1
|
Gunawardhana L, Moree W, Guo J, Artur C, Womack T, Eriksen JL, Mayerich D. Fast Photostable Expansion Microscopy Using QDots and Deconvolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625167. [PMID: 39651290 PMCID: PMC11623529 DOI: 10.1101/2024.11.25.625167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Expansion microscopy (ExM) enables sub-diffraction imaging by physically expanding labeled tissue samples. This increases the tissue volume relative to the instrument point spread function (PSF), thereby improving the effective resolution by reported factors of 4 - 20X [1, 2]. However, this volume increase dilutes the fluorescence signal, reducing both signal-to noise ratio (SNR) and acquisition speed. This paper proposes and validates a method for mitigating these challenges. We overcame the limitations of ExM by developing a fast photo-stable protocol to enable scalable widefield three-dimensional imaging with ExM. We combined widefield imaging with quantum dots (QDots). Widefield imaging provides a significantly faster acquisition of a single field-of-view (FOV). However, the uncontrolled incoherent illumination induces photobleaching. We mitigated this challenge using QDots, which exhibit a long fluorescence lifetime and improved photostability. First, we developed a protocol for QDot labeling. Next, we utilized widefield imaging to obtain 3D image stacks and applied deconvolution, which is feasible due to reduced scattering in ExM samples. We show that tissue clearing, which is a side-effect of ExM, enables widefield deconvolution, dramatically reducing the acquisition time for three-dimensional images compared to laser scanning microscopy. The proposed QDot labeling protocol is compatible with ExM and provides enhanced photostability compared to traditional fluorescent dyes. Widefield imaging significantly improves SNR and acquisition speed compared to conventional confocal microscopy. Combining widefield imaging with QDot labeling and deconvolution has the potential to be applied to ExM for faster imaging of large three-dimensional samples with improved SNR.
Collapse
|
2
|
Szczypkowski P, Pawlowska M, Lapkiewicz R. 3D super-resolution optical fluctuation imaging with temporal focusing two-photon excitation. BIOMEDICAL OPTICS EXPRESS 2024; 15:4381-4389. [PMID: 39022538 PMCID: PMC11249675 DOI: 10.1364/boe.523430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024]
Abstract
3D super-resolution fluorescence microscopy typically requires sophisticated setups, sample preparation, or long measurements. A notable exception, SOFI, only requires recording a sequence of frames and no hardware modifications whatsoever but being a wide-field method, it faces problems in thick, dense samples. We combine SOFI with temporal focusing two-photon excitation - the wide-field method that is capable of exciting a thin slice in 3D volume. Temporal focusing is simple to implement whenever the excitation path of the microscope can be accessed. The implementation of SOFI is straightforward. By merging these two methods, we obtain super-resolved 3D images of neurons stained with quantum dots. Our approach offers reduced bleaching of out-of-focus fluorescent probes and an improved signal-to-background ratio that can be used when robust resolution improvement is required in thick, dense samples.
Collapse
Affiliation(s)
- Pawel Szczypkowski
- Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, Warsaw 02-093, Poland
| | - Monika Pawlowska
- Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, Warsaw 02-093, Poland
- Nencki Institute of Experimental Biology PAS, Pasteura 3, Warsaw 02-093, Poland
| | - Radek Lapkiewicz
- Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, Warsaw 02-093, Poland
| |
Collapse
|
3
|
Kolossov VL, Kanakaraju K, Sarkar S, Arogundade OH, Kuo CW, Mara NR, Smith AM. Quantum Dot-Fab' Conjugates as Compact Immunolabels for Microtubule Imaging and Cell Classification. ACS NANO 2024; 18:15084-15095. [PMID: 38815170 PMCID: PMC11262708 DOI: 10.1021/acsnano.4c02215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Antibodies and their conjugates of fluorescent labels are widely applied in life sciences research and clinical pathology. Among diverse label types, compact quantum dots (QDs) provide advantages of multispectral multiplexing, bright signals in the deep red and infrared, and low steric hindrance. However, QD-antibody conjugates have random orientation of the antigen-binding domain which may interfere with labeling and are large (20-30 nm) and heterogeneous, which limits penetration into biospecimens. Here, we develop conjugates of compact QDs and Fab' antibody fragments as primary immunolabels. Fab' fragments are conjugated site-specifically through sulfhydryl groups distal to antigen-binding domains, and the multivalent conjugates have small and homogeneous sizes (∼12 nm) near those of full-sized antibodies. Their performance as immunolabels for intracellular antigens is evaluated quantitatively by metrics of microtubule labeling density and connectivity in fixed cells and for cytological identification in fixed brain specimens, comparing results with probes based on spectrally-matched dyes. QD-Fab' conjugates outperformed QD conjugates of full-sized antibodies and could be imaged with bright signals with 1-photon and 2-photon excitation. The results demonstrate a requirement for smaller bioaffinity agents and site-specific orientation for the success of nanomaterial-based labels to enhance penetration in biospecimens and minimize nonspecific staining.
