1
|
Senarathne DS, Shahu L, Lu HP. Probing the Epidermal Growth Factor Receptor under Piconewton Mechanical Compressive Force Manipulations. J Phys Chem B 2025. [PMID: 40423669 DOI: 10.1021/acs.jpcb.5c00800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Studying the relationship among protein structure, dynamics, and function under external compressive forces offers valuable insights. While extensive research has focused on manipulating protein dynamics and ligand-receptor interactions under pulling forces, the exploration of protein conformational changes under compressive forces has been limited. In this study, we investigate the response of unliganded epidermal growth factor receptor (EGFR) monomers, liganded EGF-EGFR monomers, and dimers when exposed to external compressive forces using a home-modified AFM setup with an ultrasoft AFM tip. We observed that both ligand-bound and unbound EGFR proteins can undergo spontaneous tertiary structural rupture under piconewton-level compressive forces, a previously hidden protein behavior that may play a significant role in protein cell signaling. The magnitudes of the threshold compressive forces obtained in our study lie in the range of tens and hundreds of piconewtons (pN), which is accessible within a live biological system. Moreover, we developed a kinetic model to exhibit that only a fraction of the uniaxial compressive force exerted by the AFM tip affects the internal tension that causes a pseudopulling force within the protein before it undergoes the tertiary structural rupture. This calculated fraction ranged from 0.45 to 0.65, depending on the protein type and the approach velocity of the AFM tip. Additionally, we employed molecular dynamics (MD) simulations, particularly Steered MD (SMD) simulations along with Umbrella Sampling (US), to investigate the dynamics of unliganded and liganded EGFR in the presence of external compressive forces. These MD simulation results offer valuable insights into the flexibilities and unfolding behaviors of both liganded and unliganded EGFR proteins when subjected to external compressive forces.
Collapse
Affiliation(s)
- Dedunu S Senarathne
- Department of Chemistry, Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, United States
| | - Lalita Shahu
- Department of Chemistry, Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, United States
| | - H Peter Lu
- Department of Chemistry, Center for Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, United States
| |
Collapse
|
2
|
Ruan H, Lemke EA. Resolving Conformational Plasticity in Mammalian Cells with High-Resolution Fluorescence Tools. Annu Rev Phys Chem 2025; 76:103-128. [PMID: 40258244 DOI: 10.1146/annurev-physchem-082423-030632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Investigating protein dynamic structural changes is fundamental for understanding protein function, drug discovery, and disease mechanisms. Traditional studies of protein dynamics often rely on investigations of purified systems, which fail to capture the complexity of the cellular environment. The intracellular milieu imposes distinct physicochemical constraints that affect macromolecular interactions and dynamics in ways not easily replicated in isolated experimental setups. We discuss the use of fluorescence resonance energy transfer, fluorescence anisotropy, and minimal photon flux imaging technologies to address these challenges and directly investigate protein conformational dynamics in mammalian cells. Key findings from the application of these techniques demonstrate their potential to reveal intricate details of protein conformational plasticity. By overcoming the limitations of traditional in vitro methods, these approaches offer a more accurate and comprehensive understanding of protein function and behavior within the complex environment of mammalian cells.
Collapse
Affiliation(s)
- Hao Ruan
- BioCenter, Johannes Gutenberg University Mainz, Mainz, Germany;
- IMB Postdoc Program, Institute of Molecular Biology, Mainz, Germany
| | - Edward A Lemke
- BioCenter, Johannes Gutenberg University Mainz, Mainz, Germany;
- Institute of Molecular Biology, Mainz, Germany
| |
Collapse
|
3
|
Ling H, Zhang W, Zhang Y, Shen J, Liu Q. Lanthanide-Doped Upconversion Nanoparticles for Single-Particle Imaging. Chembiochem 2025:e2400942. [PMID: 40134352 DOI: 10.1002/cbic.202400942] [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: 11/19/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 03/27/2025]
Abstract
Lanthanide-doped upconversion nanoparticles (UCNPs) have recently demonstrated great promise in single-particle imaging (SPI) due to their exceptional photostability and minimal background fluorescence. However, their limited brightness has posed a significant barrier to wider adoption in SPI applications. This review highlights recent advances in applying UCNPs for SPI, focusing on strategies to enhance their brightness and reduce quenching effects in aqueous environments. Additionally, it summarizes the latest progress in using UCNPs for single-particle tracking and super-resolution imaging, underscoring their potential in biomedical research. Finally, the review outlines current challenges and future directions in this field.
Collapse
Affiliation(s)
- Huan Ling
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| | - Wenrui Zhang
- 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
| | - Jie Shen
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Qian Liu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai, 200438, China
| |
Collapse
|
4
|
Isogai T, Hirosawa KM, Suzuki KGN. Recent Advancements in Imaging Techniques for Individual Extracellular Vesicles. Molecules 2024; 29:5828. [PMID: 39769916 PMCID: PMC11728280 DOI: 10.3390/molecules29245828] [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: 11/14/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs), secreted from most cells, are small lipid membranes of vesicles of 30 to 1000 nm in diameter and contain nucleic acids, proteins, and intracellular organelles originating from donor cells. EVs play pivotal roles in intercellular communication, particularly in forming niches for cancer cell metastasis. However, EVs derived from donor cells exhibit significant heterogeneity, complicating the investigation of EV subtypes using ensemble averaging methods. In this context, we highlight recent studies that characterize individual EVs using advanced techniques, including single-fluorescent-particle tracking, single-metal-nanoparticle tracking, single-non-label-particle tracking, super-resolution microscopy, and atomic force microscopy. These techniques have facilitated high-throughput analyses of the properties of individual EV particles such as their sizes, compositions, and physical properties. Finally, we address the challenges that need to be resolved via single-particle (-molecule) imaging and super-resolution microscopy in future research.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan;
| | - Koichiro M. Hirosawa
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan;
| | - Kenichi G. N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu 501-1193, Japan;
- Institute for Glyco-Core Research (iGCORE), Gifu University, Gifu 501-1193, Japan;
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo 104-0045, Japan
| |
Collapse
|
5
|
Sannigrahi A, Ghosh S, Pradhan S, Jana P, Jawed JJ, Majumdar S, Roy S, Karmakar S, Mukherjee B, Chattopadhyay K. Leishmania protein KMP-11 modulates cholesterol transport and membrane fluidity to facilitate host cell invasion. EMBO Rep 2024; 25:5561-5598. [PMID: 39482488 PMCID: PMC11624268 DOI: 10.1038/s44319-024-00302-7] [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: 07/16/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 11/03/2024] Open
Abstract
The first step of successful infection by any intracellular pathogen relies on its ability to invade its host cell membrane. However, the detailed structural and molecular understanding underlying lipid membrane modification during pathogenic invasion remains unclear. In this study, we show that a specific Leishmania donovani (LD) protein, KMP-11, forms oligomers that bridge LD and host macrophage (MΦ) membranes. This KMP-11 induced interaction between LD and MΦ depends on the variations in cholesterol (CHOL) and ergosterol (ERG) contents in their respective membranes. These variations are crucial for the subsequent steps of invasion, including (a) the initial attachment, (b) CHOL transport from MΦ to LD, and (c) detachment of LD from the initial point of contact through a liquid ordered (Lo) to liquid disordered (Ld) membrane-phase transition. To validate the importance of KMP-11, we generate KMP-11 depleted LD, which failed to attach and invade host MΦ. Through tryptophan-scanning mutagenesis and synthesized peptides, we develop a generalized mathematical model, which demonstrates that the hydrophobic moment and the symmetry sequence code at the membrane interacting protein domain are key factors in facilitating the membrane phase transition and, consequently, the host cell infection process by Leishmania parasites.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Souradeepa Ghosh
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Supratim Pradhan
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Pulak Jana
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
| | - Junaid Jibran Jawed
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, 700156, India
| | - Subrata Majumdar
- Department of Molecular Medicine, Bose Institute, Kolkata, West Bengal, 700054, India
| | - Syamal Roy
- Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
- INSA Senior Scientist, Indian Association for the Cultivation of Science, Kolkata, West Bengal, 700032, India
| | - Sanat Karmakar
- Department of Physics, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, IIT-Kharagpur, Kharagpur, West Bengal, 721302, India.
| | - Krishnananda Chattopadhyay
- Structural Biology & Bio-Informatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mallick Road, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
6
|
Abe M, Yanagawa M, Hiroshima M, Kobayashi T, Sako Y. Bilateral regulation of EGFR activity and local PI(4,5)P 2 dynamics in mammalian cells observed with superresolution microscopy. eLife 2024; 13:e101652. [PMID: 39513999 PMCID: PMC11548882 DOI: 10.7554/elife.101652] [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: 07/24/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024] Open
Abstract
Anionic lipid molecules, including phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2), are implicated in the regulation of epidermal growth factor receptor (EGFR). However, the role of the spatiotemporal dynamics of PI(4,5)P2 in the regulation of EGFR activity in living cells is not fully understood, as it is difficult to visualize the local lipid domains around EGFR. Here, we visualized both EGFR and PI(4,5)P2 nanodomains in the plasma membrane of HeLa cells using super-resolution single-molecule microscopy. The EGFR and PI(4,5)P2 nanodomains aggregated before stimulation with epidermal growth factor (EGF) through transient visits of EGFR to the PI(4,5)P2 nanodomains. The degree of coaggregation decreased after EGF stimulation and depended on phospholipase Cγ, the EGFR effector hydrolyzing PI(4,5)P2. Artificial reduction in the PI(4,5)P2 content of the plasma membrane reduced both the dimerization and autophosphorylation of EGFR after stimulation with EGF. Inhibition of PI(4,5)P2 hydrolysis after EGF stimulation decreased phosphorylation of EGFR-Thr654. Thus, EGFR kinase activity and the density of PI(4,5)P2 around EGFR molecules were found to be mutually regulated.
