1
|
Zhang O, Lew MD. Single-molecule orientation-localization microscopy: Applications and approaches. Q Rev Biophys 2024; 57:e17. [PMID: 39710866 DOI: 10.1017/s0033583524000167] [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: 12/24/2024]
Abstract
Single-molecule orientation-localization microscopy (SMOLM) builds upon super-resolved localization microscopy by imaging orientations and rotational dynamics of individual molecules in addition to their positions. This added dimensionality provides unparalleled insights into nanoscale biophysical and biochemical processes, including the organization of actin networks, movement of molecular motors, conformations of DNA strands, growth and remodeling of amyloid aggregates, and composition changes within lipid membranes. In this review, we discuss recent innovations in SMOLM and cover three key aspects: (1) biophysical insights enabled by labeling strategies that endow fluorescent probes to bind to targets with orientation specificity; (2) advanced imaging techniques that leverage the physics of light-matter interactions and estimation theory to encode orientation information with high fidelity into microscope images; and (3) computational methods that ensure accurate and precise data analysis and interpretation, even in the presence of severe shot noise. Additionally, we compare labeling approaches, imaging hardware, and publicly available analysis software to aid the community in choosing the best SMOLM implementation for their specific biophysical application. Finally, we highlight future directions for SMOLM, such as the development of probes with improved photostability and specificity, the design of “smart” adaptive hardware, and the use of advanced computational approaches to handle large, complex datasets. This review underscores the significant current and potential impact of SMOLM in deepening our understanding of molecular dynamics, paving the way for future breakthroughs in the fields of biophysics, biochemistry, and materials science.
Collapse
Affiliation(s)
- Oumeng Zhang
- Preston M. Green Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Electrical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Matthew D Lew
- Preston M. Green Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
2
|
Wang H, Kocheril PA, Yang Z, Lee D, Naji N, Du J, Lin LE, Wei L. Room-Temperature Single-Molecule Infrared Imaging and Spectroscopy through Bond-Selective Fluorescence. Angew Chem Int Ed Engl 2024; 63:e202413647. [PMID: 39312677 DOI: 10.1002/anie.202413647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 09/25/2024]
Abstract
Infrared (IR) spectroscopy stands as a workhorse for exploring bond vibrations, offering a wealth of chemical insights across diverse frontiers. With increasing focus on the regime of single molecules, obtaining IR-sensitive information from individual molecules at room temperature would provide essential information about unknown molecular properties. Here, we leverage bond-selective fluorescence microscopy, facilitated by narrowband picosecond mid-IR and near-IR double-resonance excitation, for high-throughput mid-IR structural probing of single molecules. We robustly capture single-molecule images and analyze the combined polarization dependence, vibrational peaks, linewidths, and lifetimes of probe molecules with representative scaffolds. From bulk to single molecules, we find that vibrational lifetimes remain consistent, while linewidths are narrowed by approximately twofold and anisotropy becomes more pronounced. Additionally, unexpected peak shifts from single molecules were observed, attributed to the generation of new modes due to previously unexplored dimerization, supported by quantum chemistry calculations. These findings underscore the importance of infrared analysis on individual single molecules in ambient environments, offering molecular information crucial for functional imaging and the investigation of the fundamental properties and utilities of luminescent molecules.
Collapse
Affiliation(s)
- Haomin Wang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Philip A Kocheril
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Ziguang Yang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Dongkwan Lee
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Noor Naji
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Jiajun Du
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Li-En Lin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| | - Lu Wei
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 91125, Pasadena, California, USA
| |
Collapse
|
3
|
Bunne C, Roohani Y, Rosen Y, Gupta A, Zhang X, Roed M, Alexandrov T, AlQuraishi M, Brennan P, Burkhardt DB, Califano A, Cool J, Dernburg AF, Ewing K, Fox EB, Haury M, Herr AE, Horvitz E, Hsu PD, Jain V, Johnson GR, Kalil T, Kelley DR, Kelley SO, Kreshuk A, Mitchison T, Otte S, Shendure J, Sofroniew NJ, Theis F, Theodoris CV, Upadhyayula S, Valer M, Wang B, Xing E, Yeung-Levy S, Zitnik M, Karaletsos T, Regev A, Lundberg E, Leskovec J, Quake SR. How to build the virtual cell with artificial intelligence: Priorities and opportunities. Cell 2024; 187:7045-7063. [PMID: 39672099 DOI: 10.1016/j.cell.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 12/15/2024]
Abstract
Cells are essential to understanding health and disease, yet traditional models fall short of modeling and simulating their function and behavior. Advances in AI and omics offer groundbreaking opportunities to create an AI virtual cell (AIVC), a multi-scale, multi-modal large-neural-network-based model that can represent and simulate the behavior of molecules, cells, and tissues across diverse states. This Perspective provides a vision on their design and how collaborative efforts to build AIVCs will transform biological research by allowing high-fidelity simulations, accelerating discoveries, and guiding experimental studies, offering new opportunities for understanding cellular functions and fostering interdisciplinary collaborations in open science.
Collapse
Affiliation(s)
- Charlotte Bunne
- Department of Computer Science, Stanford University, Stanford, CA, USA; Genentech, South San Francisco, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; School of Computer and Communication Sciences and School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Yusuf Roohani
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; Arc Institute, Palo Alto, CA, USA
| | - Yanay Rosen
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Ankit Gupta
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xikun Zhang
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marcel Roed
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Theo Alexandrov
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mohammed AlQuraishi
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | | | | | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Chan Zuckerberg Biohub, New York, NY, USA
| | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Abby F Dernburg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kirsty Ewing
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Emily B Fox
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Statistics, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Matthias Haury
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Amy E Herr
- Chan Zuckerberg Biohub, San Francisco, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | | | - Patrick D Hsu
- Arc Institute, Palo Alto, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA; Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | | | | - Shana O Kelley
- Chan Zuckerberg Biohub, Chicago, IL, USA; Northwestern University, Evanston, IL, USA
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Tim Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Stephani Otte
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA; Seattle Hub for Synthetic Biology, Seattle, WA, USA; Howard Hughes Medical Institute, Seattle, WA, USA
| | | | - Fabian Theis
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany; School of Computing, Information and Technology, Technical University of Munich, Munich, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Christina V Theodoris
- Gladstone Institute of Cardiovascular Disease, Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Marc Valer
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, ON, Canada; Vector Institute, Toronto, ON, Canada
| | - Eric Xing
- Carnegie Mellon University, School of Computer Science, Pittsburgh, PA, USA; Mohamed Bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Serena Yeung-Levy
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Aviv Regev
- Genentech, South San Francisco, CA, USA.
| | - Emma Lundberg
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA.
| | - Stephen R Quake
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Applied Physics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Dong J, Kuang W, Shi B, Huang ZL. Microfluidics-based automatic immunofluorescence staining for single-molecule localization microscopy. BIOMEDICAL OPTICS EXPRESS 2024; 15:6893-6904. [PMID: 39679413 PMCID: PMC11640577 DOI: 10.1364/boe.540434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/29/2024] [Accepted: 11/03/2024] [Indexed: 12/17/2024]
Abstract
Full automation of single-molecule localization microscopy (SMLM) is crucial for large-scale and high-throughput cellular imaging. It is well-known that SMLM typically consists of three major steps: immunofluorescence (IF) staining, optical imaging, and image processing. Currently, automation in optical imaging and image processing is almost complete; however, the automation of IF staining has been slow to advance, probably due to its complicated experimental operations. Here we present a low-cost automated method for IF staining, called super-resolution immunofluorescence staining by microfluidics (SRIF-fluidics). This method is suitable for both adherent and suspension cells and supports single-color and multi-color IF staining for SMLM. Our results show that SRIF-fluidics reduces antibody consumption by about 75% and shortens the sample preparation time from 5.6 hours (manual operation) to 2.5 ∼ 4.4 hours, depending on the sample types. Importantly, this method provides a satisfactory consistency of imaging results without sacrificing sample labeling quality. We believe that the method proposed in this paper is a necessary supplement to achieving fully automated SMLM and facilitating high-throughput SMLM in the near future.
Collapse
Affiliation(s)
- Jun Dong
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
| | - Weibing Kuang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bing Shi
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572025, China
| | - Zhen-li Huang
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572025, China
| |
Collapse
|
5
|
Perez D, Dowlatshahi DP, Azaldegui CA, Ansell TB, Dahlberg PD, Moerner WE. Exploring Transient States of PAmKate to Enable Improved Cryogenic Single-Molecule Imaging. J Am Chem Soc 2024; 146:28707-28716. [PMID: 39388715 PMCID: PMC11505402 DOI: 10.1021/jacs.4c05632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
Super-resolved cryogenic correlative light and electron microscopy is a powerful approach which combines the single-molecule specificity and sensitivity of fluorescence imaging with the nanoscale resolution of cryogenic electron tomography. Key to this method is active control over the emissive state of fluorescent labels to ensure sufficient sparsity to localize individual emitters. Recent work has identified fluorescent proteins (FPs) that photoactivate or photoswitch efficiently at cryogenic temperatures, but long on-times due to reduced quantum yield of photobleaching remain a challenge for imaging structures with a high density of localizations. In this work, we explore the photophysical properties of the red photoactivatable FP PAmKate and identify a 2-color process leading to enhanced turn-off of active emitters, improving localization rate. Specifically, after excitation of ground state molecules, we find that a transient state forms with a lifetime of ∼2 ms under cryogenic conditions, which can be bleached by exposure to a second wavelength. We measure the response of the transient state to different wavelengths, demonstrate how this mechanism can be used to improve imaging, and provide a blueprint for the study of other FPs at cryogenic temperatures.
Collapse
Affiliation(s)
- Davis Perez
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Dara P. Dowlatshahi
- Biosciences
Division, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
- Department
of Structural Biology, Stanford University, Stanford, California 94305, United States
| | - Christopher A. Azaldegui
- Division
of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - T. Bertie Ansell
- Division
of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Peter D. Dahlberg
- Division
of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - W. E. Moerner
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
6
|
Sasmal R, Som A, Kumari P, Nair RV, Show S, Barge NS, Pahwa M, Das Saha N, Rao S, Vasu S, Agarwal R, Agasti SS. Supramolecular Guest Exchange in Cucurbit[7]uril for Bioorthogonal Fluorogenic Imaging across the Visible Spectrum. ACS CENTRAL SCIENCE 2024; 10:1945-1959. [PMID: 39463826 PMCID: PMC11503495 DOI: 10.1021/acscentsci.4c01080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024]
Abstract
Fluorogenic probes that unmask fluorescence signals in response to bioorthogonal reactions are a powerful new addition to biological imaging. They can significantly reduce background fluorescence and minimize nonspecific signals, potentially enabling real-time, high-contrast imaging without the need to wash out excess fluorophores. While diverse classes of highly refined synthetic fluorophores are now readily available, integrating them into a bioorthogonal fluorogenic scheme still requires extensive design efforts and customized structural alterations to optimize quenching mechanisms for each specific fluorophore scaffold. Herein, we present a highly generalizable strategy that can produce an efficient bioorthogonal fluorogenic response from essentially any readily available fluorophore without further structural alterations. We designed this strategy based on the macrocyclic cucurbit[7]uril (CB7) host, where a fluorogenic response is achieved by programming a guest exchange reaction within the macrocyclic cavity. We employed this strategy to rapidly create fluorogenic probes across the visible spectrum from diverse fluorophore scaffolds, which enabled no-wash imaging in live cells and tissues with minimal background signal. Finally, we demonstrated that this strategy can be combined with metabolic labeling for fluorogenic detection of metabolically tagged mycobacteria under no-wash conditions and paired with covalently clickable probes for high-contrast super-resolution and multiplexed imaging in cells and tissues.
