositely charged cations.
Collapse
Affiliation(s)
- B Agnarsson
- Dept of Appl. Physics, Chalmers University of Technology, Göteborg, Sweden.
| | | | - F Höök
- Dept of Appl. Physics, Chalmers University of Technology, Göteborg, Sweden.
| | - A Kunze
- Dept of Appl. Physics, Chalmers University of Technology, Göteborg, Sweden. and Inst of Physical Chemistry, University of Göttingen, Göttingen, Germany
| |
Collapse
|
37
|
Lundgren A, Agnarsson B, Zirbs R, Zhdanov VP, Reimhult E, Höök F. Nonspecific Colloidal-Type Interaction Explains Size-Dependent Specific Binding of Membrane-Targeted Nanoparticles. ACS NANO 2016; 10:9974-9982. [PMID: 27783496 DOI: 10.1021/acsnano.6b04160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Emerging biomedical applications such as molecular imaging and drug delivery often require directed binding of nanoparticles to cell-membrane receptors. The specific apparent affinity of such ligand-functionalized particles is size-dependent, an observation so far solely attributed to multivalent receptor-ligand interaction. We question the universality of this explanation by demonstrating that the binding kinetics also depends on weak, attractive colloidal-type interaction between nanoparticles and a lipid membrane. Applying label-free single-particle imaging, we correlate binding of nanoparticles targeted to a cell-mimetic lipid membrane with the distribution of nontargeted particles freely diffusing close to the membrane interface. This analysis shows that already a weak, kBT-scale attraction present between 50 nm gold nanoparticles and the membrane renders these particles an order of magnitude higher avidity compared to 20 nm particles. A stronger emphasis on nonspecific particle-membrane interaction might thus be required to accurately predict nanoparticle targeting and other similar processes such as cellular uptake of exosomes and viruses.
Collapse
Affiliation(s)
- Anders Lundgren
- Department of Physics, Chalmers University of Technology , Gothenburg 412 96, Sweden
- Department of Nanobiotechnology, University of Natural Resources and Life Sciences , Vienna 1190, Austria
| | - Björn Agnarsson
- Department of Physics, Chalmers University of Technology , Gothenburg 412 96, Sweden
| | - Ronald Zirbs
- Department of Nanobiotechnology, University of Natural Resources and Life Sciences , Vienna 1190, Austria
| | - Vladimir P Zhdanov
- Department of Physics, Chalmers University of Technology , Gothenburg 412 96, Sweden
- Boreskov Institute of Catalysis, Russian Academy of Sciences , Novosibirsk 630090, Russia
| | - Erik Reimhult
- Department of Nanobiotechnology, University of Natural Resources and Life Sciences , Vienna 1190, Austria
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology , Gothenburg 412 96, Sweden
| |
Collapse
|
38
|
Yang CT, Wu L, Liu X, Tran NT, Bai P, Liedberg B, Wang Y, Thierry B. Exploiting Surface-Plasmon-Enhanced Light Scattering for the Design of Ultrasensitive Biosensing Modality. Anal Chem 2016; 88:11924-11930. [PMID: 27934101 DOI: 10.1021/acs.analchem.6b03798] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Development of new detection methodologies and amplification schemes is indispensable for plasmonic biosensors to improve the sensitivity for the detection of trace amounts of analytes. Herein, an ultrasensitive scheme for signal enhancement based on the concept of surface-plasmon-resonance-enhanced light scattering (SP-LS) was validated experimentally and theoretically. The SP-LS of gold nanoparticles' (AuNPs) tags was employed in a sandwich assay for the detection of cardiac troponin I and provided up to 2 orders of magnitude improved sensitivity over conventional AuNPs-enhanced refractometric measurements and 3 orders of magnitude improvement over label-free SPR. Simulations were also performed to provide insights into the physical mechanisms.
Collapse
Affiliation(s)
- Chih-Tsung Yang
- Future Industries Institute, University of South Australia , Mawson Lakes Campus, Mawson Lakes, South Australia 5095, Australia
| | - Lin Wu
- Electronics and Photonics Department, Institute of High Performance Computing, Agency for Science, Technology, and Research (A*STAR) , Singapore 138632
| | - Xiaohu Liu
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University , Singapore 639798
| | - Nhung Thi Tran
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University , Singapore 639798
| | - Ping Bai
- Electronics and Photonics Department, Institute of High Performance Computing, Agency for Science, Technology, and Research (A*STAR) , Singapore 138632
| | - Bo Liedberg
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University , Singapore 639798
| | - Yi Wang
- Center for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University , Singapore 639798.,Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Sciences , Wenzhou, Zhejiang 325001, China.,Institute of Biomaterials and Engineering, Wenzhou Medical University , Wenzhou, Zhejiang 325035, China
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia , Mawson Lakes Campus, Mawson Lakes, South Australia 5095, Australia
| |
Collapse
|
39
|
Rupert DLM, Shelke GV, Emilsson G, Claudio V, Block S, Lässer C, Dahlin A, Lötvall JO, Bally M, Zhdanov VP, Höök F. Dual-Wavelength Surface Plasmon Resonance for Determining the Size and Concentration of Sub-Populations of Extracellular Vesicles. Anal Chem 2016; 88:9980-9988. [PMID: 27644331 DOI: 10.1021/acs.analchem.6b01860] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Accurate concentration determination of subpopulations of extracellular vesicles (EVs), such as exosomes, is of importance both in the context of understanding their fundamental biological role and of potentially using them as disease biomarkers. In principle, this can be achieved by measuring the rate of diffusion-limited mass uptake to a sensor surface modified with a receptor designed to only bind the subpopulation of interest. However, a significant error is introduced if the targeted EV subpopulation has a size, and thus hydrodynamic diffusion coefficient, that differs from the mean size and diffusion coefficient of the whole EV population and/or if the EVs become deformed upon binding to the surface. We here demonstrate a new approach to determine the mean size (or effective film thickness) of bound nanoparticles, in general, and EV subpopulation carrying a marker of interest, in particular. The method is based on operating surface plasmon resonance simultaneously at two wavelengths with different sensing depths and using the ratio of the corresponding responses to extract the particle size on the surface. By estimating in this way the degree of deformation of adsorbed EVs, we markedly improved their bulk concentration determination and showed that EVs carrying the exosomal marker CD63 correspond to not more than around 10% of the EV sample.
Collapse
Affiliation(s)
- Deborah L M Rupert
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| | - Ganesh V Shelke
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg , SE-405 30 Gothenburg, Sweden
| | - Gustav Emilsson
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| | - Virginia Claudio
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| | - Stephan Block
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg , SE-405 30 Gothenburg, Sweden
| | - Andreas Dahlin
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| | - Jan O Lötvall
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg , SE-405 30 Gothenburg, Sweden
| | - Marta Bally
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden.,Institut Curie, Centre de Recherche, CNRS, UMR 168, Physico-Chimie Curie, F-75248 Paris, France
| | - Vladimir P Zhdanov
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden.,Boreskov Institute of Catalysis, Russian Academy of Sciences , 630090 Novosibirsk, Russia
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology , SE-412 96 Gothenburg, Sweden
| |
Collapse
|
40
|
Zhdanov VP, Agnarsson B, Höök F. Kinetics of enzyme-mediated hydrolysis of lipid vesicles. Chem Phys Lett 2016. [DOI: 10.1016/j.cplett.2016.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|