Collapse
Affiliation(s)
- Mitsuhiro Abe
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering ResearchWakoJapan
| | - Masataka Yanagawa
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering ResearchWakoJapan
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku UniversitySendaiJapan
| | - Michio Hiroshima
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering ResearchWakoJapan
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka UniversityOsakaJapan
| | - Toshihide Kobayashi
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering ResearchWakoJapan
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, Faculté de PharmacieIllkirchFrance
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering ResearchWakoJapan
| |
Collapse
|
7
|
Watanabe D, Hiroshima M, Yasui M, Ueda M. Single molecule tracking based drug screening. Nat Commun 2024; 15:8975. [PMID: 39420015 PMCID: PMC11486946 DOI: 10.1038/s41467-024-53432-w] [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: 12/29/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
The single-molecule tracking of transmembrane receptors in living cells has provided significant insights into signaling mechanisms, such as mobility and clustering upon their activation/inactivation, making it a potential screening method for drug discovery. Here we show that single-molecule tracking-based screening can be used to explore compounds both detectable and undetectable by conventional methods for disease-related receptors. Using an automated system for a fast large-scale single-molecule analysis, we screen for epidermal growth factor receptor (EGFR) from 1134 of FDA approved drugs. The 18 hit compounds include all EGFR-targeted tyrosine kinase inhibitors (TKIs) in the library that suppress any phosphorylation-dependent mobility shift of EGFR, proving the concept of this approach. The remaining hit compounds are not reported as EGFR-targeted drugs and do not inhibit EGF-induced EGFR phosphorylation. These non-TKI compounds affect the mobility and/or clustering of EGFR without EGF and induce EGFR internalization, to impede EGFR-dependent cell growth. Thus, single-molecule tracking provides an alternative modality for discovering therapeutics on various receptor functions with previously untargeted mechanisms.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research, RIKEN, Suita, Osaka, Japan
| | - Michio Hiroshima
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan.
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research, RIKEN, Suita, Osaka, Japan.
| | | | - Masahiro Ueda
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan.
- Laboratory for Cell Signaling Dynamics, Center for Biosystems Dynamics Research, RIKEN, Suita, Osaka, Japan.
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Osaka, Japan.
| |
Collapse
|
8
|
Hockenberry MA, Daugird TA, Legant WR. Cell dynamics revealed by microscopy advances. Curr Opin Cell Biol 2024; 90:102418. [PMID: 39159598 PMCID: PMC11392612 DOI: 10.1016/j.ceb.2024.102418] [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: 05/01/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Cell biology emerges from spatiotemporally coordinated molecular processes. Recent advances in live-cell microscopy, fueled by a surge in optical, molecular, and computational technologies, have enabled dynamic observations from single molecules to whole organisms. Despite technological leaps, there is still an untapped opportunity to fully leverage their capabilities toward biological insight. We highlight how single-molecule imaging has transformed our understanding of biological processes, with a focus on chromatin organization and transcription in the nucleus. We describe how this was enabled by the close integration of new imaging techniques with analysis tools and discuss the challenges to make a comparable impact at larger scales from organelles to organisms. By highlighting recent successful examples, we describe an outlook of ever-increasing data and the need for seamless integration between dataset visualization and quantification to realize the full potential warranted by advances in new imaging technologies.
Collapse
Affiliation(s)
- Max A Hockenberry
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, USA
| | - Timothy A Daugird
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wesley R Legant
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
9
|
Yoda T, Sako Y, Inoue A, Yanagawa M. Four-color single-molecule imaging system for tracking GPCR dynamics with fluorescent HiBiT peptide. Biophys Physicobiol 2024; 21:e210020. [PMID: 39802745 PMCID: PMC11718171 DOI: 10.2142/biophysico.bppb-v21.0020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/18/2024] [Indexed: 01/16/2025] Open
Abstract
Single-molecule imaging provides information on diffusion dynamics, oligomerization, and protein-protein interactions in living cells. To simultaneously monitor different types of proteins at the single-molecule level, orthogonal fluorescent labeling methods with different photostable dyes are required. G-protein-coupled receptors (GPCRs), a major class of drug targets, are prototypical membrane receptors that have been studied using single-molecule imaging techniques. Here we developed a method for labeling cell-surface GPCRs inspired by the HiBiT system, which utilizes the high affinity complementation between LgBiT and HiBiT fragments of the NanoLuc luciferase. We synthesized four fluorescence-labeled HiBiT peptides (F-FiBiTs) with a different color dye (Setau-488, TMR, SaraFluor 650 and SaraFluor 720). We constructed a multicolor total internal reflection fluorescence microscopy system that allows us to track four color dyes simultaneously. As a proof-of-concept experiment, we labeled an N-terminally LgBiT-fused GPCR (Lg-GPCR) with a mixture of the four F-FiBiTs and successfully tracked each dye within a cell at the single-molecule level. The F-FiBiT-labeled Lg-GPCRs showed agonist-dependent changes in the diffusion dynamics and accumulation into the clathrin-coated pits as observed with a conventional method using a C-terminally HaloTag-fused GPCR. Taking advantage of luciferase complementation by the F-FiBiT and Lg-GPCRs, the F-FiBiT was also applicable to bioluminescence plate-reader-based assays. By combining existing labeling methods such as HaloTag, SNAP-tag, and fluorescent proteins, the F-FiBiT method will be useful for multicolor single-molecule imaging and will enhance our understanding of GPCR signaling at the single-molecule level.
Collapse
Affiliation(s)
- Toshiki Yoda
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
| | - Asuka Inoue
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Masataka Yanagawa
- Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
| |
Collapse
|
10
|
Mashanov GI, Molloy JE. Single molecule dynamics in a virtual cell combining a 3-dimensional matrix model with random walks. Sci Rep 2024; 14:20032. [PMID: 39198682 PMCID: PMC11358523 DOI: 10.1038/s41598-024-70925-2] [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: 04/17/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024] Open
Abstract
Recent advances in light microscopy have enabled single molecules to be imaged and tracked within living cells and this approach is impacting our understanding of cell biology. Computer modeling and simulation are important adjuncts to the experimental cycle since they aid interpretation of experimental results and help refine, test and generate hypotheses. Object-oriented computer modeling is particularly well-suited for simulating random, thermal, movements of individual molecules as they interact with other molecules and subcellular structures, but current models are often limited to idealized systems consisting of unit volumes or planar surfaces. Here, a simulation tool is described that combines a 3-dimensional, voxelated, representation of the cell consisting of subcellular structures (e.g. nucleus, endoplasmic reticulum, cytoskeleton, vesicles, and filopodia) combined with numerical floating-point precision simulation of thousands of individual molecules moving and interacting within the 3-dimensional space. Simulations produce realistic time-series video sequences comprising single fluorophore intensities and realistic background noise which can be directly compared to experimental fluorescence video microscopy data sets.
Collapse
Affiliation(s)
| | - Justin E Molloy
- The Francis Crick Institute, London, NW1 1AT, UK
- Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
11
|
Winkelmann H, Richter CP, Eising J, Piehler J, Kurre R. Correlative single-molecule and structured illumination microscopy of fast dynamics at the plasma membrane. Nat Commun 2024; 15:5813. [PMID: 38987559 PMCID: PMC11236984 DOI: 10.1038/s41467-024-49876-9] [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: 09/25/2023] [Accepted: 06/21/2024] [Indexed: 07/12/2024] Open
Abstract
Total internal reflection fluorescence (TIRF) microscopy offers powerful means to uncover the functional organization of proteins in the plasma membrane with very high spatial and temporal resolution. Traditional TIRF illumination, however, shows a Gaussian intensity profile, which is typically deteriorated by overlaying interference fringes hampering precise quantification of intensities-an important requisite for quantitative analyses in single-molecule localization microscopy (SMLM). Here, we combine flat-field illumination by using a standard πShaper with multi-angular TIR illumination by incorporating a spatial light modulator compatible with fast super-resolution structured illumination microscopy (SIM). This distinct combination enables quantitative multi-color SMLM with a highly homogenous illumination. By using a dual camera setup with optimized image splitting optics, we achieve a versatile combination of SMLM and SIM with up to three channels. We deploy this setup for establishing robust detection of receptor stoichiometries based on single-molecule intensity analysis and single-molecule Förster resonance energy transfer (smFRET). Homogeneous illumination furthermore enables long-term tracking and localization microscopy (TALM) of cell surface receptors identifying spatial heterogeneity of mobility and accessibility in the plasma membrane. By combination of TALM and SIM, spatially and molecularly heterogenous diffusion properties can be correlated with nanoscale cytoskeletal organization and dynamics.
Collapse
Affiliation(s)
- Hauke Winkelmann
- Division of Biophysics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany
| | - Christian P Richter
- Division of Biophysics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany
| | - Jasper Eising
- Division of Biophysics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany
| | - Jacob Piehler
- Division of Biophysics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany.
- Center for Cellular Nanoanalytics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany.
| | - Rainer Kurre
- Division of Biophysics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany.
- Center for Cellular Nanoanalytics, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany.
- Integrated Bioimaging Facility iBiOs, Department of Biology/Chemistry, Osnabrück University, Barbarastraße 11, D-49076, Osnabrück, Germany.
| |
Collapse
|
12
|
Cichos F, Xia T, Yang H, Zijlstra P. The ever-expanding optics of single-molecules and nanoparticles. J Chem Phys 2024; 161:010401. [PMID: 38949895 DOI: 10.1063/5.0221680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- F Cichos
- Peter Debye Institute for Soft Matter Physics, Leipzig University, Leipzig, Germany
| | - T Xia
- Institute for Immunology, School of Medicine, Tsinghua University, Beijing, China
| | - H Yang
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, USA
| | - P Zijlstra
- Department of Applied Physics and Science Education, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands
| |
Collapse
|
13
|
Modak A, Kilic Z, Chattrakun K, Terry DS, Kalathur RC, Blanchard SC. Single-Molecule Imaging of Integral Membrane Protein Dynamics and Function. Annu Rev Biophys 2024; 53:427-453. [PMID: 39013028 DOI: 10.1146/annurev-biophys-070323-024308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Integral membrane proteins (IMPs) play central roles in cellular physiology and represent the majority of known drug targets. Single-molecule fluorescence and fluorescence resonance energy transfer (FRET) methods have recently emerged as valuable tools for investigating structure-function relationships in IMPs. This review focuses on the practical foundations required for examining polytopic IMP function using single-molecule FRET (smFRET) and provides an overview of the technical and conceptual frameworks emerging from this area of investigation. In this context, we highlight the utility of smFRET methods to reveal transient conformational states critical to IMP function and the use of smFRET data to guide structural and drug mechanism-of-action investigations. We also identify frontiers where progress is likely to be paramount to advancing the field.