Collapse
Affiliation(s)
- Ranjan Sasmal
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Arka Som
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Pratibha Kumari
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Resmi V. Nair
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Sushanta Show
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Nisha Sanjay Barge
- Department
of Bioengineering, Indian Institute of Science, Bengaluru 560012, Karnataka India
| | - Meenakshi Pahwa
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Nilanjana Das Saha
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Sushma Rao
- Evolutionary
and Integrative Biology Unit and Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Sheeba Vasu
- Evolutionary
and Integrative Biology Unit and Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| | - Rachit Agarwal
- Department
of Bioengineering, Indian Institute of Science, Bengaluru 560012, Karnataka India
| | - Sarit S. Agasti
- New
Chemistry Unit, Chemistry & Physics of Materials Unit, and School
of Advanced Materials (SAMat), Jawaharlal
Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka 560064, India
| |
Collapse
|
7
|
Bunne C, Roohani Y, Rosen Y, Gupta A, Zhang X, Roed M, Alexandrov T, AlQuraishi M, Brennan P, Burkhardt DB, Califano A, Cool J, Dernburg AF, Ewing K, Fox EB, Haury M, Herr AE, Horvitz E, Hsu PD, Jain V, Johnson GR, Kalil T, Kelley DR, Kelley SO, Kreshuk A, Mitchison T, Otte S, Shendure J, Sofroniew NJ, Theis F, Theodoris CV, Upadhyayula S, Valer M, Wang B, Xing E, Yeung-Levy S, Zitnik M, Karaletsos T, Regev A, Lundberg E, Leskovec J, Quake SR. How to Build the Virtual Cell with Artificial Intelligence: Priorities and Opportunities. ARXIV 2024:arXiv:2409.11654v2. [PMID: 39398201 PMCID: PMC11468656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The cell is arguably the most fundamental unit of life and is central to understanding biology. Accurate modeling of cells is important for this understanding as well as for determining the root causes of disease. Recent advances in artificial intelligence (AI), combined with the ability to generate large-scale experimental data, present novel opportunities to model cells. Here we propose a vision of leveraging advances in AI to construct virtual cells, high-fidelity simulations of cells and cellular systems under different conditions that are directly learned from biological data across measurements and scales. We discuss desired capabilities of such AI Virtual Cells, including generating universal representations of biological entities across scales, and facilitating interpretable in silico experiments to predict and understand their behavior using Virtual Instruments. We further address the challenges, opportunities and requirements to realize this vision including data needs, evaluation strategies, and community standards and engagement to ensure biological accuracy and broad utility. We envision a future where AI Virtual Cells help identify new drug targets, predict cellular responses to perturbations, as well as scale hypothesis exploration. With open science collaborations across the biomedical ecosystem that includes academia, philanthropy, and the biopharma and AI industries, a comprehensive predictive understanding of cell mechanisms and interactions has come into reach.
Collapse
Affiliation(s)
- Charlotte Bunne
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Genentech, South San Francisco, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- School of Computer and Communication Sciences and School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Yusuf Roohani
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Arc Institute, Palo Alto, CA, USA
| | - Yanay Rosen
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Ankit Gupta
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Protein Science, Stockholm, Sweden
| | - Xikun Zhang
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marcel Roed
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Theo Alexandrov
- Department of Pharmacology, University of California, San Diego, CA, USA
- Department of Bioengineering, University of California, San Diego, CA, USA
| | | | | | | | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Chan Zuckerberg Biohub New York, NY, USA
| | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Abby F Dernburg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kirsty Ewing
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Emily B Fox
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, USA
| | - Matthias Haury
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Amy E Herr
- Chan Zuckerberg Biohub San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | | | - Patrick D Hsu
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | | | | - Shana O Kelley
- Chan Zuckerberg Biohub Chicago, IL, USA
- Northwestern University, Evanston, IL, USA
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Tim Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Stephani Otte
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | | | - Fabian Theis
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Christina V Theodoris
- Gladstone Institute of Cardiovascular Disease, Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Marc Valer
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Eric Xing
- Carnegie Mellon University, School of Computer Science, Pittsburgh, PA, USA
- Mohamed Bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Serena Yeung-Levy
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Emma Lundberg
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Protein Science, Stockholm, Sweden
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Stephen R Quake
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| |
Collapse
|
8
|
Zulueta Diaz YDLM, Arnspang EC. Super-resolution microscopy to study membrane nanodomains and transport mechanisms in the plasma membrane. Front Mol Biosci 2024; 11:1455153. [PMID: 39290992 PMCID: PMC11405310 DOI: 10.3389/fmolb.2024.1455153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Biological membranes are complex, heterogeneous, and dynamic systems that play roles in the compartmentalization and protection of cells from the environment. It is still a challenge to elucidate kinetics and real-time transport routes for molecules through biological membranes in live cells. Currently, by developing and employing super-resolution microscopy; increasing evidence indicates channels and transporter nano-organization and dynamics within membranes play an important role in these regulatory mechanisms. Here we review recent advances and discuss the major advantages and disadvantages of using super-resolution microscopy to investigate protein organization and transport within plasma membranes.
Collapse
Affiliation(s)
| | - Eva C Arnspang
- Department of Green Technology, SDU Biotechnology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Wang Q, Nag D, Baldwin SL, Coler RN, McNamara RP. Antibodies as key mediators of protection against Mycobacterium tuberculosis. Front Immunol 2024; 15:1430955. [PMID: 39286260 PMCID: PMC11402706 DOI: 10.3389/fimmu.2024.1430955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Tuberculosis (TB) is caused by infection with the bacterial pathogen Mycobacterium tuberculosis (M.tb) in the respiratory tract. There was an estimated 10.6 million people newly diagnosed with TB, and there were approximately 1.3 million deaths caused by TB in 2022. Although the global prevalence of TB has remained high for decades and is an annual leading cause of death attributed to infectious diseases, only one vaccine, Bacillus Calmette-Guérin (BCG), has been approved so far to prevent/attenuate TB disease. Correlates of protection or immunological mechanisms that are needed to control M.tb remain unknown. The protective role of antibodies after BCG vaccination has also remained largely unclear; however, recent studies have provided evidence for their involvement in protection against disease, as biomarkers for the state of infection, and as potential predictors of outcomes. Interestingly, the antibodies generated post-vaccination with BCG are linked to the activation of innate immune cascades, providing further evidence that antibody effector functions are critical for protection against respiratory pathogens such as M.tb. In this review, we aim to provide current knowledge of antibody application in TB diagnosis, prevention, and treatment. Particularly, this review will focus on 1) The role of antibodies in preventing M.tb infections through preventing Mtb adherence to epithelium, antibody-mediated phagocytosis, and antibody-mediated cellular cytotoxicity; 2) The M.tb-directed antibody response generated after vaccination and how humoral profiles with different glycosylation patterns of these antibodies are linked with protection against the disease state; and 3) How antibody-mediated immunity against M.tb can be further explored as early diagnosis biomarkers and different detection methods to combat the global M.tb burden. Broadening the paradigm of differentiated antibody profiling and antibody-based detection during TB disease progression offers new directions for diagnosis, treatment, and preventative strategies. This approach involves linking the aforementioned humoral responses with the disease state, progression, and clearance.
Collapse
Affiliation(s)
- Qixin Wang
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| | - Deepika Nag
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Susan L. Baldwin
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Rhea N. Coler
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Ryan P. McNamara
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| |
Collapse
|
10
|
Perez D, Dowlatshahi DP, Azaldegui CA, Ansell TB, Dahlberg PD, Moerner WE. Exploring transient states of PAmKate to enable improved cryogenic single-molecule imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590965. [PMID: 38712218 PMCID: PMC11071506 DOI: 10.1101/2024.04.24.590965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Super-resolved cryogenic correlative light and electron microscopy is a powerful approach which combines the single-molecule specificity and sensitivity of fluorescence imaging with the nanoscale resolution of cryogenic electron tomography. Key to this method is active control over the emissive state of fluorescent labels to ensure sufficient sparsity to localize individual emitters. Recent work has identified fluorescent proteins (FPs) which photoactivate or photoswitch efficiently at cryogenic temperatures, but long on-times due to reduced quantum yield of photobleaching remains a challenge for imaging structures with a high density of localizations. In this work, we explore the photophysical properties of the red photoactivatable FP PAmKate and identify a 2-color process leading to enhanced turn-off of active emitters, improving localization rate. Specifically, after excitation of ground state molecules, we find a transient state forms with a lifetime of ~2 ms under cryogenic conditions which can be bleached by exposure to a second wavelength. We measure the response of the transient state to different wavelengths, demonstrate how this mechanism can be used to improve imaging, and provide a blueprint for study of other FPs at cryogenic temperatures.
Collapse
Affiliation(s)
- Davis Perez
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Dara P. Dowlatshahi
- Biosciences Division, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
- Department of Structural Biology, Stanford University, Stanford, California 94305, United States
| | - Christopher A. Azaldegui
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - T. Bertie Ansell
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Peter D. Dahlberg
- Division of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - W. E. Moerner
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
11
|
Kommidi SSR, Atkinson KM, Smith BD. Steric protection of near-infrared fluorescent dyes for enhanced bioimaging. J Mater Chem B 2024; 12:8310-8320. [PMID: 39101969 DOI: 10.1039/d4tb01281j] [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: 08/06/2024]
Abstract
Near-fluorescent (NIR) dyes that absorb and emit light in the wavelength range of 650-1700 nm are well-suited for bioimaging due to the improved image contrast and increased penetration of the long-wavelength light through biological tissue. However, the imaging performance of NIR fluorescent dyes is limited by several inherent photophysical and physicochemical properties including, low fluorescence quantum yield, high chemical and photochemical reactivity, propensity to self-aggregate in water, non-specific association with off-target biological sites, and non-optimal pharmacokinetic profiles in living subjects. In principle, all these drawbacks can be alleviated by steric protection which is a structural process that surrounds the fluorophore with bulky groups that block undesired intermolecular interactions. The literature methods to sterically protect a long-wavelength dye can be separated into two general strategies, non-covalent dye encapsulation and covalent steric appendage. Illustrative examples of each method show how steric protection improves bioimaging performance by providing: (a) increased fluorescence brightness, (b) higher fluorophore ground state stability, (c) decreased photobleaching, and (d) superior pharmacokinetic profile. Some sterically protected dyes are commercially available and further success with future systems will require experts in chemistry, microscopy, cell biology, medical imaging, and clinical medicine to work closely as interdisciplinary teams.
Collapse
Affiliation(s)
| | - Kirk M Atkinson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| | - Bradley D Smith
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, USA.
| |
Collapse
|
12
|
Bhattacharya A, Bagheri N, Boxer SG. A Fluorogenic Pseudoinfection Assay to Probe Transfer and Distribution of Influenza Viral Contents to Target Vesicles. Anal Chem 2024. [PMID: 39086018 DOI: 10.1021/acs.analchem.4c01142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Fusion of enveloped viruses with endosomal membranes and subsequent release of the viral genome into the cytoplasm are crucial to the viral infection cycle. It is often modeled by performing fusion between virus particles and target lipid vesicles. We utilized fluorescence microscopy to characterize the kinetic aspects of the transfer of influenza viral ribonucleoprotein (vRNP) complexes to target vesicles and their spatial distribution within the fused volumes to gain deeper insight into the mechanistic aspects of endosomal escape. The fluorogenic RNA-binding dye QuantiFluor (Promega) was found to be well-suited for direct and sensitive microscopic observation of vRNPs which facilitated background-free detection and kinetic analysis of fusion events on a single particle level. To determine the extent to which the viral contents are transferred to the target vesicles through the fusion pore, we carried out virus-vesicle fusion in a side-by-side fashion. Measurement of the Euclidean distances between the centroids of superlocalized membrane and content dye signals within the fused volumes allowed determination of any symmetry (or the lack thereof) between them as expected in the event of transfer (or the lack thereof) of vRNPs, respectively. We found that, in the case of fusion between viruses and 100 nm target vesicles, ∼39% of the events led to transfer of viral contents to the target vesicles. This methodology provides a rapid, generic, and cell-free way to assess the inhibitory effects of antiviral drugs and therapeutics on the endosomal escape behavior of enveloped viruses.
