1
|
Kishi K, Nigorikawa K, Hasegawa Y, Ohta Y, Matsugi E, Matsumoto D, Nomura W. Cell cycle-dependent regulation of CRISPR-Cas9 repetitive activation by anti-CRISPR and Cdt1 fusion in the CRISPRa system. FEBS Lett 2025; 599:828-837. [PMID: 39739523 DOI: 10.1002/1873-3468.15090] [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: 11/04/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025]
Abstract
CRISPR-Cas9 is a widely used genome-editing tool. We previously developed a method with improved homology-directed repair efficiency and reduced off-target effects by utilizing a fusion protein of AcrIIA4, a Cas9 inhibitor, and Cdt1, which accumulates in the G1 phase and activates Cas9 only in the S/G2 phase. However, it is unknown whether Cas9 inhibition by AcrIIA4 + Cdt1 occurs repeatedly in the G1 phase as the cell cycle progresses. In this study, we used the CRISPRa system to monitor changes in the interaction between Cas9 and AcrIIA4 + Cdt1 at single-cell resolution and in real time. Our findings are among the few examples of successful detection of fluctuating protein-protein interactions that oscillate over time.
Collapse
Affiliation(s)
- Kanae Kishi
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
- School of Pharmaceutical Sciences, Hiroshima University, Japan
| | - Yuki Hasegawa
- School of Pharmaceutical Sciences, Hiroshima University, Japan
| | - Yusaku Ohta
- Bioimage Informatics Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
- Interdisciplinary Research Unit, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Japan
| | - Erina Matsugi
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Daisuke Matsumoto
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
- School of Pharmaceutical Sciences, Hiroshima University, Japan
| | - Wataru Nomura
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
- School of Pharmaceutical Sciences, Hiroshima University, Japan
| |
Collapse
|
2
|
Ujlaky-Nagy L, Szöllősi J, Vereb G. EGFR-HER2 Transactivation Viewed in Space and Time Through the Versatile Spectacles of Imaging Cytometry-Implications for Targeted Therapy. Cytometry A 2025; 107:187-202. [PMID: 40052543 DOI: 10.1002/cyto.a.24922] [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: 09/27/2024] [Revised: 12/03/2024] [Accepted: 01/24/2025] [Indexed: 04/11/2025]
Abstract
Ligand-induced formation of signaling platforms composed of homo- and/or heterodimers of receptor tyrosine kinases is considered essential for their activation and consequential contribution to the progression of many cancers. Epidermal Growth Factor Receptor (EGFR) acts as a signal receiver upon EGF binding and produces mitogenic input for many cells also through receptor-heterodimerization with its ligandless partner, Human Epidermal growth factor Receptor 2 (HER2). Ligand-driven transactivation is a key step leading to changes in the cell surface pattern of EGFR and HER2; their interaction plays a key role in various malignancies, especially when HER2 molecules are overexpressed. Our clinically relevant model system is the SK-BR-3 breast tumor cell line, overexpressing HER2 and moderately expressing EGFR. This cell line shows significant dependency on EGF-driven HER2 signaling. We studied changes in the interaction between EGFR and HER2 in the cell membrane upon EGF binding, applying various biophysical approaches with different time scales. Changes in molecular proximity were characterized by fluorescence lifetime imaging microscopy (FLIM) techniques assessing Förster resonance energy transfer (FRET), which confirmed the ligand-enhanced interaction of EGFR and HER2, followed by an increase in HER2 homoassociation. EGF binding and transactivation were reflected in the phosphorylation of both receptor types as well. At the same time, superresolution Airyscan microscopy and fluorescence correlation and cross-correlation spectroscopy (FCS/FCCS), sensitive to changes in the size of stationary and diffusing aggregates, respectively, have revealed cyclic increases in the aggregation and stable co-diffusion of membrane-localized HER2, possibly caused by internalization and recycling, eventually leading to a new equilibrium. Such dynamic fluctuation of receptor interaction may open a window for the binding of therapeutic antibodies that are aimed at inhibiting heterodimerization, such as pertuzumab. The complementary array of state-of-the-art imaging cytometry approaches thus demonstrates a spatiotemporal pattern of spontaneous and induced receptor aggregation states that could provide mechanistic insights into the potential success of targeted therapies directed at the HER family of receptor tyrosine kinases.