Collapse
Affiliation(s)
- Arnab Modak
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
| | - Zeliha Kilic
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
| | - Kanokporn Chattrakun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
| | - Daniel S Terry
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
| | - Ravi C Kalathur
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
| | - Scott C Blanchard
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA; , , , , ,
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
14
|
Mongiat M, Pascal G, Poletto E, Williams DM, Iozzo RV. Proteoglycans of basement membranes: Crucial controllers of angiogenesis, neurogenesis, and autophagy. PROTEOGLYCAN RESEARCH 2024; 2:e22. [PMID: 39184370 PMCID: PMC11340296 DOI: 10.1002/pgr2.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/02/2024] [Indexed: 08/27/2024]
Abstract
Anti-angiogenic therapy is an established method for the treatment of several cancers and vascular-related diseases. Most of the agents employed target the vascular endothelial growth factor A, the major cytokine stimulating angiogenesis. However, the efficacy of these treatments is limited by the onset of drug resistance. Therefore, it is of fundamental importance to better understand the mechanisms that regulate angiogenesis and the microenvironmental cues that play significant role and influence patient treatment and outcome. In this context, here we review the importance of the three basement membrane heparan sulfate proteoglycans (HSPGs), namely perlecan, agrin and collagen XVIII. These HSPGs are abundantly expressed in the vasculature and, due to their complex molecular architecture, they interact with multiple endothelial cell receptors, deeply affecting their function. Under normal conditions, these proteoglycans exert pro-angiogenic functions. However, in pathological conditions such as cancer and inflammation, extracellular matrix remodeling leads to the degradation of these large precursor molecules and the liberation of bioactive processed fragments displaying potent angiostatic activity. These unexpected functions have been demonstrated for the C-terminal fragments of perlecan and collagen XVIII, endorepellin and endostatin. These bioactive fragments can also induce autophagy in vascular endothelial cells which contributes to angiostasis. Overall, basement membrane proteoglycans deeply affect angiogenesis counterbalancing pro-angiogenic signals during tumor progression, and represent possible means to develop new prognostic biomarkers and novel therapeutic approaches for the treatment of solid tumors.
Collapse
Affiliation(s)
- Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Gabriel Pascal
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Davion M. Williams
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology and Genomic Medicine, and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
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: 9] [Impact Index Per Article: 9.0] [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
|
Hu J, Zhao F, Ling H, Zhang Y, Liu Q. Single-particle Förster resonance energy transfer from upconversion nanoparticles to organic dyes. NANOSCALE ADVANCES 2024; 6:2945-2953. [PMID: 38817426 PMCID: PMC11134271 DOI: 10.1039/d4na00198b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/11/2024] [Indexed: 06/01/2024]
Abstract
Single-particle detection and sensing, powered by Förster resonance energy transfer (FRET), offers precise monitoring of molecular interactions and environmental stimuli at a nanometric resolution. Despite its potential, the widespread use of FRET has been curtailed by the rapid photobleaching of traditional fluorophores. This study presents a robust single-particle FRET platform utilizing upconversion nanoparticles (UCNPs), which stand out for their remarkable photostability, making them superior to conventional organic donors for energy transfer-based assays. Our comprehensive research demonstrates the influence of UCNPs' size, architecture, and dye selection on the efficiency of FRET. We discovered that small particles (∼14 nm) with a Yb3+-enriched outermost shell exhibit a significant boost in FRET efficiency, a benefit not observed in larger particles (∼25 nm). 25 nm UCNPs with an inert NaLuF4 shell demonstrated a comparable level of emission enhancement via FRET as those with a Yb3+-enriched outermost shell. At the single-particle level, these FRET-enhanced UCNPs manifested an upconversion green emission intensity that was 8.3 times greater than that of their unmodified counterparts, while maintaining notable luminescence stability. Our upconversion FRET system opens up new possibilities for developing more effective high-brightness, high-sensitivity single-particle detection, and sensing modalities.
Collapse
Affiliation(s)
- Jialing Hu
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University Shanghai 200438 China
| | - Fei Zhao
- Department of Chemistry and Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University Shanghai 200438 China
| | - Huan Ling
- 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
|
17
|
Hiroshima M, Bannai H, Matsumoto G, Ueda M. Application of single-molecule analysis to singularity phenomenon of cells. Biophys Physicobiol 2024; 21:e211018. [PMID: 39175861 PMCID: PMC11338674 DOI: 10.2142/biophysico.bppb-v21.s018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/02/2024] [Indexed: 08/24/2024] Open
Abstract
Single-molecule imaging in living cells is an effective tool for elucidating the mechanisms of cellular phenomena at the molecular level. However, the analysis was not designed for throughput and requires high expertise, preventing it from reaching large scale, which is necessary when searching for rare cells that induce singularity phenomena. To overcome this limitation, we have automated the imaging procedures by combining our own focusing device, artificial intelligence, and robotics. The apparatus, called automated in-cell single-molecule imaging system (AiSIS), achieves a throughput that is a hundred-fold higher than conventional manual imaging operations, enabling the analysis of molecular events by individual cells across a large population. Here, using AiSIS, we demonstrate the single-molecule imaging of molecular behaviors and reactions related to tau protein aggregation, which is considered a singularity phenomenon in neurological disorders. Changes in the dynamics and kinetics of molecular events were observed inside and on the basal membrane of cells after the induction of aggregation. Additionally, to detect rare cells based on the molecular behavior, we developed a method to identify the state of individual cells defined by the quantitative distribution of molecular mobility and clustering. Using this method, cellular variations in receptor behavior were shown to decrease following ligand stimulation. This cell state analysis based on large-scale single-molecule imaging by AiSIS will advance the study of molecular mechanisms causing singularity phenomena.
Collapse
Affiliation(s)
- Michio Hiroshima
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
- Laboratory for Cell Signaling Dynamics, RIKEN BDR, Osaka 565-0874, Japan
| | - Hiroko Bannai
- School of Advanced Science and Engineering, Department of Electrical Engineering and Biosciences, Waseda University, Shinjuku-ku, Tokyo 162-0056, Japan
| | - Gen Matsumoto
- Department of Neurological Disease Control, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
- Department of Anatomy and Neurobiology, Nagasaki University School of Medicine, Nagasaki 852-8523, Japan
| | - Masahiro Ueda
- Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
- Laboratory for Cell Signaling Dynamics, RIKEN BDR, Osaka 565-0874, Japan
| |
Collapse
|
18
|
Takayama M, Maeda S, Watanabe D, Takebayashi K, Hiroshima M, Ueda M. Cholesterol suppresses spontaneous activation of EGFR-mediated signal transduction. Biochem Biophys Res Commun 2024; 704:149673. [PMID: 38401305 DOI: 10.1016/j.bbrc.2024.149673] [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: 01/27/2024] [Revised: 01/30/2024] [Accepted: 02/11/2024] [Indexed: 02/26/2024]
Abstract
Epidermal growth factor receptor (EGFR)-mediated signal transduction controls cell growth and proliferation. The signaling pathway is regulated so that it is activated only by external EGF stimuli, but the mechanisms that prevent EGF-independent spontaneous activation of EGFR-mediated signaling are unknown. Here we report cholesterol depletion activates EGFR-mediated signaling without EGF. We applied automated single-molecule imaging to EGFR and characterized the lateral diffusion and cluster formation on cholesterol-depleted and cholesterol-supplemented membranes. In cells in which cholesterol was depleted by methyl-β-cyclodextrin (MβCD) treatment, EGFR exhibited a reduction in lateral diffusion, an acceleration of cluster formation, and autophosphorylation without EGF. Concurrently, extracellular signal-regulated kinase (ERK), which is regulated by EGFR-mediated signaling, exhibited phosphorylation and nuclear translocation without EGF. These cholesterol depletion-induced changes were similar, albeit less efficient, to those that occurred with EGF stimulation in normal cells without MβCD, indicating the spontaneous activation of EGFR signaling. The exogenous supplementation of cholesterol suppressed the MβCD-induced spontaneous activation of EGFR and ERK nuclear translocation. Single-molecule imaging of EGFR in a large number of cells revealed cell-to-cell heterogeneity, with a sub-population showing a high ability for spontaneous activation. These results provide evidence that EGFR-mediated signaling is properly regulated by cholesterol metabolism to prevent uncontrolled spontaneous activation.
Collapse
Affiliation(s)
- Miri Takayama
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Sakura Maeda
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Daisuke Watanabe
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Kazutoshi Takebayashi
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan
| | - Michio Hiroshima
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan.
| | - Masahiro Ueda
- Laboratory of Single Molecular Biology, Graduate School of Science and Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, 565-0871, Japan; Laboratory for Cell Signaling Dynamics, BDR (Biosystems and Dynamics Research Center), RIKEN, Suita, Osaka, 565-0874, Japan.
| |
Collapse
|
19
|
Mudumbi KC, Burns EA, Schodt DJ, Petrova ZO, Kiyatkin A, Kim LW, Mangiacapre EM, Ortiz-Caraveo I, Rivera Ortiz H, Hu C, Ashtekar KD, Lidke KA, Lidke DS, Lemmon MA. Distinct interactions stabilize EGFR dimers and higher-order oligomers in cell membranes. Cell Rep 2024; 43:113603. [PMID: 38117650 PMCID: PMC10835193 DOI: 10.1016/j.celrep.2023.113603] [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: 06/23/2023] [Revised: 09/23/2023] [Accepted: 12/05/2023] [Indexed: 12/22/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with important roles in many cellular processes as well as in cancer and other diseases. EGF binding promotes EGFR dimerization and autophosphorylation through interactions that are well understood structurally. How these dimers relate to higher-order EGFR oligomers seen in cell membranes, however, remains unclear. Here, we used single-particle tracking (SPT) and Förster resonance energy transfer imaging to examine how each domain of EGFR contributes to receptor oligomerization and the rate of receptor diffusion in the cell membrane. Although the extracellular region of EGFR is sufficient to drive receptor dimerization, we find that the EGF-induced EGFR slowdown seen by SPT requires higher-order oligomerization-mediated in part by the intracellular tyrosine kinase domain when it adopts an active conformation. Our data thus provide important insight into the interactions required for higher-order EGFR assemblies involved in EGF signaling.