Collapse
Affiliation(s)
- Ahanjit Bhattacharya
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Stanford Center for Innovation in Global Health, Stanford University, Stanford, California 94305, United States
| | - Nahal Bagheri
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Steven G Boxer
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
13
|
Sun Y, Chen S, Hou Y, Kang SH, Lin JM. Organelle Proximity Analysis for Enhanced Quantification of Mitochondria-Endoplasmic Reticulum Interactions in Single Cells via Super-Resolution Microscopy. Anal Chem 2024; 96:11557-11565. [PMID: 38959297 DOI: 10.1021/acs.analchem.4c02338] [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: 07/05/2024]
Abstract
Mitochondria (MT) and the endoplasmic reticulum (ER) maintain lipid and calcium homeostasis through membrane contacts, particularly MT-ER contacts (MERCs), spanning distances from 10 to 50 nm. However, the variation of different distance ranges and the metabolic factors influencing this variation remain poorly understood. This study employed microfluidic chip-based super-resolution microscopy in conjunction with a Moore-Neighbor tracing-incorporated organelle proximity analysis algorithm. This approach enabled precise three-dimensional localization of single-fluorescence protein molecules within narrow and irregular membrane proximities. It achieved lateral localization precision of less than 20 nm, resulting in a minimum MERC distance of approximately 8 nm in spatial and mean distances across multiple threshold ranges. Additionally, we demonstrated that the MERC distance variation was correlated with MT size rather than ER width. The proportion of each distance range varied significantly after the stimuli. Free cholesterol showed a negative correlation with various distances, while distances of 10-30 nm were associated with glucose, glutamine, and pyruvic acid. Furthermore, the 30-40 nm range was influenced by citric acid. These results underscore the role of advanced subcellular organelle analysis in elucidating the single-molecule behavior and organelle morphology in single-cell studies.
Collapse
Affiliation(s)
- Yucheng Sun
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Shiyu Chen
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Ying Hou
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Seong Ho Kang
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jin-Ming Lin
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
14
|
Liu J, Li Y, Chen T, Zhang F, Xu F. Machine Learning for Single-Molecule Localization Microscopy: From Data Analysis to Quantification. Anal Chem 2024; 96:11103-11114. [PMID: 38946062 DOI: 10.1021/acs.analchem.3c05857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Single-molecule localization microscopy (SMLM) is a versatile tool for realizing nanoscale imaging with visible light and providing unprecedented opportunities to observe bioprocesses. The integration of machine learning with SMLM enhances data analysis by improving efficiency and accuracy. This tutorial aims to provide a comprehensive overview of the data analysis process and theoretical aspects of SMLM, while also highlighting the typical applications of machine learning in this field. By leveraging advanced analytical techniques, SMLM is becoming a powerful quantitative analysis tool for biological research.
Collapse
Affiliation(s)
- Jianli Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yumian Li
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
- School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China
| | - Tailong Chen
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
- School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China
| | - Fa Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Fan Xu
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
- School of Optics and Photonics, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
15
|
Steves MA, He C, Xu K. Single-Molecule Spectroscopy and Super-Resolution Mapping of Physicochemical Parameters in Living Cells. Annu Rev Phys Chem 2024; 75:163-183. [PMID: 38360526 DOI: 10.1146/annurev-physchem-070623-034225] [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: 02/17/2024]
Abstract
By superlocalizing the positions of millions of single molecules over many camera frames, a class of super-resolution fluorescence microscopy methods known as single-molecule localization microscopy (SMLM) has revolutionized how we understand subcellular structures over the past decade. In this review, we highlight emerging studies that transcend the outstanding structural (shape) information offered by SMLM to extract and map physicochemical parameters in living mammalian cells at single-molecule and super-resolution levels. By encoding/decoding high-dimensional information-such as emission and excitation spectra, motion, polarization, fluorescence lifetime, and beyond-for every molecule, and mass accumulating these measurements for millions of molecules, such multidimensional and multifunctional super-resolution approaches open new windows into intracellular architectures and dynamics, as well as their underlying biophysical rules, far beyond the diffraction limit.
Collapse
Affiliation(s)
- Megan A Steves
- Department of Chemistry, University of California, Berkeley, California, USA;
| | - Changdong He
- Department of Chemistry, University of California, Berkeley, California, USA;
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, California, USA;
| |
Collapse
|
16
|
Agyemang E, Gonneville AN, Tiruvadi-Krishnan S, Lamichhane R. Exploring GPCR conformational dynamics using single-molecule fluorescence. Methods 2024; 226:35-48. [PMID: 38604413 PMCID: PMC11098685 DOI: 10.1016/j.ymeth.2024.03.011] [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] [Received: 12/06/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that transmit specific external stimuli into cells by changing their conformation. This conformational change allows them to couple and activate G-proteins to initiate signal transduction. A critical challenge in studying and inferring these structural dynamics arises from the complexity of the cellular environment, including the presence of various endogenous factors. Due to the recent advances in cell-expression systems, membrane-protein purification techniques, and labeling approaches, it is now possible to study the structural dynamics of GPCRs at a single-molecule level both in vitro and in live cells. In this review, we discuss state-of-the-art techniques and strategies for expressing, purifying, and labeling GPCRs in the context of single-molecule research. We also highlight four recent studies that demonstrate the applications of single-molecule microscopy in revealing the dynamics of GPCRs. These techniques are also useful as complementary methods to verify the results obtained from other structural biology tools like cryo-electron microscopy and x-ray crystallography.
Collapse
Affiliation(s)
- Eugene Agyemang
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA
| | - Alyssa N Gonneville
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Sriram Tiruvadi-Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Rajan Lamichhane
- UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN 37996, USA; Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
17
|
Chen H, Yan G, Wen MH, Brooks KN, Zhang Y, Huang PS, Chen TY. Advancements and Practical Considerations for Biophysical Research: Navigating the Challenges and Future of Super-resolution Microscopy. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:331-344. [PMID: 38817319 PMCID: PMC11134610 DOI: 10.1021/cbmi.4c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024]
Abstract
The introduction of super-resolution microscopy (SRM) has significantly advanced our understanding of cellular and molecular dynamics, offering a detailed view previously beyond our reach. Implementing SRM in biophysical research, however, presents numerous challenges. This review addresses the crucial aspects of utilizing SRM effectively, from selecting appropriate fluorophores and preparing samples to analyzing complex data sets. We explore recent technological advancements and methodological improvements that enhance the capabilities of SRM. Emphasizing the integration of SRM with other analytical methods, we aim to overcome inherent limitations and expand the scope of biological insights achievable. By providing a comprehensive guide for choosing the most suitable SRM methods based on specific research objectives, we aim to empower researchers to explore complex biological processes with enhanced precision and clarity, thereby advancing the frontiers of biophysical research.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Guangjie Yan
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Kameron N. Brooks
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Yuteng Zhang
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
18
|
Cha HJ, He C, Glover DJ, Xu K, Clark DS. STORM Super-Resolution Visualization of Self-Assembled γPFD Chaperone Ultrastructures in Methanocaldococcus jannaschii. NANO LETTERS 2024; 24:6078-6083. [PMID: 38723608 PMCID: PMC11117396 DOI: 10.1021/acs.nanolett.4c01043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
Gamma-prefoldin (γPFD), a unique chaperone found in the extremely thermophilic methanogen Methanocaldococcus jannaschii, self-assembles into filaments in vitro, which so far have been observed using transmission electron microscopy and cryo-electron microscopy. Utilizing three-dimensional stochastic optical reconstruction microscopy (3D-STORM), here we achieve ∼20 nm resolution by precisely locating individual fluorescent molecules, hence resolving γPFD ultrastructure both in vitro and in vivo. Through CF647 NHS ester labeling, we first demonstrate the accurate visualization of filaments and bundles with purified γPFD. Next, by implementing immunofluorescence labeling after creating a 3xFLAG-tagged γPFD strain, we successfully visualize γPFD in M. jannaschii cells. Through 3D-STORM and two-color STORM imaging with DNA, we show the widespread distribution of filamentous γPFD structures within the cell. These findings provide valuable insights into the structure and localization of γPFD, opening up possibilities for studying intriguing nanoscale components not only in archaea but also in other microorganisms.
Collapse
Affiliation(s)
- Hee-Jeong Cha
- Department
of Chemical and Biomolecular Engineering, University of California—Berkeley, Berkeley, California 94720, United States
| | - Changdong He
- Department
of Chemistry, University of California—Berkeley, Berkeley, California 94720, United States
| | - Dominic J. Glover
- School
of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ke Xu
- Department
of Chemistry, University of California—Berkeley, Berkeley, California 94720, United States
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, United States
| | - Douglas S. Clark
- Department
of Chemical and Biomolecular Engineering, University of California—Berkeley, Berkeley, California 94720, United States
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, California 94720, United States
| |
Collapse
|
19
|
Nelson T, Vargas-Hernández S, Freire M, Cheng S, Gustavsson AK. Multimodal illumination platform for 3D single-molecule super-resolution imaging throughout mammalian cells. BIOMEDICAL OPTICS EXPRESS 2024; 15:3050-3063. [PMID: 38855669 PMCID: PMC11161355 DOI: 10.1364/boe.521362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 06/11/2024]
Abstract
Single-molecule super-resolution imaging is instrumental in investigating cellular architecture and organization at the nanoscale. Achieving precise 3D nanometric localization when imaging structures throughout mammalian cells, which can be multiple microns thick, requires careful selection of the illumination scheme in order to optimize the fluorescence signal to background ratio (SBR). Thus, an optical platform that combines different wide-field illumination schemes for target-specific SBR optimization would facilitate more precise 3D nanoscale studies of a wide range of cellular structures. Here, we demonstrate a versatile multimodal illumination platform that integrates the sectioning and background reduction capabilities of light sheet illumination with homogeneous, flat-field epi- and TIRF illumination. Using primarily commercially available parts, we combine the fast and convenient switching between illumination modalities with point spread function engineering to enable 3D single-molecule super-resolution imaging throughout mammalian cells. For targets directly at the coverslip, the homogenous intensity profile and excellent sectioning of our flat-field TIRF illumination scheme improves single-molecule data quality by providing low fluorescence background and uniform fluorophore blinking kinetics, fluorescence signal, and localization precision across the entire field of view. The increased contrast achieved with LS illumination, when compared with epi-illumination, makes this illumination modality an excellent alternative when imaging targets that extend throughout the cell. We validate our microscopy platform for improved 3D super-resolution imaging by two-color imaging of paxillin - a protein located in the focal adhesion complex - and actin in human osteosarcoma cells.