Collapse
Affiliation(s)
- László Ujlaky-Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group (Hungarian Research Network - University of Debrecen), Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group (Hungarian Research Network - University of Debrecen), Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - György Vereb
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-DE Cell Biology and Signaling Research Group (Hungarian Research Network - University of Debrecen), Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
3
|
Hsiao WWW, Pham UK, Le TN, Lam XM, Chiang WH. Advances in aggregation-induced emission luminogens for biomedicine: From luminescence mechanisms to diagnostic applications. Biosens Bioelectron 2025; 270:116942. [PMID: 39566330 DOI: 10.1016/j.bios.2024.116942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/26/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Advancements in early detection have demonstrated the significance of biomarkers as indicators of health and disease. Traditional detection methods often face limitations, such as low sensitivity and time consumption. Fluorescence-based techniques are considered promising approaches because of their noninvasiveness and rapid response. However, these conventional methods have some drawbacks, such as low quantum yield, photobleaching, and aggregation-caused quenching. Recently, aggregation-induced emission (AIE) has emerged as a potential alternative, characterized by luminous emission upon aggregation, thus improving detection sensitivity and stability. This review explores the recent advancements in AIE luminogens (AIEgens) in biomedical engineering, with a particular focus on their application in biomarker detection. Here, we discuss the different types of AIE mechanisms and their advantages in disease diagnosis and imaging. In addition, we summarize the development of various AIEgen-based probes for the detection of diverse biomarkers. Finally, we address the remaining challenges and future directions for AIE materials in modern biomedical engineering, emphasizing the potential of AIEgens in biomarker detection and disease diagnosis strategies.
Collapse
Affiliation(s)
- Wesley Wei-Wen Hsiao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan.
| | - Uyen Khanh Pham
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan
| | - Trong-Nghia Le
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, 106319, Taiwan
| | - Xuan Mai Lam
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 106335, Taiwan; Sustainable Electrochemical Energy Development (SEED) Center, National Taiwan University of Science and Technology, Taipei City, 106335, Taiwan
| |
Collapse
|
4
|
Soleja N, Mohsin M. Exploring the landscape of FRET-based molecular sensors: Design strategies and recent advances in emerging applications. Biotechnol Adv 2024; 77:108466. [PMID: 39419421 DOI: 10.1016/j.biotechadv.2024.108466] [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: 02/28/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Probing biological processes in living organisms that could provide one-of-a-kind insights into real-time alterations of significant physiological parameters is a formidable task that calls for specialized analytic devices. Classical biochemical methods have significantly aided our understanding of the mechanisms that regulate essential biological processes. These methods, however, are typically insufficient for investigating transient molecular events since they focus primarily on the end outcome. Fluorescence resonance energy transfer (FRET) microscopy is a potent tool used for exploring non-invasively real-time dynamic interactions between proteins and a variety of biochemical signaling events using sensors that have been meticulously constructed. Due to their versatility, FRET-based sensors have enabled the rapid and standardized assessment of a large array of biological variables, facilitating both high-throughput research and precise subcellular measurements with exceptional temporal and spatial resolution. This review commences with a brief introduction to FRET theory and a discussion of the fluorescent molecules that can serve as tags in different sensing modalities for studies in chemical biology, followed by an outlining of the imaging techniques currently utilized to quantify FRET highlighting their strengths and shortcomings. The article also discusses the various donor-acceptor combinations that can be utilized to construct FRET scaffolds. Specifically, the review provides insights into the latest real-time bioimaging applications of FRET-based sensors and discusses the common architectures of such devices. There has also been discussion of FRET systems with multiplexing capabilities and multi-step FRET protocols for use in dual/multi-analyte detections. Future research directions in this exciting field are also mentioned, along with the obstacles and opportunities that lie ahead.