Collapse
Affiliation(s)
- Vladimir L Kolossov
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kaviamuthan Kanakaraju
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Suresh Sarkar
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, Indian Institute of Technology Jodhpur, Rajasthan 342037, India
| | - Opeyemi H Arogundade
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Chia-Wei Kuo
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Nihar R Mara
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Andrew M Smith
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Holonyak Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
- Carle Illinois College of Medicine, Urbana, Illinois 61801, United States
| |
Collapse
|
4
|
Feye J, Matthias J, Fischer A, Rudolph D, Treptow J, Popescu R, Franke J, Exarhos AL, Boekelheide ZA, Gerthsen D, Feldmann C, Roesky PW, Rösch ES. SMART RHESINs-Superparamagnetic Magnetite Architecture Made of Phenolic Resin Hollow Spheres Coated with Eu(III) Containing Silica Nanoparticles for Future Quantitative Magnetic Particle Imaging Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301997. [PMID: 37203272 DOI: 10.1002/smll.202301997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/15/2023] [Indexed: 05/20/2023]
Abstract
Magnetic particle imaging (MPI) is a powerful and rapidly growing tomographic imaging technique that allows for the non-invasive visualization of superparamagnetic nanoparticles (NPs) in living matter. Despite its potential for a wide range of applications, the intrinsic quantitative nature of MPI has not been fully exploited in biological environments. In this study, a novel NP architecture that overcomes this limitation by maintaining a virtually unchanged effective relaxation (Brownian plus Néel) even when immobilized is presented. This superparamagnetic magnetite architecture made of phenolic resin hollow spheres coated with Eu(III) containing silica nanoparticles (SMART RHESINs) was synthesized and studied. Magnetic particle spectroscopy (MPS) measurements confirm their suitability for potential MPI applications. Photobleaching studies show an unexpected photodynamic due to the fluorescence emission peak of the europium ion in combination with the phenol formaldehyde resin (PFR). Cell metabolic activity and proliferation behavior are not affected. Colocalization experiments reveal the distinct accumulation of SMART RHESINs near the Golgi apparatus. Overall, SMART RHESINs show superparamagnetic behavior and special luminescent properties without acute cytotoxicity, making them suitable for bimodal imaging probes for medical use like cancer diagnosis and treatment. SMART RHESINs have the potential to enable quantitative MPS and MPI measurements both in mobile and immobilized environments.
Collapse
Affiliation(s)
- Julia Feye
- Faculty of Engineering, Baden-Württemberg Cooperative State University Karlsruhe, 76133, Karlsruhe, Germany
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Jessica Matthias
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
| | - Alena Fischer
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
| | - David Rudolph
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Jens Treptow
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Radian Popescu
- Laboratory for Electron Microscopy, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Jochen Franke
- Bruker, BioSpin MRI GmbH, Preclinical Imaging Division, 76275, Ettlingen, Germany
| | | | | | - Dagmar Gerthsen
- Laboratory for Electron Microscopy, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Claus Feldmann
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Peter W Roesky
- Institute of Inorganic Chemistry, Karlsruhe Institute of Technology, 76131, Karlsruhe, Germany
| | - Esther S Rösch
- Faculty of Engineering, Baden-Württemberg Cooperative State University Karlsruhe, 76133, Karlsruhe, Germany
| |
Collapse
|
5
|
McFarlane M, Hall NJ, McConnell G. Enhanced fluorescence from semiconductor quantum dot-labelled cells excited at 280 nm. Methods Appl Fluoresc 2022; 10. [PMID: 35203075 DOI: 10.1088/2050-6120/ac5878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/24/2022] [Indexed: 11/12/2022]
Abstract
Semiconductor quantum dots (QDs) have significant advantages over more traditional fluorophores used in fluorescence microscopy including reduced photobleaching, long-term photostability and high quantum yields, but due to limitations in light sources and optics, are often excited far from their optimum excitation wavelengths in the deep-UV. Here, we present a quantitative comparison of the excitation of semiconductor QDs at a wavelength of 280 nm, compared to the longer wavelength of 365 nm, within a cellular environment. We report increased fluorescence intensity and enhanced image quality when using 280 nm excitation compared to 365 nm excitation for cell imaging across multiple datasets, with a highest average fluorescence intensity increase of 3.59-fold. We also find no significant photobleaching of QDs associated with 280 nm excitation and find that on average, ~80% of cells can tolerate exposure to high-intensity 280 nm irradiation over a 6-hour period.