Collapse
Affiliation(s)
- Tyler Nelson
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Applied Physics Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Sofía Vargas-Hernández
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Institute of Biosciences & Bioengineering, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Margareth Freire
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Siyang Cheng
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Applied Physics Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Anna-Karin Gustavsson
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
- Institute of Biosciences & Bioengineering, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Biosciences, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Electrical and Computer Engineering, Rice University, 6100 Main St, Houston, TX 77005, USA
- Center for Nanoscale Imaging Sciences, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
20
|
COPELAND CRAIGR, PINTAR ADAML, DIXSON RONALDG, CHANANA ASHISH, SRINIVASAN KARTIK, WESTLY DARONA, ROBERT ILIC B, DAVANCO MARCELOI, STAVIS SAMUELM. Traceable localization enables accurate integration of quantum emitters and photonic structures with high yield. OPTICA QUANTUM 2024; 2:72-84. [PMID: 38741706 PMCID: PMC11089896 DOI: 10.1364/opticaq.502464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/02/2024] [Indexed: 05/16/2024]
Abstract
In a popular integration process for quantum information technologies, localization microscopy of quantum emitters guides lithographic placement of photonic structures. However, a complex coupling of microscopy and lithography errors degrades registration accuracy, severely limiting device performance and process yield. We introduce a methodology to solve this widespread but poorly understood problem. A new foundation of traceable localization enables rapid characterization of lithographic standards and comprehensive calibration of cryogenic microscopes, revealing and correcting latent systematic effects. Of particular concern, we discover that scale factor deviation and complex optical distortion couple to dominate registration errors. These novel results parameterize a process model for integrating quantum dots and bullseye resonators, predicting higher yield by orders of magnitude, depending on the Purcell factor threshold as a quantum performance metric. Our foundational methodology is a key enabler of the lab-to-fab transition of quantum information technologies and has broader implications to cryogenic and correlative microscopy.
Collapse
Affiliation(s)
- CRAIG R. COPELAND
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - ADAM L. PINTAR
- Statistical Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - RONALD G. DIXSON
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - ASHISH CHANANA
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - KARTIK SRINIVASAN
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
- Joint Quantum Institute, NIST/University of Maryland, College Park, Maryland 20742, USA
| | - DARON A. WESTLY
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - B. ROBERT ILIC
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
- CNST NanoFab, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - MARCELO I. DAVANCO
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - SAMUEL M. STAVIS
- Microsystems and Nanotechnology Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| |
Collapse
|
21
|
Feld LG, Boehme SC, Morad V, Sahin Y, Kaul CJ, Dirin DN, Rainò G, Kovalenko MV. Quantifying Förster Resonance Energy Transfer from Single Perovskite Quantum Dots to Organic Dyes. ACS NANO 2024; 18:9997-10007. [PMID: 38547379 PMCID: PMC11008358 DOI: 10.1021/acsnano.3c11359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024]
Abstract
Colloidal quantum dots (QDs) are promising regenerable photoredox catalysts offering broadly tunable redox potentials along with high absorption coefficients. QDs have thus far been examined for various organic transformations, water splitting, and CO2 reduction. Vast opportunities emerge from coupling QDs with other homogeneous catalysts, such as transition metal complexes or organic dyes, into hybrid nanoassemblies exploiting energy transfer (ET), leveraging a large absorption cross-section of QDs and long-lived triplet states of cocatalysts. However, a thorough understanding and further engineering of the complex operational mechanisms of hybrid nanoassemblies require simultaneously controlling the surface chemistry of the QDs and probing dynamics at sufficient spatiotemporal resolution. Here, we probe the ET from single lead halide perovskite QDs, capped by alkylphospholipid ligands, to organic dye molecules employing single-particle photoluminescence spectroscopy with single-photon resolution. We identify a Förster-type ET by spatial, temporal, and photon-photon correlations in the QD and dye emission. Discrete quenching steps in the acceptor emission reveal stochastic photobleaching events of individual organic dyes, allowing a precise quantification of the transfer efficiency, which is >70% for QD-dye complexes with strong donor-acceptor spectral overlap. Our work explores the processes occurring at the QD/molecule interface and demonstrates the feasibility of sensitizing organic photocatalysts with QDs.
Collapse
Affiliation(s)
- Leon G. Feld
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Simon C. Boehme
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
| | - Viktoriia Morad
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
| | - Yesim Sahin
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Christoph J. Kaul
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
| | - Dmitry N. Dirin
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Gabriele Rainò
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, ETH Zürich, CH-8093 Zürich, Switzerland
| | - Maksym V. Kovalenko
- Institute
of Inorganic Chemistry, Department of Chemistry and Applied Biosciences, ETH Zürich, CH-8093 Zürich, Switzerland
- Laboratory
for Thin Films and Photovoltaics, Empa −
Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Dübendorf, Switzerland
- National
Centre of Competence in Research (NCCR) Catalysis, ETH Zürich, CH-8093 Zürich, Switzerland
| |
Collapse
|
22
|
Zhao B, Guan D, Liu J, Zhang X, Xiao S, Zhang Y, Smith BD, Liu Q. Squaraine Dyes Exhibit Spontaneous Fluorescence Blinking That Enables Live-Cell Nanoscopy. NANO LETTERS 2024:10.1021/acs.nanolett.4c00595. [PMID: 38588010 PMCID: PMC11458821 DOI: 10.1021/acs.nanolett.4c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Hampered by their susceptibility to nucleophilic attack and chemical bleaching, electron-deficient squaraine dyes have long been considered unsuitable for biological imaging. This study unveils a surprising twist: in aqueous environments, bleaching is not irreversible but rather a reversible spontaneous quenching process. Leveraging this new discovery, we introduce a novel deep-red squaraine probe tailored for live-cell super-resolution imaging. This probe enables single-molecule localization microscopy (SMLM) under physiological conditions without harmful additives or intense lasers and exhibits spontaneous blinking orchestrated by biological nucleophiles, such as glutathione or hydroxide anion. With a low duty cycle (∼0.1%) and high-emission rate (∼6 × 104 photons/s under 400 W/cm2), the squaraine probe surpasses the benchmark Cy5 dye by 4-fold and Si-rhodamine by a factor of 1.7 times. Live-cell SMLM with the probe reveals intricate structural details of cell membranes, which demonstrates the high potential of squaraine dyes for next-generation super-resolution imaging.
Collapse
Affiliation(s)
- Bingjie Zhao
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Daoming Guan
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Jinyang Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Xuebo Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Shuzhang Xiao
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, Hubei, P. R. China
| | - Yunxiang Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Bradley D. Smith
- Department of Chemistry and Biochemistry, 251 Nieuwland Science Hall, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Qian Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| |
Collapse
|
23
|
Wu Z, Zhang C, Sha J, Jing Z, He J, Bai Y, Wu J, Zhang S, Shi P. Ultrabright Xanthene Fluorescence Probe for Mitochondrial Super-Resolution Imaging. Anal Chem 2024; 96:5134-5142. [PMID: 38507805 DOI: 10.1021/acs.analchem.3c05154] [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: 03/22/2024]
Abstract
Mitochondria are important organelles that provide energy for cellular physiological activities. Changes in their structures may indicate the occurrence of diseases, and the super-resolution imaging of mitochondria is of great significance. However, developing fluorescent probes for mitochondrial super-resolution visualization still remains challenging due to insufficient fluorescence brightness and poor stability. Herein, we rationally synthesized an ultrabright xanthene fluorescence probe Me-hNR for mitochondria-specific super-resolution imaging using structured illumination microscopy (SIM). The rigid structure of Me-hNR provided its ultrahigh fluorescence quantum yield of up to 0.92 and ultrahigh brightness of up to 16,000. Occupying the para-position of the O atom in the xanthene skeleton by utilizing the smallest methyl group ensured its excellent stability. The study of the photophysical process indicated that Me-hNR mainly emitted fluorescence via radiative decay, and nonradiative decay and inter-system crossing were rare due to the slow nonradiative decay rate and large energy gap (ΔEst = 0.55 eV). Owing to these excellent merits, Me-hNR can specifically light up mitochondria at ultralow concentrations down to 5 nM. The unprecedented spatial resolution for mitochondria with an fwhm of 174 nm was also achieved. Therefore, this ultrabright xanthene fluorescence probe has great potential in visualizing the structural changes of mitochondria and revealing the pathogenesis of related diseases using SIM.
Collapse
Affiliation(s)
- Ziyong Wu
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| | - Chuangli Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Jie Sha
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Ziyang Jing
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| | - Jing He
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| | - Yang Bai
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| | - Jiasheng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| | - Pengfei Shi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, College of Medicine, Linyi University, Linyi 276000, P.R. China
| |
Collapse
|
24
|
Clark BS, Silvernail I, Gordon K, Castaneda JF, Morgan AN, Rolband LA, LeBlanc SJ. A practical guide to time-resolved fluorescence microscopy and spectroscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577300. [PMID: 38586000 PMCID: PMC10996486 DOI: 10.1101/2024.01.25.577300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Time-correlated single photon counting (TCSPC) coupled with confocal microscopy is a versatile biophysical tool that enables real-time monitoring of biomolecular dynamics across many timescales. With TCSPC, Fluorescence correlation spectroscopy (FCS) and pulsed interleaved excitation-Förster resonance energy transfer (PIE-FRET) are collected simultaneously on diffusing molecules to extract diffusion characteristics and proximity information. This article is a guide to calibrating FCS and PIE-FRET measurements with several biological samples including liposomes, streptavidin-coated quantum dots, proteins, and nucleic acids for reliable determination of diffusion coefficients and FRET efficiency. The FRET efficiency results are also compared to surface-attached single molecules using fluorescence lifetime imaging microscopy (FLIM-FRET). Combining the methods is a powerful approach to revealing mechanistic details of biological processes and pathways.
Collapse
|
25
|
Zhang J, Zhu Y. Exploiting the Photo-Physical Properties of Metal Halide Perovskite Nanocrystals for Bioimaging. Chembiochem 2024; 25:e202300683. [PMID: 38031246 DOI: 10.1002/cbic.202300683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/01/2023]
Abstract
Perovskite nanomaterials have recently been exploited for bioimaging applications due to their unique photo-physical properties, including high absorbance, good photostability, narrow emissions, and nonlinear optical properties. These attributes outperform conventional fluorescent materials such as organic dyes and metal chalcogenide quantum dots and endow them with the potential to reshape a wide array of bioimaging modalities. Yet, their full potential necessitates a deep grasp of their structure-attribute relationship and strategies for enhancing water stability through surface engineering for meeting the stringent and unique requirements of each individual imaging modality. This review delves into this evolving frontier, highlighting how their distinctive photo-physical properties can be leveraged and optimized for various bioimaging modalities, including visible light imaging, near-infrared imaging, and super-resolution imaging.
Collapse
Affiliation(s)
- Jiahui Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, 30332, USA
| | - Yifan Zhu
- Department of Materials Science and Nanoengineering, Rice University, Houston, Texas, 77005, USA
| |
Collapse
|
26
|
Zhao D, Wang J, Gao L, Huang X, Zhu F, Wang F. Visualizing the intracellular aggregation behavior of gold nanoclusters via structured illumination microscopy and scanning transmission electron microscopy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169153. [PMID: 38072282 DOI: 10.1016/j.scitotenv.2023.169153] [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: 09/23/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 01/18/2024]
Abstract
Given the growing concerns about nanotoxicity, numerous studies have focused on providing mechanistic insights into nanotoxicity by imaging the intracellular fate of nanoparticles. A suitable imaging strategy is necessary to uncover the intracellular behavior of nanoparticles. Although each conventional technique has its own limitations, scanning transmission electron microscopy (STEM) and three-dimensional structured illumination microscopy (3D-SIM) combine the advantages of chemical element mapping, ultrastructural analysis, and cell dynamic tracking. Gold nanoclusters (AuNCs), synthesized using 6-aza-2 thiothymine (ATT) and L-arginine (Arg) as reducing and protecting ligands, referred to as Arg@ATT-AuNCs, have been widely used in biological sensing and imaging, medicine, and catalyst yield. Based on their intrinsic fluorescence and high electron density, Arg@ATT-AuNCs were selected as a model. STEM imaging showed that both the single-particle and aggregated states of Arg@ATT-AuNCs were compartmentally distributed within a single cell. Real-time 3D-SIM imaging showed that the fluorescent Arg@ATT-AuNCs gradually aggregated after being located in the lysosomes of living cells, causing lysosomal damage. The aggregate formation of Arg@ATT-AuNCs was triggered by the low-pH medium, particularly in the lysosomal acidic environment. The proposed dual imaging strategy was verified using other types of AuNCs, which is valuable for studying nano-cell interactions and any associated cytotoxicity, and has the potential to be a useful approach for exploring the interaction of cells with various nanoparticles.