Collapse
Affiliation(s)
- Neha Soleja
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Mohd Mohsin
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
5
|
Gökerküçük EB, Tramier M, Bertolin G. Protocol for quantifying LC3B FRET biosensor activity in living cells using a broad-to-sensitive data analysis pipeline. STAR Protoc 2024; 5:103181. [PMID: 39178110 PMCID: PMC11387700 DOI: 10.1016/j.xpro.2024.103181] [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: 01/08/2024] [Revised: 04/10/2024] [Accepted: 06/18/2024] [Indexed: 08/25/2024] Open
Abstract
Here, we present a protocol to comprehensively quantify autophagy initiation using the readout of the microtubule associated protein 1 light chain 3 beta (LC3B) Förster's resonance energy transfer (FRET) biosensor. We describe steps for cell seeding, transfection, FRET/FLIM (fluorescence lifetime imaging microscopy) imaging, and image analysis. This protocol can be useful in any physiology- or disease-related paradigm where the LC3B biosensor can be expressed to determine whether autophagy has been initiated or is stalled. The analysis pipeline presented here can be applied to any other genetically encoded FRET sensor imaged using FRET/FLIM. For complete details on the use and execution of this protocol, please refer to Gökerküçük et al.1.
Collapse
Affiliation(s)
- Elif Begüm Gökerküçük
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France.
| | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France.
| |
Collapse
|
6
|
Yang Q, Miki T. Characterization of peptide-fused protein assemblies in living cells. Methods Enzymol 2024; 697:293-319. [PMID: 38816127 DOI: 10.1016/bs.mie.2024.01.022] [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: 06/01/2024]
Abstract
Assembly of de novo peptides designed from scratch is in a semi-rational manner and creates artificial supramolecular structures with unique properties. Considering that the functions of various proteins in living cells are highly regulated by their assemblies, building artificial assemblies within cells holds the potential to simulate the functions of natural protein assemblies and engineer cellular activities for controlled manipulation. How can we evaluate the self-assembly of designed peptides in cells? The most effective approach involves the genetic fusion of fluorescent proteins (FPs). Expressing a self-assembling peptide fused with an FP within cells allows for evaluating assemblies through fluorescence signal. When µm-scale assemblies such as condensates are formed, the peptide assemblies can be directly observed by imaging. For sub-µm-scale assemblies, fluorescence correlation spectroscopy analysis is more practical. Additionally, the fluorescence resonance energy transfer (FRET) signal between FPs is valuable evidence of proximity. The decrease in fluorescence anisotropy associated with homo-FRET reveals the properties of self-assembly. Furthermore, by combining two FPs, one acting as a donor and the other as an acceptor, the heteromeric interaction between two different components can be studied through the FRET signal. In this chapter, we provide detailed protocols, from designing and constructing plasmid DNA expressing the peptide-fused protein to analysis of self-assembly in living cells.
Collapse
Affiliation(s)
- Qinxuan Yang
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takayuki Miki
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan; School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.
| |
Collapse
|
7
|
Riggi M, Torrez RM, Iwasa JH. 3D animation as a tool for integrative modeling of dynamic molecular mechanisms. Structure 2024; 32:122-130. [PMID: 38183978 PMCID: PMC10872329 DOI: 10.1016/j.str.2023.12.007] [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: 09/22/2023] [Revised: 11/01/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024]
Abstract
As the scientific community accumulates diverse data describing how molecular mechanisms occur, creating and sharing visual models that integrate the richness of this information has become increasingly important to help us explore, refine, and communicate our hypotheses. Three-dimensional (3D) animation is a powerful tool to capture dynamic hypotheses that are otherwise difficult or impossible to visualize using traditional 2D illustration techniques. This perspective discusses the current and future roles that 3D animation can play in the research sphere.
Collapse
Affiliation(s)
- Margot Riggi
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Rachel M Torrez
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Janet H Iwasa
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
8
|
Caron C, Bertolin G. Cristae shaping and dynamics in mitochondrial function. J Cell Sci 2024; 137:jcs260986. [PMID: 38197774 DOI: 10.1242/jcs.260986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Mitochondria are multifunctional organelles of key importance for cell homeostasis. The outer mitochondrial membrane (OMM) envelops the organelle, and the inner mitochondrial membrane (IMM) is folded into invaginations called cristae. As cristae composition and functions depend on the cell type and stress conditions, they recently started to be considered as a dynamic compartment. A number of proteins are known to play a role in cristae architecture, such as OPA1, MIC60, LETM1, the prohibitin (PHB) complex and the F1FO ATP synthase. Furthermore, phospholipids are involved in the maintenance of cristae ultrastructure and dynamics. The use of new technologies, including super-resolution microscopy to visualize cristae dynamics with superior spatiotemporal resolution, as well as high-content techniques and datasets have not only allowed the identification of new cristae proteins but also helped to explore cristae plasticity. However, a number of open questions remain in the field, such as whether cristae-resident proteins are capable of changing localization within mitochondria, or whether mitochondrial proteins can exit mitochondria through export. In this Review, we present the current view on cristae morphology, stability and composition, and address important outstanding issues that might pave the way to future discoveries.