Collapse
Affiliation(s)
- Krishna C Mudumbi
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA.
| | - Eric A Burns
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - David J Schodt
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87106, USA
| | - Zaritza O Petrova
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Anatoly Kiyatkin
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Lucy W Kim
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Emma M Mangiacapre
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Irais Ortiz-Caraveo
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Hector Rivera Ortiz
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Chun Hu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Kumar D Ashtekar
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA
| | - Keith A Lidke
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87106, USA
| | - Diane S Lidke
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Mark A Lemmon
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cancer Biology Institute, Yale University West Campus, West Haven, CT 06516, USA.
| |
Collapse
|
20
|
Zhao X, Na N, Ouyang J. Functionalized DNA nanoplatform for multi-target simultaneous imaging: Establish the atlas of cancer cell species. Talanta 2024; 267:125222. [PMID: 37778181 DOI: 10.1016/j.talanta.2023.125222] [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: 06/09/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Detection and imaging of cell membrane receptor proteins have gained widespread interest in recent years. However, recognition based on a single biomarker can induce false positive feedback, including off-target phenomenon caused by the absence of tumor-specific antigens. In addition, nucleic acid probes often cause nonspecific and undesired cell internalization during cell imaging. In this work, we constructed a logic gate DNA nano-platform (LGDP) for single-molecule imaging of cell membrane proteins to synergistically diagnose cancer cells. The traffic light-like color response of LGDP facilitates the precise discrimination among different cell lines. Combined with single molecule technology, the target proteins were qualitatively and quantitatively analyzed synergistically. Logic-gated recognition integrated in aptamer-functionalized molecular machines will prompt fast cells analysis, laying the foundation of cancer early diagnosis and treatment.
Collapse
Affiliation(s)
- Xuan Zhao
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Na Na
- College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Jin Ouyang
- College of Chemistry, Beijing Normal University, Beijing, 100875, China; Department of Chemistry, College of Arts and Sciences, Beijing Normal University at Zhuhai, Zhuhai City, 519087, Guangdong Province, China.
| |
Collapse
|
21
|
Nagl M, Mönnich D, Rosier N, Schihada H, Sirbu A, Konar N, Reyes-Resina I, Navarro G, Franco R, Kolb P, Annibale P, Pockes S. Fluorescent Tools for the Imaging of Dopamine D 2 -Like Receptors. Chembiochem 2024; 25:e202300659. [PMID: 37942961 DOI: 10.1002/cbic.202300659] [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: 09/26/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/10/2023]
Abstract
The family of dopamine D2 -like receptors represents an interesting target for a variety of neurological diseases, e. g. Parkinson's disease (PD), addiction, or schizophrenia. In this study we describe the synthesis of a new set of fluorescent ligands as tools for visualization of dopamine D2 -like receptors. Pharmacological characterization in radioligand binding studies identified UR-MN212 (20) as a high-affinity ligand for D2 -like receptors (pKi (D2long R)=8.24, pKi (D3 R)=8.58, pKi (D4 R)=7.78) with decent selectivity towards D1 -like receptors. Compound 20 is a neutral antagonist in a Go1 activation assay at the D2long R, D3 R, and D4 R, which is an important feature for studies using whole cells. The neutral antagonist 20, equipped with a 5-TAMRA dye, displayed rapid association to the D2long R in binding studies using confocal microscopy demonstrating its suitability for fluorescence microscopy. Furthermore, in molecular brightness studies, the ligand's binding affinity could be determined in a single-digit nanomolar range that was in good agreement with radioligand binding data. Therefore, the fluorescent compound can be used for quantitative characterization of native D2 -like receptors in a broad variety of experimental setups.
Collapse
Affiliation(s)
- Martin Nagl
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Denise Mönnich
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Niklas Rosier
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Alexei Sirbu
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Nergis Konar
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marbacher Weg 6, 35037, Marburg, Germany
| | - Paolo Annibale
- Max Delbrück Center for Molecular Medicine, Berlin, 13125, Germany
- School of Physics and Astronomy, University of St Andrews, North Haugh, St Andrews, Scotland
| | - Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, D-93053, Regensburg, Germany
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, 55414, USA
| |
Collapse
|
22
|
Yang M, Batey JE, Dong B. Automated Five-Dimensional Single Particle Tracking by Bifocal Parallax Dark-Field Microscopy with Electronic Tunable Lens. Anal Chem 2024; 96:1-5. [PMID: 38153091 DOI: 10.1021/acs.analchem.3c04543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
We present a novel method for the precise tracking of plasmonic gold nanorods (AuNRs) in live cells, enabling a comprehensive understanding of the nanocargo's cellular dynamics. Traditional single particle tracking (SPT) struggles with accurately determining all five spatial parameters (x, y, z, ϕ, and θ) in live cells due to various challenges. Our innovation combines electronic tunable lens (ETL) technology with bifocal parallax dark-field (DF) microscopy, allowing continuous adjustment of the imaging focal plane for automatic tracking of both translational and rotational movements of AuNRs. This 5D single-particle orientation and rotational tracking (5D SPORT) method achieves remarkable precision, with 3D localization precisions of 9 (x), 10 (y), and 15 nm (z) and angular resolutions below 2°. To showcase its applicability, we investigated intracellular transport of nanocargos using transferrin-modified AuNRs as the imaging probe. Differentiated transport stages, such as active transport and pause period, were clearly unveiled from the observed dynamics in 5D. This advancement in single particle tracking holds promise for a wide range of applications in biomedical research, particularly when combined with other imaging modalities, such as light sheet fluorescence microscopy.
Collapse
Affiliation(s)
- Meek Yang
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - James Ethan Batey
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Bin Dong
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
23
|
Yang J, Ran K, Ma W, Chen Y, Chen Y, Zhang C, Ye H, Lu Y, Ran C. Degradation of Amyloid-β Species by Multi-Copper Oxidases. J Alzheimers Dis 2024; 101:525-539. [PMID: 39213075 DOI: 10.3233/jad-240625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Reduction of the production of amyloid-β (Aβ) species has been intensively investigated as potential therapeutic approaches for Alzheimer's disease (AD). However, the degradation of Aβ species, another potential beneficial approach, has been far less explored. Objective To investigate the potential of multi-copper oxidases (MCOs) in degrading Aβ peptides and their potential benefits for AD treatment. Methods We investigated the degradation efficiency of MCOs by using electrophoresis and validated the ceruloplasmin (CP)-Aβ interaction using total internal reflection fluorescence microscopy, fluorescence photometer, and fluorescence polarization measurement. We also investigated the therapeutic effect of ascorbate oxidase (AO) by using induced pluripotent stem (iPS) neuron cells and electrophysiological analysis with brain slices. Results We discovered that CP, an important MCO in human blood, could degrade Aβ peptides. We also found that other MCOs could induce Aβ degradation as well. Remarkably, we revealed that AO had the strongest degrading effect among the tested MCOs. Using iPS neuron cells, we observed that AO could rescue neuron toxicity which induced by Aβ oligomers. In addition, our electrophysiological analysis with brain slices suggested that AO could prevent an Aβ-induced deficit in synaptic transmission in the hippocampus. Conclusions To the best of our knowledge, our report is the first to demonstrate that MCOs have a degrading function for peptides/proteins. Further investigations are warranted to explore the possible benefits of MCOs for future AD treatment.
Collapse
Affiliation(s)
- Jing Yang
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Charlestown, Boston, MA, USA
- School of Engineering, China Pharmaceutical University, Nanjing, China
| | - Kathleen Ran
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Charlestown, Boston, MA, USA
| | - Wenzhe Ma
- Department of System Biology, Harvard Medical School, Boston, MA, USA
| | - Yanshi Chen
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Charlestown, Boston, MA, USA
| | - Yanxin Chen
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Charlestown, Boston, MA, USA
| | - Can Zhang
- Department of Neurology, Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hui Ye
- Department of Biology, Loyola University Chicago, IL, USA
| | - Ying Lu
- Department of System Biology, Harvard Medical School, Boston, MA, USA
| | - Chongzhao Ran
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Charlestown, Boston, MA, USA
| |
Collapse
|
24
|
Walker G, Brown C, Ge X, Kumar S, Muzumdar MD, Gupta K, Bhattacharyya M. Oligomeric organization of membrane proteins from native membranes at nanoscale spatial and single-molecule resolution. NATURE NANOTECHNOLOGY 2024; 19:85-94. [PMID: 38012273 PMCID: PMC10981947 DOI: 10.1038/s41565-023-01547-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
The oligomeric organization of membrane proteins in native cell membranes is a critical regulator of their function. High-resolution quantitative measurements of oligomeric assemblies and how they change under different conditions are indispensable to understanding membrane protein biology. We report Native-nanoBleach, a total internal reflection fluorescence microscopy-based single-molecule photobleaching step analysis technique to determine the oligomeric distribution of membrane proteins directly from native membranes at an effective spatial resolution of ~10 nm. We achieved this by capturing target membrane proteins in native nanodiscs with their proximal native membrane environment using amphipathic copolymers. We applied Native-nanoBleach to quantify the oligomerization status of structurally and functionally diverse membrane proteins, including a receptor tyrosine kinase (TrkA) and a small GTPase (KRas) under growth-factor binding and oncogenic mutations, respectively. Our data suggest that Native-nanoBleach provides a sensitive, single-molecule platform to quantify membrane protein oligomeric distributions in native membranes under physiologically and clinically relevant conditions.
Collapse
Affiliation(s)
- Gerard Walker
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Department of Cell Biology, Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University, West Haven, CT, USA
| | - Caroline Brown
- Department of Cell Biology, Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University, West Haven, CT, USA
| | - Xiangyu Ge
- Department of Pathology, Yale University, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Shailesh Kumar
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Mandar D Muzumdar
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
- Department of Genetics, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Yale Cancer Center, New Haven, USA
| | - Kallol Gupta
- Department of Cell Biology, Yale University, New Haven, CT, USA
- Nanobiology Institute, Yale University, West Haven, CT, USA
| | | |
Collapse
|
25
|
Kaizu K, Takahashi K. Technologies for whole-cell modeling: Genome-wide reconstruction of a cell in silico. Dev Growth Differ 2023; 65:554-564. [PMID: 37856476 PMCID: PMC11520977 DOI: 10.1111/dgd.12897] [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: 11/20/2022] [Revised: 09/06/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023]
Abstract
With advances in high-throughput, large-scale in vivo measurement and genome modification techniques at the single-nucleotide level, there is an increasing demand for the development of new technologies for the flexible design and control of cellular systems. Computer-aided design is a powerful tool to design new cells. Whole-cell modeling aims to integrate various cellular subsystems, determine their interactions and cooperative mechanisms, and predict comprehensive cellular behaviors by computational simulations on a genome-wide scale. It has been applied to prokaryotes, yeasts, and higher eukaryotic cells, and utilized in a wide range of applications, including production of valuable substances, drug discovery, and controlled differentiation. Whole-cell modeling, consisting of several thousand elements with diverse scales and properties, requires innovative model construction, simulation, and analysis techniques. Furthermore, whole-cell modeling has been extended to multiple scales, including high-resolution modeling at the single-nucleotide and single-amino acid levels and multicellular modeling of tissues and organs. This review presents an overview of the current state of whole-cell modeling, discusses the novel computational and experimental technologies driving it, and introduces further developments toward multihierarchical modeling on a whole-genome scale.