Collapse
Affiliation(s)
- Dan Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, China
| | - Jing Wang
- Institute of Photonic Chips, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Lu Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoyu Huang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fengping Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200052, China.
| | - Fu Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Shanghai 200240, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
27
|
Yan G, Zhang Y, Allamprese A, Brooks KN, Chen W, Yan S, Chen TY. From Molecules to Classrooms: A Comprehensive Guide to Single-Molecule Localization Microscopy. JOURNAL OF CHEMICAL EDUCATION 2024; 101:514-520. [PMID: 39070090 PMCID: PMC11271931 DOI: 10.1021/acs.jchemed.3c00938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Single-molecule localization microscopy (SMLM) has revolutionized our ability to visualize cellular structures, offering unprecedented detail. However, the intricate biophysical principles that underlie SMLM can be daunting for newcomers, particularly undergraduate and graduate students. To address this challenge, we introduce the fundamental concepts of SMLM, providing a solid theoretical foundation. In addition, we have developed an intuitive graphical interface APP that simplifies these core concepts, making them more accessible for students. This APP clarifies how super-resolved images are fitted and highlights the crucial factors determining image quality. Our approach deepens students' understanding of SMLM by combining theoretical instruction with practical learning. This development equips them with the skills to carry out single-molecule super-resolved experiments and explore the microscopic world beyond the diffraction limit.
Collapse
Affiliation(s)
| | | | | | - Kameron N Brooks
- Department of Chemistry, University of Houston, Houston, Texas 77204
| | - Wenkai Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204
| | - Shudan Yan
- Department of Chemistry, University of Houston, Houston, Texas 77204
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, Texas 77204
| |
Collapse
|
28
|
Nelson T, Vargas-Hernández S, freire M, Cheng S, Gustavsson AK. Multimodal illumination platform for 3D single-molecule super-resolution imaging throughout mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.579549. [PMID: 38405960 PMCID: PMC10888752 DOI: 10.1101/2024.02.08.579549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Single-molecule super-resolution imaging is instrumental for investigating cellular architecture and organization at the nanoscale. Achieving precise 3D nanometric localization when imaging structures throughout mammalian cells, which can be multiple microns thick, requires careful selection of the illumination scheme in order to optimize the fluorescence signal to background ratio (SBR). Thus, an optical platform that combines different wide-field illumination schemes for target-specific SBR optimization would facilitate more precise, 3D nanoscale studies of a wide range of cellular structures. Here we demonstrate a versatile multimodal illumination platform that integrates the sectioning and background reduction capabilities of light sheet illumination with homogeneous, flat-field epi-and TIRF illumination. Using primarily commercially available parts, we combine the fast and convenient switching between illumination modalities with point spread function engineering to enable 3D single-molecule super-resolution imaging throughout mammalian cells. For targets directly at the coverslip, the homogenous intensity profile and excellent sectioning of our flat-field TIRF illumination scheme improves single-molecule data quality by providing low fluorescence background and uniform fluorophore blinking kinetics, fluorescence signal, and localization precision across the entire field of view. The increased contrast achieved with LS illumination, when compared with epi-illumination, makes this illumination modality an excellent alternative when imaging targets that extend throughout the cell. We validate our microscopy platform for improved 3D super-resolution imaging by two-color imaging of paxillin - a protein located in the focal adhesion complex - and actin in human osteosarcoma cells.
Collapse
Affiliation(s)
- Tyler Nelson
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Applied Physics Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Sofía Vargas-Hernández
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Systems, Synthetic, and Physical Biology Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Institute of Biosciences & Bioengineering, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Margareth freire
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Siyang Cheng
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Applied Physics Program, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
| | - Anna-Karin Gustavsson
- Department of Chemistry, Rice University, 6100 Main St, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA
- Institute of Biosciences & Bioengineering, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Biosciences, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Electrical and Computer Engineering, Rice University, 6100 Main St, Houston, TX 77005, USA
- Center for Nanoscale Imaging Sciences, Rice University, 6100 Main St, Houston, TX 77005, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
29
|
Mlinac-Jerkovic K, Kalanj-Bognar S, Heffer M, Blažetić S. Methodological Pitfalls of Investigating Lipid Rafts in the Brain: What Are We Still Missing? Biomolecules 2024; 14:156. [PMID: 38397393 PMCID: PMC10886647 DOI: 10.3390/biom14020156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The purpose of this review is to succinctly examine the methodologies used in lipid raft research in the brain and to highlight the drawbacks of some investigative approaches. Lipid rafts are biochemically and biophysically different from the bulk membrane. A specific lipid environment within membrane domains provides a harbor for distinct raftophilic proteins, all of which in concert create a specialized platform orchestrating various cellular processes. Studying lipid rafts has proved to be arduous due to their elusive nature, mobility, and constant dynamic reorganization to meet the cellular needs. Studying neuronal lipid rafts is particularly cumbersome due to the immensely complex regional molecular architecture of the central nervous system. Biochemical fractionation, performed with or without detergents, is still the most widely used method to isolate lipid rafts. However, the differences in solubilization when various detergents are used has exposed a dire need to find more reliable methods to study particular rafts. Biochemical methods need to be complemented with other approaches such as live-cell microscopy, imaging mass spectrometry, and the development of specific non-invasive fluorescent probes to obtain a more complete image of raft dynamics and to study the spatio-temporal expression of rafts in live cells.
Collapse
Affiliation(s)
| | | | - Marija Heffer
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Senka Blažetić
- Department of Biology, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
30
|
Seabury AG, Khodabocus AJ, Kogan IM, Hoy GR, DeSalvo GA, Wustholz KL. Blinking characteristics of organic fluorophores for blink-based multiplexing. Commun Chem 2024; 7:18. [PMID: 38280979 PMCID: PMC10821931 DOI: 10.1038/s42004-024-01106-5] [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: 09/20/2023] [Accepted: 01/12/2024] [Indexed: 01/29/2024] Open
Abstract
Single-molecule fluorescence experiments have transformed our understanding of complex materials and biological systems. Whether single molecules are used to report on their nano-environment or provide for localization, understanding their blinking dynamics (i.e., stochastic fluctuations in emission intensity under continuous illumination) is paramount. We recently demonstrated another use for blinking dynamics called blink-based multiplexing (BBM), where individual emitters are classified using a single excitation laser based on blinking dynamics, rather than color. This study elucidates the structure-activity relationships governing BBM performance in a series of model rhodamine, BODIPY, and anthraquinone fluorophores that undergo different photo-physical and-chemical processes during blinking. Change point detection and multinomial logistic regression analyses show that BBM can leverage spectral fluctuations, electron and proton transfer kinetics, as well as photostability for molecular classification-even within the context of a shared blinking mechanism. In doing so, we demonstrate two- and three-color BBM with ≥ 93% accuracy using spectrally-overlapped fluorophores.
Collapse
Affiliation(s)
| | | | | | - Grayson R Hoy
- Chemistry Department, William & Mary, Williamsburg, VA, USA
| | | | | |
Collapse
|
31
|
Liu J, Zhao B, Zhang X, Guan D, Sun K, Zhang Y, Liu Q. Thiolation for Enhancing Photostability of Fluorophores at the Single-Molecule Level. Angew Chem Int Ed Engl 2024; 63:e202316192. [PMID: 37975636 DOI: 10.1002/anie.202316192] [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] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
Fluorescent probes are essential for single-molecule imaging. However, their application in biological systems is often limited by the short photobleaching lifetime. To overcome this, we developed a novel thiolation strategy for squaraine dyes. By introducing thiolation of the central cyclobutene of squaraine (thio-squaraine), we observed a ≈5-fold increase in photobleaching lifetime. Our single-molecule data analysis attributes this improvement to improved photostability resulting from thiolation. Interestingly, bulk measurements show rapid oxidation of thio-squaraine to its oxo-analogue under irradiation, giving the perception of inferior photostability. This discrepancy between bulk and single-molecule environments can be ascribed to the factors in the latter, including larger intermolecular distances and restricted mobility, which reduce the interactions between a fluorophore and reactive oxygen species produced by other fluorophores, ultimately impacting photobleaching and photoconversion rate. We demonstrate the remarkable performance of thio-squaraine probes in various imaging buffers, such as glucose oxidase with catalase (GLOX) and GLOX+trolox. We successfully employed these photostable probes for single-molecule tracking of CD56 membrane protein and monitoring mitochondria movements in live neurons. CD56 tracking revealed distinct motion states and the corresponding protein fractions. This investigation is expected to propel the development of single-molecule imaging probes, particularly in scenarios where bulk measurements show suboptimal performance.
Collapse
Affiliation(s)
- Jinyang Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Bingjie Zhao
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Xuebo Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Daoming Guan
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Kuangshi Sun
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Yunxiang Zhang
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Qian Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| |
Collapse
|
32
|
Graziotto ME, Kidman CJ, Adair LD, James SA, Harris HH, New EJ. Towards multimodal cellular imaging: optical and X-ray fluorescence. Chem Soc Rev 2023; 52:8295-8318. [PMID: 37910139 DOI: 10.1039/d3cs00509g] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Imaging techniques permit the study of the molecular interactions that underlie health and disease. Each imaging technique collects unique chemical information about the cellular environment. Multimodal imaging, using a single probe that can be detected by multiple imaging modalities, can maximise the information extracted from a single cellular sample by combining the results of different imaging techniques. Of particular interest in biological imaging is the combination of the specificity and sensitivity of optical fluorescence microscopy (OFM) with the quantitative and element-specific nature of X-ray fluorescence microscopy (XFM). Together, these techniques give a greater understanding of how native elements or therapeutics affect the cellular environment. This review focuses on recent studies where both techniques were used in conjunction to study cellular systems, demonstrating the breadth of biological models to which this combination of techniques can be applied and the potential for these techniques to unlock untapped knowledge of disease states.
Collapse
Affiliation(s)
- Marcus E Graziotto
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Clinton J Kidman
- Department of Chemistry, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Liam D Adair
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Simon A James
- Australian Nuclear Science and Technology Organisation, Clayton, Victoria, 3168, Australia
| | - Hugh H Harris
- Department of Chemistry, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Elizabeth J New
- School of Chemistry, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
33
|
He C, Wu CY, Li W, Xu K. Multidimensional Super-Resolution Microscopy Unveils Nanoscale Surface Aggregates in the Aging of FUS Condensates. J Am Chem Soc 2023; 145:24240-24248. [PMID: 37782826 PMCID: PMC10691933 DOI: 10.1021/jacs.3c08674] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The intracellular liquid-liquid phase separation (LLPS) of biomolecules gives rise to condensates that act as membrane-less organelles with vital functions. FUS, an RNA-binding protein, natively forms condensates through LLPS and further provides a model system for the often disease-linked liquid-to-solid transition of biomolecular condensates during aging. However, the mechanism of such maturation processes, as well as the structural and physical properties of the system, remains unclear, partly attributable to difficulties in resolving the internal structures of the micrometer-sized condensates with diffraction-limited optical microscopy. Harnessing a set of multidimensional super-resolution microscopy tools that uniquely map out local physicochemical parameters through single-molecule spectroscopy, here, we uncover nanoscale heterogeneities in FUS condensates and elucidate their evolution over aging. Through spectrally resolved single-molecule localization microscopy (SR-SMLM) with a solvatochromic dye, we unveil distinct hydrophobic nanodomains at the condensate surface. Through SMLM with a fluorogenic amyloid probe, we identify these nanodomains as amyloid aggregates. Through single-molecule displacement/diffusivity mapping (SMdM), we show that such nanoaggregates drastically impede local diffusion. Notably, upon aging or mechanical shears, these nanoaggregates progressively expand on the condensate surface, thus leading to a growing low-diffusivity shell while leaving the condensate interior diffusion-permitting. Together, beyond uncovering fascinating structural arrangements and aging mechanisms in the single-component FUS condensates, the demonstrated synergy of multidimensional super-resolution approaches in this study opens new paths for understanding LLPS systems at the nanoscale.