Collapse
Affiliation(s)
- Claire Caron
- Univ. Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| | - Giulia Bertolin
- Univ. Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000 Rennes, France
| |
Collapse
|
9
|
Fueyo-González F, Vilanova G, Ningoo M, Marjanovic N, González-Vera JA, Orte Á, Fribourg M. Small-molecule TIP60 inhibitors enhance regulatory T cell induction through TIP60-P300 acetylation crosstalk. iScience 2023; 26:108491. [PMID: 38094248 PMCID: PMC10716589 DOI: 10.1016/j.isci.2023.108491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/12/2023] [Accepted: 11/14/2023] [Indexed: 12/29/2023] Open
Abstract
Foxp3 acetylation is essential to regulatory T (Treg) cell stability and function, but pharmacologically increasing it remains an unmet challenge. Here, we report that small-molecule compounds that inhibit TIP60, an acetyltransferase known to acetylate Foxp3, unexpectedly increase Foxp3 acetylation and Treg induction. Utilizing a dual experimental/computational approach combined with a newly developed FRET-based methodology compatible with flow cytometry to measure Foxp3 acetylation, we unraveled the mechanism of action of these small-molecule compounds in murine and human Treg induction cell cultures. We demonstrate that at low-mid concentrations they activate TIP60 to acetylate P300, a different acetyltransferase, which in turn increases Foxp3 acetylation, thereby enhancing Treg cell induction. These results reveal a potential therapeutic target relevant to autoimmunity and transplant.
Collapse
Affiliation(s)
- Francisco Fueyo-González
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Guillermo Vilanova
- LaCàN, Universitat Politècnica de Catalunya-BarcelonaTech, 08034 Barcelona Spain
| | - Mehek Ningoo
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nada Marjanovic
- Deparment of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan A. González-Vera
- Deparment of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nanoscopy-UGR Laboratory, Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain
| | - Ángel Orte
- Deparment of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nanoscopy-UGR Laboratory, Departamento de Fisicoquímica, Unidad de Excelencia de Química Aplicada a Biomedicina y Medioambiente, Facultad de Farmacia, Universidad de Granada, Campus Cartuja, 18071 Granada, Spain
| | - Miguel Fribourg
- Translational Transplant Research Center, Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
10
|
Starling T, Carlon-Andres I, Iliopoulou M, Kraemer B, Loidolt-Krueger M, Williamson DJ, Padilla-Parra S. Multicolor lifetime imaging and its application to HIV-1 uptake. Nat Commun 2023; 14:4994. [PMID: 37591879 PMCID: PMC10435470 DOI: 10.1038/s41467-023-40731-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 08/04/2023] [Indexed: 08/19/2023] Open
Abstract
Simultaneous imaging of nine fluorescent proteins is demonstrated in a single acquisition using fluorescence lifetime imaging microscopy combined with pulsed interleaved excitation of three laser lines. Multicolor imaging employing genetically encodable fluorescent proteins permits spatio-temporal live cell imaging of multiple cues. Here, we show that multicolor lifetime imaging allows visualization of quadruple labelled human immunodeficiency viruses on host cells that in turn are also labelled with genetically encodable fluorescent proteins. This strategy permits to simultaneously visualize different sub-cellular organelles (mitochondria, cytoskeleton, and nucleus) during the process of virus entry with the potential of imaging up to nine different spectral channels in living cells.