Collapse
|
26
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
27
|
Zalejski J, Sun J, Sharma A. Unravelling the Mystery inside Cells by Using Single-Molecule Fluorescence Imaging. J Imaging 2023; 9:192. [PMID: 37754956 PMCID: PMC10532472 DOI: 10.3390/jimaging9090192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Live-cell imaging is a powerful technique to study the dynamics and mechanics of various biological molecules like proteins, organelles, DNA, and RNA. With the rapid evolution of optical microscopy, our understanding of how these molecules are implicated in the cells' most critical physiological roles deepens. In this review, we focus on how spatiotemporal nanoscale live-cell imaging at the single molecule level allows for profound contributions towards new discoveries in life science. This review will start by summarizing how single-molecule tracking has been used to analyze membrane dynamics, receptor-ligand interactions, protein-protein interactions, inner- and extra-cellular transport, gene expression/transcription, and whole organelle tracking. We then move on to how current authors are trying to improve single-molecule tracking and overcome current limitations by offering new ways of labeling proteins of interest, multi-channel/color detection, improvements in time-lapse imaging, and new methods and programs to analyze the colocalization and movement of targets. We later discuss how single-molecule tracking can be a beneficial tool used for medical diagnosis. Finally, we wrap up with the limitations and future perspectives of single-molecule tracking and total internal reflection microscopy.
Collapse
Affiliation(s)
| | | | - Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL 60607, USA; (J.Z.); (J.S.)
| |
Collapse
|
28
|
Yang J, Ran K, Ma W, Chen L, Chen C, Zhang C, Ye H, Lu Y, Ran C. Degradation of amyloid beta species by multi-copper oxidases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.02.547398. [PMID: 37461701 PMCID: PMC10350030 DOI: 10.1101/2023.07.02.547398] [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: 07/24/2023]
Abstract
Reduction of the production of amyloid beta (Aβ) species has been intensively investigated as potential therapeutic approaches for Alzheimer's disease (AD). However, the degradation of Aβ species, another potential beneficial approach, has been far less explored. In this study, we discovered that ceruloplasmin (CP), an important multi-copper oxidase (MCO) in human blood, could degrade Aβ peptides. We also found that the presence of Vitamin C could enhance the degrading effect in a concentration-dependent manner. We then validated the CP-Aβ interaction using total internal reflection fluorescence (TIRF) microscopy, fluorescence photometer, and fluorescence polarization measurement. Based on the above discovery, we hypothesized that other MCOs had similar Aβ-degrading functions. Indeed, we found that other MCOs could induce Aβ degradation as well. Remarkably, we revealed that ascorbate oxidase (AO) had the strongest degrading effect among the tested MCOs. Using induced pluripotent stem (iPS) neuron cells, we observed that AO could rescue neuron toxicity which induced by Aβ oligomers. In addition, our electrophysiological analysis with brain slices suggested that AO could prevent an Ab-induced deficit in synaptic transmission in the hippocampus. To the best of our knowledge, our report is the first to demonstrate that MCOs have a degrading function for peptides/proteins. Further investigations are warranted to explore the possible benefits of MCOs for future AD treatment.
Collapse
Affiliation(s)
- Jing Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Kathleen Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129
| | - Wenzhe Ma
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Lucy Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129
| | - Cindy Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hui Ye
- Department of Biology, Loyola University Chicago, Chicago, IL, 60660
| | - Ying Lu
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Room 2301, Building 149, Charlestown, Boston, Massachusetts 02129
| |
Collapse
|
29
|
Martin‐Fernandez ML. A perspective of fluorescence microscopy for cellular structural biology with EGFR as witness. J Microsc 2023; 291:73-91. [PMID: 36282005 PMCID: PMC10952613 DOI: 10.1111/jmi.13151] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 09/20/2022] [Accepted: 10/11/2022] [Indexed: 10/31/2022]
Abstract
The epidermal growth factor receptor (EGFR) is a poster child for the understanding of receptor behaviour, and of paramount importance to cell function and human health. Cloned almost forty years ago, the interest in EGFR's structure/function relationships remains unabated, not least because changes in oncogenic EGFR mutants are key drivers of the formation of lung and brain tumours. The structure of the assemblies formed by EGFR have been comprehensibly investigated by techniques such as high-resolution X-ray crystallography, NMR and all-atom molecular dynamics (MD) simulations. However, the complexity embedded in the portfolio of EGFR states that are only possible in the physiological environment of cells has often proved refractory to cell-free structural methods. Conversely, some key inroads made by quantitative fluorescence microscopy and super-resolution have depended on exploiting the wealth of structures available. Here, a brief personal perspective is provided on how quantitative fluorescence microscopy and super-resolution methods have cross-fertilised with cell-free-derived EGFR structural information. I primarily discuss areas in which my research group has made a contribution to fill gaps in EGFR's cellular structural biology and towards developing new tools to investigate macromolecular assemblies in cells.
Collapse
Affiliation(s)
- M. L. Martin‐Fernandez
- Central Laser FacilityScience and Technology Facilities Council, Rutherford Appleton LaboratoryDidcotUK
| |
Collapse
|
30
|
Ruan Q, Zhao C. A method for parallel microscale protein labeling and precise control over the average degree of labeling (aDoL). Sci Rep 2023; 13:8961. [PMID: 37268718 DOI: 10.1038/s41598-023-36163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
A widely used approach for protein conjugation is through the lysine residues reacting with NHS- or other active esters. However, it is a challenge to precisely control the degree of labeling (DoL) due to the instability of active ester and variability of reaction efficiencies. Here, we provide a protocol for better control of aDoL using existing Copper-free Click Chemistry reagents. It is a two-step reaction with one purification in between. Briefly, proteins of interest were first activated with azide-NHS. After removing unreacted azide-NHS, the protein-N3 is then reacted with a limited amount of complementary click tag. Our studies have shown the click tag will fully react with the protein-N3 after 24 h' incubation, and therefore does not require additional purification steps. As such, the aDoL is equal to the input molar ratio of the click tag and the protein. Furthermore, this approach offers a much simpler and more economical way to perform parallel microscale labeling. Once a protein is pre-activated with N3-NHS, any fluorophore or molecule with the complementary click tag can be attached to the protein by mixing the two ingredients. Quantities of the protein used in the click reaction can be at any desired amount. In one example, we labeled an antibody in parallel with 9 different fluorophores using a total of 0.5 mg of antibody. In another example, we labeled Ab with targeted aDoL value from 2 to 8. In a stability comparison study, we have found the conjugated fluorophore using the suggested click protocol stayed attached to the protein longer than with standard NHS-fluorophore labeling.
Collapse
Affiliation(s)
- Qiaoqiao Ruan
- Applied Research and Technology, Abbott Diagnostics Division, AP-20, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL, 60064-6016, USA.
| | - Cheng Zhao
- Applied Research and Technology, Abbott Diagnostics Division, AP-20, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, IL, 60064-6016, USA
| |
Collapse
|
31
|
Sugiyama MG, Brown AI, Vega-Lugo J, Borges JP, Scott AM, Jaqaman K, Fairn GD, Antonescu CN. Confinement of unliganded EGFR by tetraspanin nanodomains gates EGFR ligand binding and signaling. Nat Commun 2023; 14:2681. [PMID: 37160944 PMCID: PMC10170156 DOI: 10.1038/s41467-023-38390-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/28/2023] [Indexed: 05/11/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is a central regulator of cell physiology. EGFR is activated by ligand binding, triggering receptor dimerization, activation of kinase activity, and intracellular signaling. EGFR is transiently confined within various plasma membrane nanodomains, yet how this may contribute to regulation of EGFR ligand binding is poorly understood. To resolve how EGFR nanoscale compartmentalization gates ligand binding, we developed single-particle tracking methods to track the mobility of ligand-bound and total EGFR, in combination with modeling of EGFR ligand binding. In comparison to unliganded EGFR, ligand-bound EGFR is more confined and distinctly regulated by clathrin and tetraspanin nanodomains. Ligand binding to unliganded EGFR occurs preferentially in tetraspanin nanodomains, and disruption of tetraspanin nanodomains impairs EGFR ligand binding and alters the conformation of the receptor's ectodomain. We thus reveal a mechanism by which EGFR confinement within tetraspanin nanodomains regulates receptor signaling at the level of ligand binding.
Collapse
Affiliation(s)
- Michael G Sugiyama
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Aidan I Brown
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Jesus Vega-Lugo
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jazlyn P Borges
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, La Trobe University, Melbourne, VIC, Australia
| | - Khuloud Jaqaman
- Department of Biophysics, UT Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Gregory D Fairn
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Costin N Antonescu
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, ON, Canada.
| |
Collapse
|
32
|
Dahal L, Walther N, Tjian R, Darzacq X, Graham TG. Single-molecule tracking (SMT): a window into live-cell transcription biochemistry. Biochem Soc Trans 2023; 51:557-569. [PMID: 36876879 PMCID: PMC10212543 DOI: 10.1042/bst20221242] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/07/2023]
Abstract
How molecules interact governs how they move. Single-molecule tracking (SMT) thus provides a unique window into the dynamic interactions of biomolecules within live cells. Using transcription regulation as a case study, we describe how SMT works, what it can tell us about molecular biology, and how it has changed our perspective on the inner workings of the nucleus. We also describe what SMT cannot yet tell us and how new technical advances seek to overcome its limitations. This ongoing progress will be imperative to address outstanding questions about how dynamic molecular machines function in live cells.