Collapse
Affiliation(s)
- Changdong He
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Chun Ying Wu
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Wan Li
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| |
Collapse
|
34
|
Hsu CC, Rückel M, Bonn D, Brouwer AM. Super-resolution Fluorescence Imaging of Recycled Polymer Blends via Hydrogen Bond-Assisted Adsorption of a Nile Red Derivative. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:14652-14659. [PMID: 37788122 PMCID: PMC10586370 DOI: 10.1021/acs.langmuir.3c01976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/19/2023] [Indexed: 10/05/2023]
Abstract
A key challenge in the recycling of multilayer plastic films of polyethylene and polyamide, as typically used for food packaging, is to assess and control the phase separation of the two types of polymers in the recycled material, the specifics of which determine the mechanical strength of the recycled material. However, visualizing the polyamide-in-polyethylene domains with conventional fluorescence methods or electron microscopy is challenging. We present a new approach that combines the point accumulation in nanoscale topography (PAINT) super-resolution method with a newly synthesized Nile Red probe (diOHNR) as the fluorescent label. The molecule was modified to undergo a hydrogen bond-assisted interaction with the polyamide phase in the blend due to its two additional hydroxyl groups but preserves the spectral properties of Nile Red. As a result, the localization density of the probe in the PAINT image is 13 times larger at the polyamide phase than at the polyethylene phase, enabling quantitative evaluation of the spatial polyamide/polyethylene distribution down to the nanoscale. The method achieved a spatial resolution of 18.8 nm, and we found that over half of the polyamide particles in a recycled sample were smaller than the optical diffraction limit. Being able to image the blends with nanoscopic resolution can help to optimize the composition and mechanical properties of recycled materials and thus contribute to an increased reuse of plastics.
Collapse
Affiliation(s)
- Chao-Chun Hsu
- van’t
Hoff Institute for Molecular Sciences, University
of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Markus Rückel
- Group
Research, BASF SE, Ludwigshafen D-67056, Germany
| | - Daniel Bonn
- van
der Waals-Zeeman Institute, Institute of
Physics, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Albert M. Brouwer
- van’t
Hoff Institute for Molecular Sciences, University
of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
35
|
Samanta S, Lai K, Wu F, Liu Y, Cai S, Yang X, Qu J, Yang Z. Xanthene, cyanine, oxazine and BODIPY: the four pillars of the fluorophore empire for super-resolution bioimaging. Chem Soc Rev 2023; 52:7197-7261. [PMID: 37743716 DOI: 10.1039/d2cs00905f] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
In the realm of biological research, the invention of super-resolution microscopy (SRM) has enabled the visualization of ultrafine sub-cellular structures and their functions in live cells at the nano-scale level, beyond the diffraction limit, which has opened up a new window for advanced biomedical studies to unravel the complex unknown details of physiological disorders at the sub-cellular level with unprecedented resolution and clarity. However, most of the SRM techniques are highly reliant on the personalized special photophysical features of the fluorophores. In recent times, there has been an unprecedented surge in the development of robust new fluorophore systems with personalized features for various super-resolution imaging techniques. To date, xanthene, cyanine, oxazine and BODIPY cores have been authoritatively utilized as the basic fluorophore units in most of the small-molecule-based organic fluorescent probe designing strategies for SRM owing to their excellent photophysical characteristics and easy synthetic acquiescence. Since the future of next-generation SRM studies will be decided by the availability of advanced fluorescent probes and these four fluorescent building blocks will play an important role in progressive new fluorophore design, there is an urgent need to review the recent advancements in designing fluorophores for different SRM methods based on these fluorescent dye cores. This review article not only includes a comprehensive discussion about the recent developments in designing fluorescent probes for various SRM techniques based on these four important fluorophore building blocks with special emphasis on their effective integration into live cell super-resolution bio-imaging applications but also critically evaluates the background of each of the fluorescent dye cores to highlight their merits and demerits towards developing newer fluorescent probes for SRM.
Collapse
Affiliation(s)
- Soham Samanta
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Kaitao Lai
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Feihu Wu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Yingchao Liu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Songtao Cai
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Xusan Yang
- Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, China
| | - Junle Qu
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Zhigang Yang
- Center for Biomedical Optics and Photonics & Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
36
|
Beatty KE, López CS. Characteristics of genetic tags for correlative light and electron microscopy. Curr Opin Chem Biol 2023; 76:102369. [PMID: 37453163 DOI: 10.1016/j.cbpa.2023.102369] [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] [Received: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Fluorescence microscopy is indispensable in live cell studies of fluorescently-labeled proteins, but has limited resolution and context. Electron microscopy offers high-resolution imaging of cellular ultrastructure, including membranes, organelles, and other nanoscale features. However, identifying proteins by EM remains a substantial challenge. There is potential to combine the strengths of both FM and EM through correlative light and EM (CLEM), and bridging the two modalities enables new discoveries and biological insights. CLEM enables cellular proteins to be observed dynamically, across size scales, and in relationship to sub-cellular structures. A central limitation to using CLEM is the scarcity of methods for labeling proteins with CLEM reporters. This review will describe the characteristics of genetic tags for CLEM that are available today, including fixation-resistant fluorescent proteins, 3,3'-diaminobenzidine (DAB)-based tags, metal-chelating tags, DNA origami tags, and VIP tags.
Collapse
Affiliation(s)
- Kimberly E Beatty
- Department of Chemical Physiology and Biochemistry Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Claudia S López
- Department of Biomedical Engineering Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
37
|
Wang H, Lee D, Cao Y, Bi X, Du J, Miao K, Wei L. Bond-selective fluorescence imaging with single-molecule sensitivity. NATURE PHOTONICS 2023; 17:846-855. [PMID: 38162388 PMCID: PMC10756635 DOI: 10.1038/s41566-023-01243-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/25/2023] [Indexed: 01/03/2024]
Abstract
Bioimaging harnessing optical contrasts and chemical specificity is of vital importance in probing complex biology. Vibrational spectroscopy based on mid-infrared (mid-IR) excitation can reveal rich chemical information about molecular distributions. However, its full potential for bioimaging is hindered by the achievable sensitivity. Here, we report bond selective fluorescence-detected infrared-excited (BonFIRE) spectral microscopy. BonFIRE employs two-photon excitation in the mid-IR and near-IR to upconvert vibrational excitations to electronic states for fluorescence detection, thus encoding vibrational information into fluorescence. The system utilizes tuneable narrowband picosecond pulses to ensure high sensitivity, biocompatibility, and robustness for bond-selective biological interrogations over a wide spectrum of reporter molecules. We demonstrate BonFIRE spectral imaging in both fingerprint and cell-silent spectroscopic windows with single-molecule sensitivity for common fluorescent dyes. We then demonstrate BonFIRE imaging on various intracellular targets in fixed and live cells, neurons, and tissues, with promises for further vibrational multiplexing. For dynamic bioanalysis in living systems, we implement a high-frequency modulation scheme and demonstrate time-lapse BonFIRE microscopy of live HeLa cells. We expect BonFIRE to expand the bioimaging toolbox by providing a new level of bond-specific vibrational information and facilitate functional imaging and sensing for biological investigations.
Collapse
Affiliation(s)
- Haomin Wang
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Dongkwan Lee
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Yulu Cao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Xiaotian Bi
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Jiajun Du
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Kun Miao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Lu Wei
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
38
|
Fernando SI, Martineau JT, Hobson RJ, Vu TN, Baker B, Mueller BD, Menon R, Jorgensen EM, Gerton JM. Simultaneous spectral differentiation of multiple fluorophores in super-resolution imaging using a glass phase plate. OPTICS EXPRESS 2023; 31:33565-33581. [PMID: 37859135 PMCID: PMC10544955 DOI: 10.1364/oe.499929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/10/2023] [Indexed: 10/21/2023]
Abstract
By engineering the point-spread function (PSF) of single molecules, different fluorophore species can be imaged simultaneously and distinguished by their unique PSF patterns. Here, we insert a silicon-dioxide phase plate at the Fourier plane of the detection path of a wide-field fluorescence microscope to produce distinguishable PSFs (X-PSFs) at different wavelengths. We demonstrate that the resulting PSFs can be localized spatially and spectrally using a maximum-likelihood estimation algorithm and can be utilized for hyper-spectral super-resolution microscopy of biological samples. We produced superresolution images of fixed U2OS cells using X-PSFs for dSTORM imaging with simultaneous illumination of up to three fluorophore species. The species were distinguished only by the PSF pattern. We achieved ∼21-nm lateral localization precision (FWHM) and ∼17-nm axial precision (FWHM) with an average of 1,800 - 3,500 photons per PSF and a background as high as 130 - 400 photons per pixel. The modified PSF distinguished fluorescent probes with ∼80 nm separation between spectral peaks.
Collapse
Affiliation(s)
- Sanduni I. Fernando
- University of Utah Department of Physics and Astronomy, 201 James Fletcher Bldg. 115 S. 1400 E Salt Lake City, UT 84112-0830, USA
| | - Jason T. Martineau
- University of Utah Department of Physics and Astronomy, 201 James Fletcher Bldg. 115 S. 1400 E Salt Lake City, UT 84112-0830, USA
| | - Robert J. Hobson
- University of Utah School of Biological Sciences, 257 South 1400 East Salt Lake City, Utah 84112, USA
| | - Thien N. Vu
- University of Utah School of Biological Sciences, 257 South 1400 East Salt Lake City, Utah 84112, USA
| | - Brian Baker
- University of Utah Nanofab 36 S. Wasatch Drive, SMBB Room 2500 Salt Lake City, UT 84112, USA
| | - Brian D. Mueller
- University of Utah School of Biological Sciences, 257 South 1400 East Salt Lake City, Utah 84112, USA
| | - Rajesh Menon
- University of Utah Department of Electrical and Computer Engineering 50 S. Central Campus Drive, MEB Room 2110 Salt Lake City, UT 84112, USA
| | - Erik M. Jorgensen
- University of Utah School of Biological Sciences, 257 South 1400 East Salt Lake City, Utah 84112, USA
| | - Jordan M. Gerton
- University of Utah Department of Physics and Astronomy, 201 James Fletcher Bldg. 115 S. 1400 E Salt Lake City, UT 84112-0830, USA
| |
Collapse
|
39
|
Colson L, Kwon Y, Nam S, Bhandari A, Maya NM, Lu Y, Cho Y. Trends in Single-Molecule Total Internal Reflection Fluorescence Imaging and Their Biological Applications with Lab-on-a-Chip Technology. SENSORS (BASEL, SWITZERLAND) 2023; 23:7691. [PMID: 37765748 PMCID: PMC10537725 DOI: 10.3390/s23187691] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023]
Abstract
Single-molecule imaging technologies, especially those based on fluorescence, have been developed to probe both the equilibrium and dynamic properties of biomolecules at the single-molecular and quantitative levels. In this review, we provide an overview of the state-of-the-art advancements in single-molecule fluorescence imaging techniques. We systematically explore the advanced implementations of in vitro single-molecule imaging techniques using total internal reflection fluorescence (TIRF) microscopy, which is widely accessible. This includes discussions on sample preparation, passivation techniques, data collection and analysis, and biological applications. Furthermore, we delve into the compatibility of microfluidic technology for single-molecule fluorescence imaging, highlighting its potential benefits and challenges. Finally, we summarize the current challenges and prospects of fluorescence-based single-molecule imaging techniques, paving the way for further advancements in this rapidly evolving field.