Collapse
Affiliation(s)
- Tobias Starling
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, UK
| | - Irene Carlon-Andres
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, UK
| | - Maro Iliopoulou
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Randall Division of Cell and Molecular Biophysics and Department of Physics, King's College London, London, UK
| | - Benedikt Kraemer
- PicoQuant GmbH, Rudower Chaussee 29 (IGZ), 12489, Berlin, Germany
| | | | - David J Williamson
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, UK
| | - Sergi Padilla-Parra
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, UK.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
11
|
Smith JT, Sinsuebphon N, Rudkouskaya A, Michalet X, Intes X, Barroso M. In vivo quantitative FRET small animal imaging: Intensity versus lifetime-based FRET. BIOPHYSICAL REPORTS 2023; 3:100110. [PMID: 37251213 PMCID: PMC10209493 DOI: 10.1016/j.bpr.2023.100110] [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: 01/26/2023] [Accepted: 04/27/2023] [Indexed: 05/31/2023]
Abstract
Förster resonance energy transfer (FRET) microscopy is used in numerous biophysical and biomedical applications to monitor inter- and intramolecular interactions and conformational changes in the 2-10 nm range. FRET is currently being extended to in vivo optical imaging, its main application being in quantifying drug-target engagement or drug release in animal models of cancer using organic dye or nanoparticle-labeled probes. Herein, we compared FRET quantification using intensity-based FRET (sensitized emission FRET analysis with the three-cube approach using an IVIS imager) and macroscopic fluorescence lifetime (MFLI) FRET using a custom system using a time-gated-intensified charge-coupled device, for small animal optical in vivo imaging. The analytical expressions and experimental protocols required to quantify the product f D E of the FRET efficiency E and the fraction of donor molecules involved in FRET, f D , are described in detail for both methodologies. Dynamic in vivo FRET quantification of transferrin receptor-transferrin binding was acquired in live intact nude mice upon intravenous injection of a near-infrared-labeled transferrin FRET pair and benchmarked against in vitro FRET using hybridized oligonucleotides. Even though both in vivo imaging techniques provided similar dynamic trends for receptor-ligand engagement, we demonstrate that MFLI-FRET has significant advantages. Whereas the sensitized emission FRET approach using the IVIS imager required nine measurements (six of which are used for calibration) acquired from three mice, MFLI-FRET needed only one measurement collected from a single mouse, although a control mouse might be needed in a more general situation. Based on our study, MFLI therefore represents the method of choice for longitudinal preclinical FRET studies such as that of targeted drug delivery in intact, live mice.
Collapse
Affiliation(s)
- Jason T. Smith
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, New York
| | - Nattawut Sinsuebphon
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, New York
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Xavier Michalet
- Department of Chemistry & Biochemistry, University of California at Los Angeles, Los Angeles, California
| | - Xavier Intes
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, New York
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
12
|
Smith JT, Sinsuebphon N, Rudkouskaya A, Michalet X, Intes X, Barroso M. in vivo quantitative FRET small animal imaging: intensity versus lifetime-based FRET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525411. [PMID: 36747671 PMCID: PMC9900789 DOI: 10.1101/2023.01.24.525411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Förster Resonance Energy Transfer (FRET) microscopy is used in numerous biophysical and biomedical applications to monitor inter- and intramolecular interactions and conformational changes in the 2-10 nm range. FRET is currently being extended to in vivo optical imaging, its main application being in quantifying drug-target engagement or drug release in animal models of cancer using organic dye or nanoparticle-labeled probes. Herein, we compared FRET quantification using intensity-based FRET (sensitized emission FRET analysis with the 3-cube approach using an IVIS imager) and macroscopic fluorescence lifetime (MFLI) FRET using a custom system using a time-gated ICCD, for small animal optical in vivo imaging. The analytical expressions and experimental protocols required to quantify the product f D E of the FRET efficiency E and the fraction of donor molecules involved in FRET, f D , are described in detail for both methodologies. Dynamic in vivo FRET quantification of transferrin receptor-transferrin binding was acquired in live intact nude mice upon intravenous injection of near infrared-labeled transferrin FRET pair and benchmarked against in vitro FRET using hybridized oligonucleotides. Even though both in vivo imaging techniques provided similar dynamic trends for receptor-ligand engagement, we demonstrate that MFLI FRET has significant advantages. Whereas the sensitized emission FRET approach using the IVIS imager required 9 measurements (6 of which are used for calibration) acquired from three mice, MFLI FRET needed only one measurement collected from a single mouse, although a control mouse might be needed in a more general situation. Based on our study, MFLI therefore represents the method of choice for longitudinal preclinical FRET studies such as that of targeted drug delivery in intact, live mice.