Collapse
Affiliation(s)
- Liza Dahal
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, U.S.A
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, U.S.A
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, Berkeley, U.S.A
| | - Nike Walther
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, U.S.A
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, Berkeley, U.S.A
| | - Robert Tjian
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, U.S.A
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, U.S.A
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, Berkeley, U.S.A
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, U.S.A
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, Berkeley, U.S.A
| | - Thomas G.W. Graham
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, U.S.A
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, U.S.A
- Li Ka Shing Center for Biomedical & Health Sciences, University of California, Berkeley, Berkeley, U.S.A
| |
Collapse
|
33
|
Mudumbi KC, Burns EA, Schodt DJ, Petrova ZO, Kiyatkin A, Kim LW, Mangiacapre EM, Ortiz-Caraveo I, Ortiz HR, Hu C, Ashtekar KD, Lidke KA, Lidke DS, Lemmon MA. Distinct interactions stabilize EGFR dimers and higher-order oligomers in cell membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536273. [PMID: 37090557 PMCID: PMC10120646 DOI: 10.1101/2023.04.10.536273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase (RTK) with important roles in many cellular processes as well as cancer and other diseases. EGF binding promotes EGFR dimerization and autophosphorylation through interactions that are well understood structurally. However, it is not clear how these dimers relate to higher-order EGFR oligomers detected at the cell surface. We used single-particle tracking (SPT) and Förster resonance energy transfer (FRET) imaging to examine how each domain within EGFR contributes to receptor dimerization and the rate of its diffusion in the cell membrane. We show that the EGFR extracellular region is sufficient to drive receptor dimerization, but that the EGF-induced EGFR slow-down seen by SPT requires formation of higher order oligomers, mediated in part by the intracellular tyrosine kinase domain - but only when in its active conformation. Our data thus provide important insight into higher-order EGFR interactions required for EGF signaling.
Collapse
|
34
|
Sobhanan J, Anas A, Biju V. Nanomaterials for Fluorescence and Multimodal Bioimaging. CHEM REC 2023; 23:e202200253. [PMID: 36789795 DOI: 10.1002/tcr.202200253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/27/2023] [Indexed: 02/16/2023]
Abstract
Bioconjugated nanomaterials replace molecular probes in bioanalysis and bioimaging in vitro and in vivo. Nanoparticles of silica, metals, semiconductors, polymers, and supramolecular systems, conjugated with contrast agents and drugs for image-guided (MRI, fluorescence, PET, Raman, SPECT, photodynamic, photothermal, and photoacoustic) therapy infiltrate into preclinical and clinical settings. Small bioactive molecules like peptides, proteins, or DNA conjugated to the surfaces of drugs or probes help us to interface them with cells and tissues. Nevertheless, the toxicity and pharmacokinetics of nanodrugs, nanoprobes, and their components become the clinical barriers, underscoring the significance of developing biocompatible next-generation drugs and contrast agents. This account provides state-of-the-art advancements in the preparation and biological applications of bioconjugated nanomaterials and their molecular, cell, and in vivo applications. It focuses on the preparation, bioimaging, and bioanalytical applications of monomodal and multimodal nanoprobes composed of quantum dots, quantum clusters, iron oxide nanoparticles, and a few rare earth metal ion complexes.
Collapse
Affiliation(s)
- Jeladhara Sobhanan
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Department of Chemistry, Rice University, Houston, Texas 77005, United States
| | - Abdulaziz Anas
- CSIR-National Institute of Oceanography, Regional Centre Kochi, Kerala, 682 018, India
| | - Vasudevanpillai Biju
- Graduate School of Environmental Science, Hokkaido University, N10 W5, Sapporo, Hokkaido, 060-0810, Japan.,Research Institute for Electronic Science, Hokkaido University, Sapporo, 001-0020, Japan
| |
Collapse
|
35
|
Walker G, Brown C, Ge X, Kumar S, Muzumdar MD, Gupta K, Bhattacharyya M. Determination of oligomeric organization of membrane proteins from native membranes at nanoscale-spatial and single-molecule resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.19.529138. [PMID: 36865290 PMCID: PMC9980011 DOI: 10.1101/2023.02.19.529138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
The oligomeric organization of membrane proteins in native cell membranes is a critical regulator of their function. High-resolution quantitative measurements of oligomeric assemblies and how they change under different conditions are indispensable to the understanding of membrane protein biology. We report a single-molecule imaging technique (Native-nanoBleach) to determine the oligomeric distribution of membrane proteins directly from native membranes at an effective spatial resolution of ∼10 nm. We achieved this by capturing target membrane proteins in "native nanodiscs" with their proximal native membrane environment using amphipathic copolymers. We established this method using structurally and functionally diverse membrane proteins with well-established stoichiometries. We then applied Native-nanoBleach to quantify the oligomerization status of a receptor tyrosine kinase (TrkA) and a small GTPase (KRas) under conditions of growth-factor binding or oncogenic mutations, respectively. Native-nanoBleach provides a sensitive, single-molecule platform to quantify membrane protein oligomeric distributions in native membranes at an unprecedented spatial resolution.
Collapse
|
36
|
Refinement of Singer-Nicolson fluid-mosaic model by microscopy imaging: Lipid rafts and actin-induced membrane compartmentalization. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184093. [PMID: 36423676 DOI: 10.1016/j.bbamem.2022.184093] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022]
Abstract
This year celebrates the 50th anniversary of the Singer-Nicolson fluid mosaic model for biological membranes. The next level of sophistication we have achieved for understanding plasma membrane (PM) structures, dynamics, and functions during these 50 years includes the PM interactions with cortical actin filaments and the partial demixing of membrane constituent molecules in the PM, particularly raft domains. Here, first, we summarize our current knowledge of these two structures and emphasize that they are interrelated. Second, we review the structure, molecular dynamics, and function of raft domains, with main focuses on raftophilic glycosylphosphatidylinositol-anchored proteins (GPI-APs) and their signal transduction mechanisms. We pay special attention to the results obtained by single-molecule imaging techniques and other advanced microscopy methods. We also clarify the limitations of present optical microscopy methods for visualizing raft domains, but emphasize that single-molecule imaging techniques can "detect" raft domains associated with molecules of interest in the PM.
Collapse
|
37
|
Imaging strategies for receptor tyrosine kinase dimers in living cells. Anal Bioanal Chem 2023; 415:67-82. [PMID: 36190534 DOI: 10.1007/s00216-022-04334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 01/10/2023]
Abstract
Receptor tyrosine kinases (RTKs) are the essential regulators of cell signal transduction pathways and play important roles in biological processes. RTK dimerization is generally considered the first step in receptor activation and cell communication. And the abnormal expression of RTK dimers is closely related to the occurrence and development of many diseases. Therefore, the visualization of RTK dimerization is of great significance for monitoring physiological processes. The genetic and nongenetic imaging strategies have attracted widespread attention due to their high efficiency and high sensitivity. In this review, the RTKs and their dimers as well as the advances in strategies for imaging RTK dimers are introduced. Furthermore, we analyze the limitations of existing imaging strategies and put forward suggestions for the future development of imaging probes. We expect that this review will inspire more in-depth investigation of RTK dimers, which will also broaden the application of strategies of RTK dimers in biomedical areas.
Collapse
|
38
|
Zhang L, Chu M, Ji C, Wei J, Yang Y, Huang Z, Tan W, Tan J, Yuan Q. In Situ Visualization of Epidermal Growth Factor Receptor Nuclear Translocation with Circular Bivalent Aptamer. Anal Chem 2022; 94:17413-17421. [PMID: 36469021 DOI: 10.1021/acs.analchem.2c02762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epidermal growth factor receptor (EGFR) nuclear translocation correlates with the abnormal proliferation, migration, and anti-apoptosis of tumor cells. Monitoring EGFR nuclear translocation provides insights into the molecular mechanisms underlying cancers. EGFR nuclear translocation includes two processes, EGFR phosphorylation and phosphorylated EGFR translocation to the nucleus. With the help of aptamers, probes that can achieve the first step of anchoring phosphorylated EGFR have been developed. However, the EGFR nuclear translocation can last for hours, posing a challenge to monitor the entire nuclear translocation in living cells. Herein, we designed a circular bivalent aptamer-functionalized optical probe with greatly enhanced stability for long-term visualization of EGFR nuclear translocation in situ. The results of cell experiments show that the probe could monitor the entire nuclear translocation of EGFR. The findings of tissue and in vivo experiments demonstrate that the probe can evaluate the development and progression of tumors by imaging EGFR nuclear translocation in situ. The proposed approach allows us to monitor EGFR nuclear translocation in the long term, indicating its great potential in investigating the mechanisms of cancers and guiding for tumor treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Mengge Chu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Cailing Ji
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Junyuan Wei
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yanbing Yang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Zhongnan Huang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.,Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jie Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Quan Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China.,College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
39
|
Li R, Li T, Lu G, Cao Z, Chen B, Wang Y, Du J, Li P. Programming cell-surface signaling by phase-separation-controlled compartmentalization. Nat Chem Biol 2022; 18:1351-1360. [DOI: 10.1038/s41589-022-01192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 09/30/2022] [Indexed: 11/18/2022]
|
40
|
Xie KX, Li Z, Fang JH, Cao SH, Li YQ. Au-Ag Alloy Nanoshuttle Mediated Surface Plasmon Coupling for Enhanced Fluorescence Imaging. BIOSENSORS 2022; 12:bios12111014. [PMID: 36421131 PMCID: PMC9688416 DOI: 10.3390/bios12111014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 05/27/2023]
Abstract
Surface plasmon-coupled emission (SPCE), a novel signal enhancement technology generated by the interactions between surface plasmons and excited fluorophores in close vicinity to metallic film, has shown excellent performance in bioimaging. Variable-angle nanoplasmonic fluorescence microscopy (VANFM), based on an SPCE imaging system, can selectively modulate the imaging depth by controlling the excitation angles. In order to further improve the imaging performance, Au-Ag alloy nanoshuttles were introduced into an Au substrate to mediate the plasmonic properties. Benefiting from the strong localized plasmon effect of the modified SPCE chip, better imaging brightness, signal-to-background ratio and axial resolution for imaging of the cell membrane region were obtained, which fully displays the imaging advantages of SPCE system. Meanwhile, the imaging signal obtained from the critical angle excitation mode was also amplified, which helps to acquire a more visible image of the cell both from near- and far-field in order to comprehensively investigate the cellular interactions.
Collapse
Affiliation(s)
- Kai-Xin Xie
- College of Chemistry and Materials, Taiyuan Normal University, Jinzhong 030619, China
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Zhao Li
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jia-Hua Fang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuo-Hui Cao
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Department of Electronic Science, Xiamen University, Xiamen 361005, China
| | - Yao-Qun Li
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
41
|
Graham TGW, Ferrie JJ, Dailey GM, Tjian R, Darzacq X. Detecting molecular interactions in live-cell single-molecule imaging with proximity-assisted photoactivation (PAPA). eLife 2022; 11:e76870. [PMID: 35976226 PMCID: PMC9531946 DOI: 10.7554/elife.76870] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Single-molecule imaging provides a powerful way to study biochemical processes in live cells, yet it remains challenging to track single molecules while simultaneously detecting their interactions. Here, we describe a novel property of rhodamine dyes, proximity-assisted photoactivation (PAPA), in which one fluorophore (the 'sender') can reactivate a second fluorophore (the 'receiver') from a dark state. PAPA requires proximity between the two fluorophores, yet it operates at a longer average intermolecular distance than Förster resonance energy transfer (FRET). We show that PAPA can be used in live cells both to detect protein-protein interactions and to highlight a subpopulation of labeled protein complexes in which two different labels are in proximity. In proof-of-concept experiments, PAPA detected the expected correlation between androgen receptor self-association and chromatin binding at the single-cell level. These results establish a new way in which a photophysical property of fluorophores can be harnessed to study molecular interactions in single-molecule imaging of live cells.