Collapse
Affiliation(s)
- Mollie McFarlane
- Department of Physics , University of Strathclyde, John Anderson Building, 107 Rottenrow, Glasgow, G4 0NG, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Nicholas James Hall
- Department of Physics, University of Strathclyde, John Anderson Building, 107 Rottenrow, Glasgow, G4 0NG, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Gail McConnell
- Department of Physics & Applied Physics, Strathclyde University, John Anderson Building, 107 Rottenrow, Glasgow, G4 0NG, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| |
Collapse
|
6
|
Stelate A, Tihlaříková E, Schwarzerová K, Neděla V, Petrášek J. Correlative Light-Environmental Scanning Electron Microscopy of Plasma Membrane Efflux Carriers of Plant Hormone Auxin. Biomolecules 2021; 11:1407. [PMID: 34680040 PMCID: PMC8533460 DOI: 10.3390/biom11101407] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Fluorescence light microscopy provided convincing evidence for the domain organization of plant plasma membrane (PM) proteins. Both peripheral and integral PM proteins show an inhomogeneous distribution within the PM. However, the size of PM nanodomains and protein clusters is too small to accurately determine their dimensions and nano-organization using routine confocal fluorescence microscopy and super-resolution methods. To overcome this limitation, we have developed a novel correlative light electron microscopy method (CLEM) using total internal reflection fluorescence microscopy (TIRFM) and advanced environmental scanning electron microscopy (A-ESEM). Using this technique, we determined the number of auxin efflux carriers from the PINFORMED (PIN) family (NtPIN3b-GFP) within PM nanodomains of tobacco cell PM ghosts. Protoplasts were attached to coverslips and immunostained with anti-GFP primary antibody and secondary antibody conjugated to fluorochrome and gold nanoparticles. After imaging the nanodomains within the PM with TIRFM, the samples were imaged with A-ESEM without further processing, and quantification of the average number of molecules within the nanodomain was performed. Without requiring any post-fixation and coating procedures, this method allows to study details of the organization of auxin carriers and other plant PM proteins.
Collapse
Affiliation(s)
- Ayoub Stelate
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic; (A.S.); (K.S.)
| | - Eva Tihlaříková
- Institute of Scientific Instruments, Academy of Sciences of the Czech Republic, Královopolská 147, 612 64 Brno, Czech Republic; (E.T.); (V.N.)
| | - Kateřina Schwarzerová
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic; (A.S.); (K.S.)
| | - Vilém Neděla
- Institute of Scientific Instruments, Academy of Sciences of the Czech Republic, Královopolská 147, 612 64 Brno, Czech Republic; (E.T.); (V.N.)
| | - Jan Petrášek
- Department of Experimental Plant Biology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic; (A.S.); (K.S.)
| |
Collapse
|
7
|
Ma L, Geng J, Kolossov VL, Han Z, Pei Y, Lim SJ, Kilian KA, Smith AM. Antibody Self-Assembly Maximizes Cytoplasmic Immunostaining Accuracy of Compact Quantum Dots. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2021; 33:4877-4889. [PMID: 35221487 PMCID: PMC8880911 DOI: 10.1021/acs.chemmater.1c00164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Antibody conjugates of quantum dots (QDs) are expected to transform immunofluorescence staining by expanding multiplexed analysis and improving target quantification. Recently, a new generation of small QDs coated with multidentate polymers has improved QD labeling density in diverse biospecimens, but new challenges prevent their routine use. In particular, these QDs exhibit nonspecific binding to fixed cell nuclei and their antibody conjugates have random attachment orientations. This report describes four high-efficiency chemical approaches to conjugate antibodies to compact QDs. Methods include click chemistry and self-assembly through polyhistidine coordination, both with and without adaptor proteins that directionally orient antibodies. Specific and nonspecific labeling are independently analyzed after application of diverse blocking agent classes, and a new assay is developed to quantitatively measure intracellular labeling density based on microtubule stain connectivity. Results show that protein conjugation to the QD surface is required to simultaneously eliminate nonspecific binding and maintain antigen specificity. Of the four conjugation schemes, polyhistidine-based coordination of adaptor proteins with antibody self-assembly yields the highest intracellular staining density and the simplest conjugation procedure. Therefore, antibody and adaptor protein orientation, in addition to blocking optimization, are important determinants of labeling outcomes, insights that can inform translational development of these more compact nanomaterials.