Collapse
Affiliation(s)
- Louis Colson
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; (L.C.); (A.B.); (N.M.M.); (Y.L.)
| | - Youngeun Kwon
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (S.N.)
| | - Soobin Nam
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (S.N.)
| | - Avinashi Bhandari
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; (L.C.); (A.B.); (N.M.M.); (Y.L.)
| | - Nolberto Martinez Maya
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; (L.C.); (A.B.); (N.M.M.); (Y.L.)
| | - Ying Lu
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; (L.C.); (A.B.); (N.M.M.); (Y.L.)
| | - Yongmin Cho
- Department of Chemical Engineering, Myongji University, Yongin 17058, Republic of Korea; (Y.K.); (S.N.)
| |
Collapse
|
40
|
Procházková M, Kuchovská E, Killinger M, Klepárník K. Novel Förster Resonance Energy Transfer probe with quantum dot for a long-time imaging of active caspases inside individual cells. Anal Chim Acta 2023; 1267:341334. [PMID: 37257963 DOI: 10.1016/j.aca.2023.341334] [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: 12/19/2022] [Revised: 02/28/2023] [Accepted: 05/07/2023] [Indexed: 06/02/2023]
Abstract
With the goal to investigate biological phenomena at a single-cell level, we designed, synthesized and tested a molecular probe based on Förster resonance energy transfer (FRET) between a highly luminescent quantum dot (QD) as a donor and a fluorophore or fluorescence quencher as an acceptor linked by a specific peptide. In principle, QD luminescence, effectively dissipated in the probe, is switched on after the cleavage of the peptide by a protease and the release of the quencher. We proposed a novel synthesis strategy of a probe. A two-step synthesis consists of: (i) Conjugation of CdTe QDs functionalized by -COOH groups of succinic acid on the nanoparticle surface with the designed specific peptide (GTADVEDTSC) using a ligand-exchange approach; (ii) A fast, high-yield reaction of amine-reactive succinimidyl group on the BHQ-2 quencher with N-terminal of the peptide. This way, any crosslinking between individual nanoparticles and any nonspecific conjugation bonds are excluded. The analysis of the product after the first step proved a high reaction yield and nearly no occurrence of unreacted QDs, a prerequisite of the specificity of our luminescent probe. Its parameters evaluated as Michaelis-Menten description of enzymatic kinetics are similar to products published by other groups. Our research is focused on the fluorescence microscopy analyses of biologically active molecules, such as proteolytic active caspases, playing important roles in cell signaling regulations in normal and diseased states. Consequently, they are attractive targets for clinical diagnosis and medical therapy. The ultimate goal of our work was to synthesize a new QD luminescent probe for a long-time quantitative monitoring of active caspase-3/7 distribution in apoptotic osteoblastic MC3T3-E1 cells treated with camptothecin. As a result of comparison, our synthetized luminescent probe provides longer imaging times of caspases than commercial products. The probe proved the stability of the luminescence signal inside cells for more than 14 days.
Collapse
Affiliation(s)
- Markéta Procházková
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, 602 00, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic.
| | - Eliška Kuchovská
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, 602 00, Brno, Czech Republic.
| | - Michael Killinger
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, 602 00, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kotlářská 267/2, 611 37, Brno, Czech Republic.
| | - Karel Klepárník
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, 602 00, Brno, Czech Republic.
| |
Collapse
|
41
|
He C, Wu CY, Li W, Xu K. Multidimensional super-resolution microscopy unveils nanoscale surface aggregates in the aging of FUS condensates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548239. [PMID: 37503034 PMCID: PMC10369965 DOI: 10.1101/2023.07.12.548239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The intracellular liquid-liquid phase separation (LLPS) of biomolecules gives rise to condensates that act as membrane-less organelles with vital functions. FUS, an RNA-binding protein, natively forms condensates through LLPS and further provides a model system for the often disease-linked liquid-to-solid transition of biomolecular condensates during aging. However, the mechanism of such maturation processes, as well as the structural and physical properties of the system, remain unclear, partly attributable to difficulties in resolving the internal structures of the micrometer-sized condensates with diffraction-limited optical microscopy. Harnessing a set of multidimensional super-resolution microscopy tools that uniquely map out local physicochemical parameters through single-molecule spectroscopy, here we uncover nanoscale heterogeneities in the aging process of FUS condensates. Through spectrally resolved single-molecule localization microscopy (SR-SMLM) with a solvatochromic dye, we unveil distinct hydrophobic nanodomains at the condensate surface. Through SMLM with a fluorogenic amyloid probe, we identify these nanodomains as amyloid aggregates. Through single-molecule displacement/diffusivity mapping (SM d M), we show that such nanoaggregates drastically impede local diffusion. Notably, upon aging or mechanical shears, these nanoaggregates progressively expand on the condensate surface, thus leading to a growing low-diffusivity shell while leaving the condensate interior diffusion-permitting. Together, beyond uncovering fascinating nanoscale structural arrangements and aging mechanisms in the single-component FUS condensates, the demonstrated synergy of multidimensional super-resolution approaches in this study opens new paths for understanding LLPS systems.
Collapse
|
42
|
Rathi P, Gupta P, Debnath A, Baldi H, Wang Y, Gupta R, Raman B, Singamaneni S. Plasmon-Enhanced Expansion Microscopy. NANO LETTERS 2023. [PMID: 37307329 DOI: 10.1021/acs.nanolett.3c01256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Expansion microscopy (ExM) is a rapidly emerging super-resolution microscopy technique that involves isotropic expansion of biological samples to improve spatial resolution. However, fluorescence signal dilution due to volumetric expansion is a hindrance to the widespread application of ExM. Here, we introduce plasmon-enhanced expansion microscopy (p-ExM) by harnessing an ultrabright fluorescent nanoconstruct, called plasmonic-fluor (PF), as a nanolabel. The unique structure of PFs renders nearly 15000-fold brighter fluorescence signal intensity and higher fluorescence retention following the ExM protocol (nearly 76%) compared to their conventional counterparts (<16% for IR-650). Individual PFs can be easily imaged using conventional fluorescence microscopes, making them excellent "digital" labels for ExM. We demonstrate that p-ExM enables improved tracing and decrypting of neural networks labeled with PFs, as evidenced by improved quantification of morphological markers (nearly a 2.5-fold increase in number of neurite terminal points). Overall, p-ExM complements the existing ExM techniques for probing structure-function relationships of various biological systems.
Collapse
Affiliation(s)
- Priya Rathi
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Prashant Gupta
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Avishek Debnath
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Harsh Baldi
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Yixuan Wang
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Baranidharan Raman
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, and Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
43
|
Almahayni K, Nestola G, Spiekermann M, Möckl L. Simple, Economic, and Robust Rail-Based Setup for Super-Resolution Localization Microscopy. J Phys Chem A 2023; 127:4553-4560. [PMID: 37163339 DOI: 10.1021/acs.jpca.3c01351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Research during the past 2 decades has showcased the power of single-molecule localization microscopy (SMLM) as a tool for exploring the nanoworld. However, SMLM systems are typically available in specialized laboratories and imaging facilities, owing to their expensiveness as well as complex assembly and alignment procedure. Here, we lay out the blueprint of a sturdy, rail-based, cost-efficient, multicolor SMLM setup that is easy to construct and align in service of simplifying the accessibility of SMLM. We characterize the optical properties of the design and assess its capabilities, robustness, and stability. The performance of the system is assayed using super-resolution imaging of biological samples. We believe that this design will make SMLM more affordable and broaden its availability.
Collapse
Affiliation(s)
- Karim Almahayni
- Max Planck Institute for the Science of Light, Staudtstr. 2, 91058 Erlangen, Germany
- Department of Physics, Friedrich-Alexander-University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Gianluca Nestola
- Max Planck Institute for the Science of Light, Staudtstr. 2, 91058 Erlangen, Germany
| | - Malte Spiekermann
- Max Planck Institute for the Science of Light, Staudtstr. 2, 91058 Erlangen, Germany
| | - Leonhard Möckl
- Max Planck Institute for the Science of Light, Staudtstr. 2, 91058 Erlangen, Germany
| |
Collapse
|
44
|
Hoy GR, DeSalvo GA, Haile SH, Smith EN, Wustholz KL. Rapid, Accurate Classification of Single Emitters in Various Conditions and Environments for Blinking-Based Multiplexing. J Phys Chem A 2023; 127:3518-3525. [PMID: 37023466 DOI: 10.1021/acs.jpca.3c00917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Although single-molecule imaging is widely applied in biology and materials science, most studies are limited by their reliance on spectrally distinct fluorescent probes. We recently introduced blinking-based multiplexing (BBM), a simple approach to differentiate spectrally overlapped single emitters based solely on their intrinsic blinking dynamics. The original proof-of-concept study implemented two methods for emitter classification: an empirically derived metric and a deep learning algorithm, both of which have significant drawbacks. Here, a multinomial logistic regression (LR) classification is applied to rhodamine 6G (R6G) and CdSe/ZnS quantum dots (QDs) in various experimental conditions (i.e., excitation power and bin time) and environments (i.e., glass versus polymer). We demonstrate that LR analysis is rapid and generalizable, and classification accuracies of 95% are routinely observed, even within a complex polymer environment where multiple factors contribute to blinking heterogeneity. In doing so, this study (1) reveals the experimental conditions (i.e., Pexc = 1.2 μW and tbin = 10 ms) that optimize BBM for QD and R6G and (2) demonstrates that BBM via multinomial LR can accurately classify both emitter and environment, opening the door to new opportunities in single-molecule imaging.
Collapse
Affiliation(s)
- Grayson R Hoy
- Department of Chemistry, William & Mary, P.O. Box 8795, Williamsburg, Virginia 23187, United States
| | - Grace A DeSalvo
- Department of Chemistry, William & Mary, P.O. Box 8795, Williamsburg, Virginia 23187, United States
| | - Sophia H Haile
- Department of Chemistry, William & Mary, P.O. Box 8795, Williamsburg, Virginia 23187, United States
| | - Emma N Smith
- Department of Chemistry, William & Mary, P.O. Box 8795, Williamsburg, Virginia 23187, United States
| | - Kristin L Wustholz
- Department of Chemistry, William & Mary, P.O. Box 8795, Williamsburg, Virginia 23187, United States
| |
Collapse
|
45
|
Scalisi S, Pisignano D, Cella Zanacchi F. Single-molecule localization microscopy goes quantitative. Microsc Res Tech 2023; 86:494-504. [PMID: 36601697 DOI: 10.1002/jemt.24281] [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: 11/15/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023]
Abstract
In the last few years, single-molecule localization (SMLM) techniques have been used to address biological questions in different research fields. More recently, super-resolution has also been proposed as a quantitative tool for quantifying protein copy numbers at the nanoscale level. In this scenario, quantitative approaches, mainly based on stepwise photobleaching and quantitative SMLM assisted by calibration standards, offer an exquisite tool for investigating protein complexes. This primer focuses on the basic concepts behind quantitative super-resolution microscopy, also providing strategies to overcome the technical hurdles that could limit their application.