Collapse
Affiliation(s)
- Thomas GW Graham
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - John Joseph Ferrie
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Gina M Dailey
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Robert Tjian
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
42
|
Deng H, Lei Q, Yang N, Dai S, Peng H, Yang K, Xiao Z, Wang D, Yu Z, Li N, Li W. Expanded Application of a Photoaffinity Probe to Study Epidermal Growth Factor Receptor Tyrosine Kinase with Functional Activity. Anal Chem 2022; 94:10118-10126. [PMID: 35729862 DOI: 10.1021/acs.analchem.2c01340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The abnormal activation of the epidermal growth factor receptor (EGFR) is strongly associated with cancer invasion and metastasis. Tools and methods are required to study and visualize EGFR activation under (patho)physiological conditions. Here, we report the development of a two-step photoaffinity probe (HX101) by incorporation of a diazirine as a photoreactive group and an alkyne as a ligation handle to quantitively study EGFR kinase activity in native cellular contexts and human tissue slices. HX101 is a multifunctional probe based on the pharmacophore of the EGFR tyrosine kinase inhibitor (EGFR-TKI) and can covalently target the EGFR upon photoactivation. The incorporated alkyne serves as a versatile ligation handle and enables HX101 to introduce distinct reporter groups (e.g., fluorophore and biotin) via click chemistry. With variable reporter tags, HX101 enables visualization and target engagement studies of the active EGFR in a panel of cancer cells using flow cytometry, confocal microscopy, and mass spectrometry. Furthermore, as a proof of concept study, we applied HX101 in stochastic optical reconstruction microscopy super-resolution imaging to study EGFR activation in live cells. Importantly, HX101 was also applied to visualize EGFR mutant activity in tumor tissues from lung cancer patients for prediction of EGFR-TKI sensitivity. Altogether, our results demonstrate the wide application of a selective photoaffinity probe in multi-modal assessment/visualization of EGFR activity in both live cells and tissue slices. We anticipate that these diverse applications can facilitate the translation of a strategically functionalized probe into medical use.
Collapse
Affiliation(s)
- Hui Deng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qian Lei
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Na Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shengkun Dai
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Huipai Peng
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Kai Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zhaolin Xiao
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dongpeng Wang
- Biological Science Instruments Division, Nikon Instruments (Shanghai), Chengdu, Sichuan, 610041, China
| | - Zhiyi Yu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Nan Li
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.,Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
43
|
Komatsu R, Tanimoto Y, Ando K, Yasuhara K, Iwasaki Y, Hayashi F, Morigaki K. Nanofluidic Model Membrane for the Single-Molecule Observation of Membrane Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:7234-7243. [PMID: 35641430 DOI: 10.1021/acs.langmuir.2c00724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Membrane proteins play essential roles in the cell, and they constitute one of the most important targets of drugs. Studying membrane proteins in a controlled model membrane environment can provide unambiguous, quantitative information on their molecular properties and functions. However, reconstituting membrane proteins in a model system poses formidable technological challenges. Here, we developed a novel model membrane platform for highly sensitive observation of membrane proteins by combining a micropatterned lipid membrane and a nanofluidic channel. A micropatterned model membrane was generated by lithographically integrating a polymerized lipid bilayer and a natural (fluid) lipid bilayer. A nanofluidic channel having a defined thickness was formed between the fluid bilayer and a polydimethylsiloxane (PDMS) slab by attaching the polymeric bilayer and PDMS slab using an adhesion layer composed of silica nanoparticles that are coated with a biocompatible polymer brush. As we reconstituted rhodopsin (Rh), a G-protein-coupled receptor (GPCR), from a detergent-solubilized state into the fluid bilayer, only successfully reconstituted Rh molecules diffused laterally in the lipid bilayer and migrated into the nanogap junction, where they could be observed with a vastly improved signal-to-background ratio. The nanogap junction effectively separates the sites of reconstitution and observation and provides a novel platform for studying the molecular properties and functions of membrane proteins at the single-molecular level.
Collapse
Affiliation(s)
- Ryota Komatsu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Yasushi Tanimoto
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Koji Ando
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuma Yasuhara
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
- Center for Digital Green-innovation, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Yasuhiko Iwasaki
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita 564-0836, Japan
| | - Fumio Hayashi
- Graduate School of Science, Kobe University, Kobe 657-8501, Japan
| | - Kenichi Morigaki
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
- Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
44
|
Wollman AJM, Fournier C, Llorente-Garcia I, Harriman O, Payne-Dwyer AL, Shashkova S, Zhou P, Liu TC, Ouaret D, Wilding J, Kusumi A, Bodmer W, Leake MC. Critical roles for EGFR and EGFR-HER2 clusters in EGF binding of SW620 human carcinoma cells. J R Soc Interface 2022; 19:20220088. [PMID: 35612280 PMCID: PMC9131850 DOI: 10.1098/rsif.2022.0088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor (EGF) signalling regulates normal epithelial and other cell growth, with EGF receptor (EGFR) overexpression reported in many cancers. However, the role of EGFR clusters in cancer and their dependence on EGF binding is unclear. We present novel single-molecule total internal reflection fluorescence microscopy of (i) EGF and EGFR in living cancer cells, (ii) the action of anti-cancer drugs that separately target EGFR and human EGFR2 (HER2) on these cells and (iii) EGFR–HER2 interactions. We selected human epithelial SW620 carcinoma cells for their low level of native EGFR expression, for stable transfection with fluorescent protein labelled EGFR, and imaged these using single-molecule localization microscopy to quantify receptor architectures and dynamics upon EGF binding. Prior to EGF binding, we observe pre-formed EGFR clusters. Unexpectedly, clusters likely contain both EGFR and HER2, consistent with co-diffusion of EGFR and HER2 observed in a different model CHO-K1 cell line, whose stoichiometry increases following EGF binding. We observe a mean EGFR : EGF stoichiometry of approximately 4 : 1 for plasma membrane-colocalized EGFR–EGF that we can explain using novel time-dependent kinetics modelling, indicating preferential ligand binding to monomers. Our results may inform future cancer drug developments.
Collapse
Affiliation(s)
- Adam J M Wollman
- Department of Physics, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Charlotte Fournier
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK.,Science and Technology Group, Okinawa Institute of Science and Technology Graduate University (OIST), 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | | | - Oliver Harriman
- Department of Physics, Clarendon Laboratory, University of Oxford, Oxford OX1 3PU, UK
| | | | | | - Peng Zhou
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Ta-Chun Liu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Djamila Ouaret
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Jenny Wilding
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Akihiro Kusumi
- Membrane Cooperativity Unit, OIST, 1919 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Walter Bodmer
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| | - Mark C Leake
- Department of Physics, University of York, York, UK.,Department of Biology, University of York, York, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
45
|
Mutlu L, Tymon-Rosario J, Harold J, Menderes G. Targeted treatment options for the management of metastatic/persistent and recurrent cervical cancer. Expert Rev Anticancer Ther 2022; 22:633-645. [PMID: 35533682 DOI: 10.1080/14737140.2022.2075348] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Cervical cancer is the overall fourth most common malignancy and the fourth most common cause of cancer related deaths worldwide. Despite vaccination and screening programs, many women continue to present with advanced stage cervical cancer, wherein the treatment options have been limited. AREAS COVERED In this review, immunotherapy and the potential targeted therapies that have demonstrated promise in the treatment of persistent, recurrent, and metastatic cervical cancer are discussed. EXPERT OPINION Our global goal in the gynecologic oncology community is to eliminate cervical cancer, by increasing the uptake of preventive vaccination and screening programs. For unfortunate patients who present with metastatic, persistent, and recurrent cervical cancer, pembrolizumab with chemotherapy, with or without bevacizumab is the new first line therapy for PD-L1 positive patients. For this patient population as a second line therapy, tisotumab vedotin (i.e. ADC) has shown significant efficacy in Phase II trials, leading to FDA approval. Combination regimens inclusive of immune checkpoint inhibitors, DNA damage repair inhibitors, antibody drug conjugates are potential breakthrough treatment strategies and are currently being investigated.