Collapse
Affiliation(s)
- Liang Ma
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Holonyak Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Junlong Geng
- Department of Bioengineering, Carl R. Woese Institute for Genomic Biology, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Vladimir L Kolossov
- Holonyak Micro and Nanotechnology Laboratory and Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Zhiyuan Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Holonyak Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yi Pei
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Holonyak Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; School of Materials Science and Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sung Jun Lim
- Holonyak Micro and Nanotechnology Laboratory and Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Division of Nanotechnology, Dalseong-Gun 42988, Republic of Korea
| | - Kristopher A Kilian
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Holonyak Micro and Nanotechnology Laboratory and Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; School of Materials Science and Engineering and School of Chemistry, Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Andrew M Smith
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Holonyak Micro and Nanotechnology Laboratory, Department of Bioengineering, Carl R. Woese Institute for Genomic Biology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States; Carle Illinois College of Medicine, Urbana, Illinois 61801, United States
| |
Collapse
|
8
|
Ma RY, Zhang H, Li XF, Zhang CB, Selli C, Tagliavini G, Lam AD, Prost S, Sims AH, Hu HY, Ying T, Wang Z, Ye Z, Pollard JW, Qian BZ. Monocyte-derived macrophages promote breast cancer bone metastasis outgrowth. J Exp Med 2020; 217:e20191820. [PMID: 32780802 PMCID: PMC7596825 DOI: 10.1084/jem.20191820] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/08/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Bone metastasis is the major cause of death in breast cancer. The lack of effective treatment suggests that disease mechanisms are still largely unknown. As a key component of the tumor microenvironment, macrophages promote tumor progression and metastasis. In this study, we found that macrophages are abundant in human and mouse breast cancer bone metastases. Macrophage ablation significantly inhibited bone metastasis growth. Lineage tracking experiments indicated that these macrophages largely derive from Ly6C+CCR2+ inflammatory monocytes. Ablation of the chemokine receptor, CCR2, significantly inhibited bone metastasis outgrowth and prolonged survival. Immunophenotyping identified that bone metastasis-associated macrophages express high levels of CD204 and IL4R. Furthermore, monocyte/macrophage-restricted IL4R ablation significantly inhibited bone metastasis growth, and IL4R null mutant monocytes failed to promote bone metastasis outgrowth. Together, this study identified a subset of monocyte-derived macrophages that promote breast cancer bone metastasis in an IL4R-dependent manner. This suggests that IL4R and macrophage inhibition can have potential therapeutic benefit against breast cancer bone disease.