Collapse
Affiliation(s)
- Jason T. Smith
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Present address: Elephas, 1 Erdman Pl., Madison, WI 53705, USA
| | - Nattawut Sinsuebphon
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Present address: Assistive Technology and Medical Devices Research Center, National Science and Technology Development Agency, 12120 Pathum Thani, Thailand
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Xavier Michalet
- Department of Chemistry & Biochemistry, University of California at Los Angeles, Los Angeles, California, CA 90095, USA
| | - Xavier Intes
- Center for Modeling, Simulation and Imaging in Medicine (CeMSIM), Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
13
|
Camponeschi F, Banci L. Metal trafficking in the cell: Combining atomic resolution with cellular dimension. FEBS Lett 2023; 597:122-133. [PMID: 36285633 DOI: 10.1002/1873-3468.14524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023]
Abstract
Metals are widely present in biological systems as simple ions or complex cofactors, and are involved in a variety of processes essential for life. Their transport inside cells and insertion into the binding sites of the proteins that need metals to function occur through complex and selective pathways involving dedicated multiprotein machineries specifically and transiently interacting with each other, often sharing the coordination of metal ions and/or cofactors. The understanding of these machineries requires integrated approaches, ranging from bioinformatics to experimental investigations, possibly in the cellular context. In this review, we report two case studies where the use of integrated in vitro and in cellulo approaches is necessary to clarify at atomic resolution essential aspects of metal trafficking in cells.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy.,Department of Chemistry, University of Florence, Italy
| |
Collapse
|
14
|
Ley-Ngardigal S, Bertolin G. Approaches to monitor ATP levels in living cells: where do we stand? FEBS J 2022; 289:7940-7969. [PMID: 34437768 DOI: 10.1111/febs.16169] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
ATP is the most universal and essential energy molecule in cells. This is due to its ability to store cellular energy in form of high-energy phosphate bonds, which are extremely stable and readily usable by the cell. This energy is key for a variety of biological functions such as cell growth and division, metabolism, and signaling, and for the turnover of biomolecules. Understanding how ATP is produced and hydrolyzed with a spatiotemporal resolution is necessary to understand its functions both in physiological and in pathological contexts. In this review, first we will describe the organization of the electron transport chain and ATP synthase, the main molecular motor for ATP production in mitochondria. Second, we will review the biochemical assays currently available to estimate ATP quantities in cells, and we will compare their readouts, strengths, and weaknesses. Finally, we will explore the palette of genetically encoded biosensors designed for microscopy-based approaches, and show how their spatiotemporal resolution opened up the possibility to follow ATP levels in living cells.
Collapse
Affiliation(s)
- Seyta Ley-Ngardigal
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France.,LVMH Research Perfumes and Cosmetics, Saint-Jean-de-Braye, France
| | - Giulia Bertolin
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France
| |
Collapse
|
15
|
Kim H, Baek IY, Seong J. Genetically encoded fluorescent biosensors for GPCR research. Front Cell Dev Biol 2022; 10:1007893. [PMID: 36247000 PMCID: PMC9559200 DOI: 10.3389/fcell.2022.1007893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) regulate a wide range of physiological and pathophysiological cellular processes, thus it is important to understand how GPCRs are activated and function in various cellular contexts. In particular, the activation process of GPCRs is dynamically regulated upon various extracellular stimuli, and emerging evidence suggests the subcellular functions of GPCRs at endosomes and other organelles. Therefore, precise monitoring of the GPCR activation process with high spatiotemporal resolution is required to investigate the underlying molecular mechanisms of GPCR functions. In this review, we will introduce genetically encoded fluorescent biosensors that can precisely monitor the real-time GPCR activation process in live cells. The process includes the binding of extracellular GPCR ligands, conformational change of GPCR, recruitment of G proteins or β-arrestin, GPCR internalization and trafficking, and the GPCR-related downstream signaling events. We will introduce fluorescent GPCR biosensors based on a variety of strategies such as fluorescent resonance energy transfer (FRET), bioluminescence resonance energy transfer (BRET), circular permuted fluorescent protein (cpFP), and nanobody. We will discuss the pros and cons of these GPCR biosensors as well as their applications in GPCR research.