Collapse
Affiliation(s)
- Levent Mutlu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Joan Tymon-Rosario
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Justin Harold
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| | - Gulden Menderes
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, Smilow Cancer Hospital, Yale University, School of Medicine
| |
Collapse
|
46
|
Qiu MZ, Zhang Y, Guo Y, Guo W, Nian W, Liao W, Xu Z, Zhang W, Zhao HY, Wei X, Xue L, Tang W, Wu Y, Ren G, Wang L, Xi J, Jin Y, Li H, Hu C, Xu RH. Evaluation of Safety of Treatment With Anti-Epidermal Growth Factor Receptor Antibody Drug Conjugate MRG003 in Patients With Advanced Solid Tumors: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol 2022; 8:1042-1046. [PMID: 35511148 PMCID: PMC9073657 DOI: 10.1001/jamaoncol.2022.0503] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Question What is the safety and antitumor activity of MRG003 in patients with advanced solid tumors? Findings In this phase 1 clinical trial of 61 patients with advanced or metastatic solid tumors, treatment with MRG003 exhibited manageable safety and showed encouraging antitumor activity in squamous cell carcinomas of the head and neck and nasopharyngeal carcinoma, with a confirmed objective response rates of 40% and 44%, respectively. Meaning The study findings suggest the safety of treatment with MRG003 and an acceptable tolerance in most patients with epidermal growth factor receptor–expressing solid tumors, as well as encouraging antitumor activity in patients with squamous cell carcinomas of the head and neck and nasopharyngeal carcinoma. Importance The antibody drug conjugate drug MRG003 comprises an anti–epidermal growth factor receptor (EGFR) humanized immunoglobulin G1 monoclonal antibody that is conjugated with monomethyl auristatin E via a valine-citrulline linker. There is currently insufficient evidence of this drug's safety and efficacy. Objective To evaluate the safety and maximum tolerated dose of MRG003 in a phase 1a study and investigate the preliminary antitumor activity in EGFR-expressing patients in a phase 1b study. Design, Setting, and Participants This nonrandomized open-label, single-arm, phase 1, multicenter study of solid tumors was divided into 2 parts, phase 1a dose escalation and phase 1b dose expansion. Patients with advanced or metastatic solid tumors who had failed outcomes from or were not able to receive standard treatment were enrolled in phase 1a without EGFR prescreening. Phase 1b recruited EGFR-positive patients with refractory advanced squamous cell carcinomas of the head and neck (SCCHN), nasopharyngeal carcinoma (NPC), and colorectal cancer (CRC). This study was conducted at 7 Chinese centers between April 11, 2018, and March 29, 2021 (data cutoff date). Data analysis took place between April 2021 and June 2021. Interventions An intravenous dose of 0.1 to 2.5 mg/kg of MRG003 was administered every 3 weeks during phase 1a. During phase 1b, patients were administered the recommended dose identified in phase 1a. Main Outcomes and Measures The primary end points were dose-limiting toxic effects in phase 1a and objective response rate in phase 1b. The safety, tolerability, immunogenicity, and pharmacokinetics of MRG003 were assessed. Tumor assessment was evaluated by RECIST 1.1. Results Twenty-two patients (mean [range] age, 54.5 [32.0-67.0] years; 9 women [41%]) were enrolled in phase 1a and 39 patients (mean [range] age, 50.4 [27.0-75.0] years; 8 women [21%]) in phase 1b. The recommended dose was identified as 2.5 mg/kg. Eighty-nine percent of adverse events (AEs) were associated with MRG003 treatment, and most AEs were grade 1 to 2. Nineteen patients (31%) reported grade 3 or greater treatment-related AEs, including hyponatremia, leukocytopenia, neutropenia, increased aspartate aminotransferase levels, and febrile neutropenia. In phase 1a, 1 patient (5%) achieved a partial response, and 5 (23%) achieved stable disease. In phase 1b, 8 patients (21%) achieved a confirmed partial response, and 12 (31%) achieved stable disease. The objective response rates for SCCHN, NPC, and CRC were 40%, 44%, and 0%, and the disease control rates were 100%, 89%, and 25%, respectively. Conclusions and Relevance The findings of this nonrandomized clinical trial suggest that MRG003 showed a manageable safety profile and promising antitumor activity in patients with EGFR-positive NPC and SCCHN. Trial Registration Clinicaltrials.gov Identifier: NCT04868344
Collapse
Affiliation(s)
- Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yang Zhang
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ye Guo
- Department of Oncology, Shanghai East Hospital Tongji University School of Medicine, Shanghai, China
| | - Wei Guo
- Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqi Nian
- Chongqing Cancer Institute, Chongqing University Cancer Hospital, Chongqing, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongyuan Xu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenxue Zhang
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin, Tianjin, China
| | - Hong-Yun Zhao
- Department of Clinical Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xiaoli Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liqiong Xue
- Department of Oncology, Shanghai East Hospital Tongji University School of Medicine, Shanghai, China
| | - Wenbo Tang
- Department of Oncology, Shanghai East Hospital Tongji University School of Medicine, Shanghai, China
| | - Yunteng Wu
- Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoxin Ren
- Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Wang
- Chongqing Cancer Institute, Chongqing University Cancer Hospital, Chongqing, China
| | - Jingle Xi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | - Hu Li
- Shanghai Miracogen Inc, Shanghai, China
| | | | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.,Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, People's Republic of China
| |
Collapse
|
47
|
Sato O, Sakai T, Choo YY, Ikebe R, Watanabe TM, Ikebe M. Mitochondria-associated myosin 19 processively transports mitochondria on actin tracks in living cells. J Biol Chem 2022; 298:101883. [PMID: 35367209 PMCID: PMC9065997 DOI: 10.1016/j.jbc.2022.101883] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondria are fundamentally important in cell function, and their malfunction can cause the development of cancer, cardiovascular disease, and neuronal disorders. Myosin 19 (Myo19) shows discrete localization with mitochondria and is thought to play an important role in mitochondrial dynamics and function; however, the function of Myo19 in mitochondrial dynamics at the cellular and molecular levels is poorly understood. Critical missing information is whether Myo19 is a processive motor that is suitable for transportation of mitochondria. Here, we show for the first time that single Myo19 molecules processively move on actin filaments and can transport mitochondria in cells. We demonstrate that Myo19 dimers having a leucine zipper processively moved on cellular actin tracks in demembraned cells with a velocity of 50 to 60 nm/s and a run length of ∼0.4 μm, similar to the movement of isolated mitochondria from Myo19 dimer-transfected cells on actin tracks, suggesting that the Myo19 dimer can transport mitochondria. Furthermore, we show single molecules of Myo19 dimers processively moved on single actin filaments with a large step size of ∼34 nm. Importantly, WT Myo19 single molecules without the leucine zipper processively move in filopodia in living cells similar to Myo19 dimers, whereas deletion of the tail domain abolished such active movement. These results suggest that Myo19 can processively move on actin filaments when two Myo19 monomers form a dimer, presumably as a result of tail-tail association. In conclusion, Myo19 molecules can directly transport mitochondria on actin tracks within living cells.
Collapse
Affiliation(s)
- Osamu Sato
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Tsuyoshi Sakai
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Young-Yeon Choo
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Reiko Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA
| | - Tomonobu M Watanabe
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, University of Texas at Tyler Health Science Center, Tyler, Texas, USA.
| |
Collapse
|
48
|
Chauhan N, Saxena K, Jain U. Single molecule detection; from microscopy to sensors. Int J Biol Macromol 2022; 209:1389-1401. [PMID: 35413320 DOI: 10.1016/j.ijbiomac.2022.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 01/31/2023]
Abstract
Single molecule detection is necessary to find out physical, chemical properties and their mechanism involved in the normal functioning of body cells. In this way, they can provide a new direction to the healthcare system. Various techniques have been developed and employed for their successful detection. Herein, we have emphasized various traditional methods as well as biosensing technology which offer single molecule sensitivity. The various methods including plasmonic resonance, nanopores, whispering gallery mode, Simoa assay and recognition tunneling are discussed in the initial part which has been followed by a discussion about biosensor-based detection. Plasmonic, SERS, CRISPR/Cas, and other types of biosensors are focused in this review and found to be highly sensitive for single molecule detection. This review provides an overview of progression in different techniques employed for single molecule detection.
Collapse
Affiliation(s)
- Nidhi Chauhan
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida 201313, U.P., India
| | - Kirti Saxena
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida 201313, U.P., India
| | - Utkarsh Jain
- Amity Institute of Nanotechnology (AINT), Amity University Uttar Pradesh (AUUP), Noida 201313, U.P., India.
| |
Collapse
|
49
|
Liu Y, Li B, Wang YJ, Fan Z, Du Y, Li B, Liu YJ, Liu B. In Situ Single-Molecule Imaging of MicroRNAs in Switchable Migrating Cells under Biomimetic Confinement. Anal Chem 2022; 94:4030-4038. [PMID: 35213802 DOI: 10.1021/acs.analchem.1c05223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Spatial imaging of RNAs in single cells is extremely charming for deciphering of regulatory mechanisms in multiple migration modes during tumor metastasis. Herein, enzyme-free-mediated cascade amplified nanoprobes were designed for in situ single-molecule imaging of dual-microRNAs (miRNAs) in switchable migrating cells. Differential expression and localization of dual-miRNAs were clearly exhibited in multiple cell lines attributed to enhanced sensitivity via the cascade signal amplification strategy. Significantly, in situ three-dimensional (3D) imaging of dual-miRNAs in transition of cell migration phenotypes was successfully reconstructed in both non-confined and confined microenvironments in vitro, of which differential spatial distribution was observed in a single cell. This is very promising for exploring key roles of spatial RNA distribution in migrating cells at the single-molecule level, which will advance revealing the molecular mechanism and physical principle in 3D cell migration in vivo.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Binxiao Li
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Ya-Jun Wang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Zihui Fan
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Yang Du
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Bin Li
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Yan-Jun Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Lab of Molecular Engineering of Polymers, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai 200433, China
| |
Collapse
|
50
|
Wang Q, Zhang Q, He H, Feng Z, Mao J, Hu X, Wei X, Bi S, Qin G, Wang X, Ge B, Yu D, Ren H, Huang F. Carbon Dot Blinking Fingerprint Uncovers Native Membrane Receptor Organizations via Deep Learning. Anal Chem 2022; 94:3914-3921. [PMID: 35188385 DOI: 10.1021/acs.analchem.1c04947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Oligomeric organization of G protein-coupled receptors is proposed to regulate receptor signaling and function, yet rapid and precise identification of the oligomeric status especially for native receptors on a cell membrane remains an outstanding challenge. By using blinking carbon dots (CDs), we now develop a deep learning (DL)-based blinking fingerprint recognition method, named deep-blinking fingerprint recognition (BFR), which allows automatic classification of CD-labeled receptor organizations on a cell membrane. This DL model integrates convolutional layers, long-short-term memory, and fully connected layers to extract time-dependent blinking features of CDs and is trained to a high accuracy (∼95%) for identifying receptor organizations. Using deep blinking fingerprint recognition, we found that CXCR4 mainly exists as 87.3% monomers, 12.4% dimers, and <1% higher-order oligomers on a HeLa cell membrane. We further demonstrate that the heterogeneous organizations can be regulated by various stimuli at different degrees. The receptor-binding ligands, agonist SDF-1α and antagonist AMD3100, can induce the dimerization of CXCR4 to 33.1 and 20.3%, respectively. In addition, cytochalasin D, which inhibits actin polymerization, similarly prompts significant dimerization of CXCR4 to 30.9%. The multi-pathway organization regulation will provide an insight for understanding the oligomerization mechanism of CXCR4 as well as for elucidating their physiological functions.
Collapse
Affiliation(s)
- Qian Wang
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Qian Zhang
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Hua He
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Zhenzhen Feng
- Technical Center of Qingdao Customs District, Qingdao 266500, China
| | - Jian Mao
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiang Hu
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiaoyun Wei
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Simin Bi
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Guangyong Qin
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiaojuan Wang
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Baosheng Ge
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Daoyong Yu
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Hao Ren
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Fang Huang
- State Key Laboratory of Heavy Oil Processing and College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|