Collapse
Affiliation(s)
- Ruo-Yu Ma
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Hui Zhang
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY
| | - Xue-Feng Li
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cheng-Bin Zhang
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Cigdem Selli
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Giulia Tagliavini
- Medical Research Council Centre for Inflammation Research, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Alyson D. Lam
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Sandrine Prost
- Medical Research Council Centre for Inflammation Research, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Andrew H. Sims
- Edinburgh Cancer Research UK Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Hai-Yan Hu
- Shanghai Jiao Tong University Affiliated Sixth People`s Hospital, Shanghai, China
| | - Tianlei Ying
- Ministry of Education/National Health Commission/Chinese Academy of Medical Sciences Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhan Wang
- Department of Orthopaedics, Centre for Orthopaedic Research, Orthopaedics Research Institute of Zhejiang University, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopaedics, Centre for Orthopaedic Research, Orthopaedics Research Institute of Zhejiang University, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Jeffrey W. Pollard
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY
| | - Bin-Zhi Qian
- Medical Research Council Centre for Reproductive Health, College of Medicine and Veterinary Medicine, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research UK Centre, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Guangdong Provincial Education Department Key Laboratory of Nano-immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Ehrenberg AJ, Morales DO, Piergies AMH, Li SH, Tejedor JS, Mladinov M, Mulder J, Grinberg LT. A manual multiplex immunofluorescence method for investigating neurodegenerative diseases. J Neurosci Methods 2020; 339:108708. [PMID: 32243897 PMCID: PMC7269157 DOI: 10.1016/j.jneumeth.2020.108708] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/19/2020] [Accepted: 03/27/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neurodegenerative diseases feature stereotypical deposits of protein aggregates that selectively accumulate in vulnerable cells. The ability to simultaneously localize multiple targets in situ is critical to facilitate discovery and validation of pathogenic molecular pathways. Immunostaining methods enable in situ detection of specific targets. Effective stripping of antibodies, allowing successive rounds of staining while maintaining tissue adhesion and antigen integrity, is the main roadblock for enabling multiplex immunostaining in standard labs. Furthermore, stripping techniques require antibody-specific optimization, validation, and quality control steps. NEW METHOD Aiming to create protocols for multiplex localization of neurodegenerative-related processes, without the need for specialized equipment, we evaluated several antibody stripping techniques. We also recommend quality control steps to validate stripping efficacy and ameliorate concerns of cross-reactivity and false positives based on extensive testing. RESULTS A protocol using β-mercaptoethanol and SDS consistently enables reliable antibody stripping across multiple rounds of staining and minimizes the odds of cross-reactivity while preserving tissue adhesion and antigen integrity in human postmortem tissue. COMPARISON WITH EXISTING METHODS Our proposed method is optimal for standard lab settings and shows consistent efficacy despite the intricacies of suboptimal human postmortem tissue and the need to strip markers bound to highly aggregated proteins. Additionally, it incorporates quality control steps to validate antibody stripping. CONCLUSIONS Multiplex immunofluorescence methods for studying neurodegenerative diseases in human postmortem tissue are feasible even in standard laboratories. Nevertheless, evaluation of stripping parameters during optimization and validation phases of experiments is prudent.
Collapse
Affiliation(s)
- Alexander J Ehrenberg
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA; University of California, Berkeley, Helen Wills Neuroscience Institute; Berkeley, CA, USA; University of California, Berkeley, Dept. of Integrative Biology; Berkeley, CA, USA
| | - Dulce Ovando Morales
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA
| | - Antonia M H Piergies
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA
| | - Song Hua Li
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA
| | - Jorge Santos Tejedor
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA; Karolinska Instituet, Department of Neuroscience, Stockholm, Sweden
| | - Mihovil Mladinov
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA
| | - Jan Mulder
- Karolinska Instituet, Department of Neuroscience, Stockholm, Sweden
| | - Lea T Grinberg
- University of California, San Francisco, Memory and Aging Center, Weill Institute for Neurosciences; San Francisco, CA, USA; University of São Paulo School of Medicine, São Paulo, Brazil; University of California, San Francisco, Global Brain Health Institute; San Francisco, CA, USA.
| |
Collapse
|
10
|
Enhancing the Value of Histopathological Assessment of Allograft Biopsy Monitoring. Transplantation 2020; 103:1306-1322. [PMID: 30768568 DOI: 10.1097/tp.0000000000002656] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Traditional histopathological allograft biopsy evaluation provides, within hours, diagnoses, prognostic information, and mechanistic insights into disease processes. However, proponents of an array of alternative monitoring platforms, broadly classified as "invasive" or "noninvasive" depending on whether allograft tissue is needed, question the value proposition of tissue histopathology. The authors explore the pros and cons of current analytical methods relative to the value of traditional and illustrate advancements of next-generation histopathological evaluation of tissue biopsies. We describe the continuing value of traditional histopathological tissue assessment and "next-generation pathology (NGP)," broadly defined as staining/labeling techniques coupled with digital imaging and automated image analysis. Noninvasive imaging and fluid (blood and urine) analyses promote low-risk, global organ assessment, and "molecular" data output, respectively; invasive alternatives promote objective, "mechanistic" insights by creating gene lists with variably increased/decreased expression compared with steady state/baseline. Proponents of alternative approaches contrast their preferred methods with traditional histopathology and: (1) fail to cite the main value of traditional and NGP-retention of spatial and inferred temporal context available for innumerable objective analyses and (2) belie an unfamiliarity with the impact of advances in imaging and software-guided analytics on emerging histopathology practices. Illustrative NGP examples demonstrate the value of multidimensional data that preserve tissue-based spatial and temporal contexts. We outline a path forward for clinical NGP implementation where "software-assisted sign-out" will enable pathologists to conduct objective analyses that can be incorporated into their final reports and improve patient care.