Collapse
Affiliation(s)
- Silvia Scalisi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Dario Pisignano
- Dipartimento di Fisica "E. Fermi", Università di Pisa, Pisa, Italy
| | - Francesca Cella Zanacchi
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
- Dipartimento di Fisica "E. Fermi", Università di Pisa, Pisa, Italy
| |
Collapse
|
46
|
Sartor A, Dahlberg PD, Perez D, Moerner WE. Characterization of mApple as a Red Fluorescent Protein for Cryogenic Single-Molecule Imaging with Turn-Off and Turn-On Active Control Mechanisms. J Phys Chem B 2023; 127:2690-2700. [PMID: 36943356 PMCID: PMC10069424 DOI: 10.1021/acs.jpcb.2c08995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/01/2023] [Indexed: 03/23/2023]
Abstract
Single-molecule superresolution microscopy is a powerful tool for the study of biological structures on size scales smaller than the optical diffraction limit. Imaging samples at cryogenic temperatures (77 K) reduces the quantum yield of photobleaching for many fluorescent labels, yielding localization precisions below 10 nm. Cryogenic imaging further enables correlation with cryogenic electron tomography. A key limitation in applying methods such as PALM and STORM to samples maintained at 77 K is the limited number of fluorophores known to undergo efficient turn-on and turn-off mechanisms necessary to control the sparsity of active emitters. We find that mApple, a red-emitting fluorescent protein, undergoes a novel turn-off mechanism in response to simultaneous illumination with two colors of light. This turn-off mechanism enables localization of many individual molecules in initially bright samples, but the final density of localizable emitters is limited by relatively inefficient turn-on (photoactivation). Bulk excitation and emission spectroscopy shows that mApple has access to two distinct emissive states as well as dark states accessible optically or through changes in pH. The bright and stable emission of mApple enables widefield collection of single-molecule emission spectra, which highlight the complex nature and environmental sensitivity of states observed in red fluorescent proteins.
Collapse
Affiliation(s)
- Annina
M. Sartor
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Peter D. Dahlberg
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Division
of CryoEM and Bioimaging, Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Davis Perez
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - W. E. Moerner
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
47
|
Kozhina E, Bedin S, Martynov A, Andreev S, Piryazev A, Grigoriev Y, Gorbunova Y, Naumov A. Ultrasensitive Optical Fingerprinting of Biorelevant Molecules by Means of SERS-Mapping on Nanostructured Metasurfaces. BIOSENSORS 2022; 13:46. [PMID: 36671881 PMCID: PMC9855407 DOI: 10.3390/bios13010046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
The most relevant technique for portable (on-chip) sensors is Surface Enhanced Raman Scattering (SERS). This strategy crashes in the case of large (biorelevant) molecules and nano-objects, whose SERS spectra are irreproducible for "homeopathic" concentrations. We suggested solving this problem by SERS-mapping. We analyzed the distributions of SERS parameters for relatively "small" (malachite green (MG)) and "large" (phthalocyanine, H2Pc*) molecules. While fluctuations of spectra for "small" MG were negligible, noticeable distribution of spectra was observed for "large" H2Pc*. We show that the latter is due to a random arrangement of molecules with respect to "hot spot" areas, which have limited sizes, thus amplifying the lines corresponding to vibrations of different molecule parts. We have developed a method for engineering low-cost SERS substrates optimized for the best enhancement efficiency and a measurement protocol to obtain a reliable Raman spectrum, even for a countable number of large molecules randomly distributed over the substrate.
Collapse
Affiliation(s)
- Elizaveta Kozhina
- Laboratory of Plasmonics, Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, bld. 1, 121205 Moscow, Russia
- Department of Advanced Photonics and Sensorics, Lebedev Physical Institute RAS, Troitsk Branch, Fizicheskaya Str. 11, 108840 Moscow, Troitsk, Russia
| | - Sergey Bedin
- Department of Advanced Photonics and Sensorics, Lebedev Physical Institute RAS, Troitsk Branch, Fizicheskaya Str. 11, 108840 Moscow, Troitsk, Russia
- Laboratory of Physics of Advanced Materials and Nanostructures, Moscow State Pedagogical University, Malaya Pirogovskaya St. 1-1, 119991 Moscow, Russia
- Laboratory for the Growth of Thin Films and Inorganic Nanostructures Center of Crystallography and Photonics of RAS, Leninskiy Prosp. 59, 119333 Moscow, Russia
| | - Alexander Martynov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Leninskiy Prosp., 31 Building 4, 119071 Moscow, Russia
| | - Stepan Andreev
- Laboratory of Physics of Advanced Materials and Nanostructures, Moscow State Pedagogical University, Malaya Pirogovskaya St. 1-1, 119991 Moscow, Russia
| | - Alexey Piryazev
- Department of Chemistry, Moscow State University, Leninskie Gory, 1, 119991 Moscow, Russia
- Research Center of Genetics and Life Sciences, Research Direction–Biomaterials, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
| | - Yuri Grigoriev
- Laboratory for the Growth of Thin Films and Inorganic Nanostructures Center of Crystallography and Photonics of RAS, Leninskiy Prosp. 59, 119333 Moscow, Russia
| | - Yulia Gorbunova
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Leninskiy Prosp., 31 Building 4, 119071 Moscow, Russia
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, Leninskiy Prosp., 31, 119991 Moscow, Russia
| | - Andrey Naumov
- Department of Advanced Photonics and Sensorics, Lebedev Physical Institute RAS, Troitsk Branch, Fizicheskaya Str. 11, 108840 Moscow, Troitsk, Russia
- Laboratory of Physics of Advanced Materials and Nanostructures, Moscow State Pedagogical University, Malaya Pirogovskaya St. 1-1, 119991 Moscow, Russia
- Laboratory for Spectroscopy of Electronic Spectra of Molecules, Institute for Spectroscopy RAS, Fizicheskaya Str. 5, 108840 Moscow, Troitsk, Russia
| |
Collapse
|
48
|
Pramanik SK, Sreedharan S, Tiwari R, Dutta S, Kandoth N, Barman S, Aderinto SO, Chattopadhyay S, Das A, Thomas JA. Nanoparticles for super-resolution microscopy: intracellular delivery and molecular targeting. Chem Soc Rev 2022; 51:9882-9916. [PMID: 36420611 DOI: 10.1039/d1cs00605c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Following an overview of the approaches and techniques used to acheive super-resolution microscopy, this review presents the advantages supplied by nanoparticle based probes for these applications. The various clases of nanoparticles that have been developed toward these goals are then critically described and these discussions are illustrated with a variety of examples from the recent literature.
Collapse
Affiliation(s)
- Sumit Kumar Pramanik
- CSIR - Central Salt and Marine Chemicals Research Institute, Gijubhai Badheka Marg, Bhavnagar, Gujarat 364002, India.
| | - Sreejesh Sreedharan
- Human Science Research Centre, University of Derby, Kedleston road, DE22 1GB, UK
| | - Rajeshwari Tiwari
- CSIR - Central Salt and Marine Chemicals Research Institute, Gijubhai Badheka Marg, Bhavnagar, Gujarat 364002, India.
| | - Sourav Dutta
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, West Bengal, India.
| | - Noufal Kandoth
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, West Bengal, India.
| | - Surajit Barman
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, West Bengal, India.
| | - Stephen O Aderinto
- Department of Chemistry, University of Sheffield, Western Bank, Sheffield, S3 7HF, UK.
| | - Samit Chattopadhyay
- Department of Biological Sciences, BITS-Pilani, K K Birla Goa Campus, NH 17B, Zuarinagar, Goa 403726, India.
| | - Amitava Das
- Department of Chemical Sciences and Centre for Advanced Functional Materials, Indian Institute of Science Education and Research, Kolkata, West Bengal, India.
| | - Jim A Thomas
- Department of Chemistry, University of Sheffield, Western Bank, Sheffield, S3 7HF, UK.
| |
Collapse
|
49
|
Rajeev P, Singh N, Kechkar A, Butler C, Ramanan N, Sibarita JB, Jose M, Nair D. Nanoscale regulation of Ca2+ dependent phase transitions and real-time dynamics of SAP97/hDLG. Nat Commun 2022; 13:4236. [PMID: 35869063 PMCID: PMC9307800 DOI: 10.1038/s41467-022-31912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 07/08/2022] [Indexed: 11/20/2022] Open
Abstract
Synapse associated protein-97/Human Disk Large (SAP97/hDLG) is a conserved, alternatively spliced, modular, scaffolding protein critical in regulating the molecular organization of cell-cell junctions in vertebrates. We confirm that the molecular determinants of first order phase transition of SAP97/hDLG is controlled by morpho-functional changes in its nanoscale organization. Furthermore, the nanoscale molecular signatures of these signalling islands and phase transitions are altered in response to changes in cytosolic Ca2+. Additionally, exchange kinetics of alternatively spliced isoforms of the intrinsically disordered region in SAP97/hDLG C-terminus shows differential sensitivities to Ca2+ bound Calmodulin, affirming that the molecular signatures of local phase transitions of SAP97/hDLG depends on their nanoscale heterogeneity and compositionality of isoforms. SAP97/hDLG is a ubiquitous, alternatively spliced, and conserved modular scaffolding protein involved in the organization cell junctions and excitatory synapses. Here, authors confirm that SAP97/hDLG condenses in to nanosized molecular domains in both heterologous cells and hippocampal pyramidal neurons. Authors demonstrate that in vivo and in vitro condensation, molecular signatures of nanoscale condensates and exchange kinetics of SAP97/hDLG is modulated by the local availability of alternatively spliced isoforms. Additionally, SAP97/hDLG isoforms exhibits a differential sensitivity to Ca2+ bound Calmodulin, resulting in altered properties of nanocondensates and their real-time regulation
Collapse
|
50
|
Monaghan JW, O'Dell ZJ, Sridhar S, Paranzino B, Sundaresan V, Willets KA. Calcite-Assisted Localization and Kinetics (CLocK) Microscopy. J Phys Chem Lett 2022; 13:10527-10533. [PMID: 36342334 DOI: 10.1021/acs.jpclett.2c03028] [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: 06/16/2023]
Abstract
Localization-based super-resolution imaging techniques have improved the spatial resolution of optical microscopy well below the diffraction limit, yet encoding additional information into super-resolved images, such as anisotropy and orientation, remains a challenge. Here we introduce calcite-assisted localization and kinetics (CLocK) microscopy, a multiparameter super-resolution imaging technique easily integrated into any existing optical microscope setup at low cost and with straightforward analysis. By placing a rotating calcite crystal in the infinity space of an optical microscope, CLocK microscopy provides immediate polarization and orientation information while maintaining the ability to localize an emitter/scatterer with <10 nm resolution. Further, kinetic information an order of magnitude shorter than the integration time of the camera is encoded in the unique point spread function of a CLocK image, allowing for new mechanistic insight into dynamic processes such as single-nanoparticle dissolution and single-molecule surface-enhanced Raman scattering.
Collapse
Affiliation(s)
- Joseph W Monaghan
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania19122, United States
| | - Zachary J O'Dell
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania19122, United States
| | - Sanjay Sridhar
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania19122, United States
| | - Bianca Paranzino
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania19122, United States
| | - Vignesh Sundaresan
- Department of Chemistry and Biochemistry, University of Mississippi, University, Mississippi38677, United States
| | - Katherine A Willets
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania19122, United States
| |
Collapse
|