Collapse
|
11
|
Guo J, Artur C, Womack T, Eriksen JL, Mayerich D. Multiplex protein-specific microscopy with ultraviolet surface excitation. BIOMEDICAL OPTICS EXPRESS 2020; 11:99-108. [PMID: 32010503 PMCID: PMC6968765 DOI: 10.1364/boe.11.000099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 06/01/2023]
Abstract
Immunohistochemical techniques, such as immunofluorescence (IF) staining, enable microscopic imaging of local protein expression within tissue samples. Molecular profiling enabled by IF is critical to understanding pathogenesis and is often involved in complex diagnoses. A recent innovation, known as microscopy with ultraviolet surface excitation (MUSE), uses deep ultraviolet (≈280 nm) illumination to excite labels at the tissue surface, providing equivalent images without fixation, embedding, and sectioning. However, MUSE has not yet been integrated into traditional IF pipelines. This limits its application in more complex diagnoses that rely on protein-specific markers. This paper aims to broaden the applicability of MUSE to multiplex immunohistochemistry using quantum dot nanoparticles. We demonstrate the advantages of quantum dot labels for protein-specific MUSE imaging on both paraffin-embedded and intact tissue, significantly expanding MUSE applicability to protein-specific applications. Furthermore, with recent innovations in three-dimensional ultraviolet fluorescence microscopy, this opens the door to three-dimensional IF imaging with quantum dots using ultraviolet excitation.
Collapse
Affiliation(s)
- Jiaming Guo
- University of Houston, Department of Electrical and Computer Engineering, Houston, TX 77004, USA
- These authors contributed equally to this work
| | - Camille Artur
- University of Houston, Department of Electrical and Computer Engineering, Houston, TX 77004, USA
- These authors contributed equally to this work
| | - Tasha Womack
- University of Houston, Department of Pharmacology, Houston, TX 77004, USA
| | - Jason L. Eriksen
- University of Houston, Department of Pharmacology, Houston, TX 77004, USA
| | - David Mayerich
- University of Houston, Department of Electrical and Computer Engineering, Houston, TX 77004, USA
- University of Houston, NSF I/UCRC BRAIN Center, Houston, TX 77004, USA
| |
Collapse
|
12
|
Mehnert JM, Monjazeb AM, Beerthuijzen JMT, Collyar D, Rubinstein L, Harris LN. The Challenge for Development of Valuable Immuno-oncology Biomarkers. Clin Cancer Res 2017; 23:4970-4979. [PMID: 28864725 PMCID: PMC5657536 DOI: 10.1158/1078-0432.ccr-16-3063] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/06/2017] [Accepted: 07/10/2017] [Indexed: 12/25/2022]
Abstract
The development of immunotherapy is an important breakthrough for the treatment of cancer, with antitumor efficacy observed in a wide variety of tumors. To optimize immunotherapy use, approaches must be developed to identify which patients are likely to achieve benefit. To minimize therapeutic toxicities and costs, understanding the ideal choice and sequencing of the numerous immuno-oncology agents available for individual patients is thus critical, but fraught with challenges. The immune tumor microenvironment (TME) is a unique aspect of the response to immuno-oncology agents and measurement of single biomarkers does not adequately capture these complex interactions. Therefore, multiple potential biomarkers are likely needed. Current candidates in this area include PD-L1 expression, CD8+ tumor-infiltrating lymphocytes, tumor mutation load and neoantigen burden, immune-related gene signatures, and multiplex IHC assays that examine the pharmacodynamic and spatial interactions of the TME. The most fruitful investigations are likely to use several techniques to predict response and interrogate mechanisms of resistance. Immuno-oncology biomarker research must employ validated assays to ask focused research questions utilizing clinically annotated tissue collections and biomarker-focused clinical trial designs to investigate specific endpoints. Real-time input from patients and their advocates into biomarker discovery is necessary to ensure that the investigations pursued will improve both clinical outcomes and quality of life. We herein provide a framework of recommendations to guide the search for immuno-oncology biomarkers of value.
Collapse
Affiliation(s)
- Janice M Mehnert
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey.
- Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Arta M Monjazeb
- UC Davis Comprehensive Cancer Center, Sacramento, California
| | | | | | | | | |
Collapse
|