Collapse
Affiliation(s)
- Hyunbin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
| | - In-Yeop Baek
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, South Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, South Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
16
|
Barylko B, Hedde PN, Taylor CA, Binns DD, Huang YK, Molinaro G, Huber KM, Jameson DM, Albanesi JP. Palmitoylation-regulated interactions of the pseudokinase calmodulin kinase-like vesicle-associated with membranes and Arc/Arg3.1. Front Synaptic Neurosci 2022; 14:926570. [PMID: 35965782 PMCID: PMC9371321 DOI: 10.3389/fnsyn.2022.926570] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Calmodulin kinase-like vesicle-associated (CaMKv), a pseudokinase belonging to the Ca2+/calmodulin-dependent kinase family, is expressed predominantly in brain and neural tissue. It may function in synaptic strengthening during spatial learning by promoting the stabilization and enrichment of dendritic spines. At present, almost nothing is known regarding CaMKv structure and regulation. In this study we confirm prior proteomic analyses demonstrating that CaMKv is palmitoylated on Cys5. Wild-type CaMKv is enriched on the plasma membrane, but this enrichment is lost upon mutation of Cys5 to Ser. We further show that CaMKv interacts with another regulator of synaptic plasticity, Arc/Arg3.1, and that the interaction between these two proteins is weakened by mutation of the palmitoylated cysteine in CamKv.
Collapse
Affiliation(s)
- Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Per Niklas Hedde
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- Laboratory for Fluorescence Dynamics, University of California, Irvine, Irvine, CA, United States
| | - Clinton A. Taylor
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Derk D. Binns
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yu-Kai Huang
- Laboratory for Fluorescence Dynamics, University of California, Irvine, Irvine, CA, United States
| | - Gemma Molinaro
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kimberly M. Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David M. Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
17
|
David G, Reboutier D, Deschamps S, Méreau A, Taylor W, Padilla-Parra S, Tramier M, Audic Y, Paillard L. The RNA-binding proteins CELF1 and ELAVL1 cooperatively control the alternative splicing of CD44. Biochem Biophys Res Commun 2022; 626:79-84. [DOI: 10.1016/j.bbrc.2022.07.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/29/2022]
|
18
|
White D, Yang Q. Genetically Encoded ATP Biosensors for Direct Monitoring of Cellular ATP Dynamics. Cells 2022; 11:1920. [PMID: 35741049 PMCID: PMC9221525 DOI: 10.3390/cells11121920] [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: 05/26/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 12/06/2022] Open
Abstract
Adenosine 5'-triphosphate, or ATP, is the primary molecule for storing and transferring energy in cells. ATP is mainly produced via oxidative phosphorylation in mitochondria, and to a lesser extent, via glycolysis in the cytosol. In general, cytosolic glycolysis is the primary ATP producer in proliferative cells or cells subjected to hypoxia. On the other hand, mitochondria produce over 90% of cellular ATP in differentiated cells under normoxic conditions. Under pathological conditions, ATP demand rises to meet the needs of biosynthesis for cellular repair, signaling transduction for stress responses, and biochemical processes. These changes affect how mitochondria and cytosolic glycolysis function and communicate. Mitochondria undergo remodeling to adapt to the imbalanced demand and supply of ATP. Otherwise, a severe ATP deficit will impair cellular function and eventually cause cell death. It is suggested that ATP from different cellular compartments can dynamically communicate and coordinate to adapt to the needs in each cellular compartment. Thus, a better understanding of ATP dynamics is crucial to revealing the differences in cellular metabolic processes across various cell types and conditions. This requires innovative methodologies to record real-time spatiotemporal ATP changes in subcellular regions of living cells. Over the recent decades, numerous methods have been developed and utilized to accomplish this task. However, this is not an easy feat. This review evaluates innovative genetically encoded biosensors available for visualizing ATP in living cells, their potential use in the setting of human disease, and identifies where we could improve and expand our abilities.
Collapse
Affiliation(s)
- Donnell White
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Qinglin Yang
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
19
|
Valdez S, Robertson M, Qiang Z. Fluorescence Resonance Energy Transfer Measurements in Polymer Science: A Review. Macromol Rapid Commun 2022; 43:e2200421. [PMID: 35689335 DOI: 10.1002/marc.202200421] [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] [Received: 05/01/2022] [Revised: 06/06/2022] [Indexed: 12/27/2022]
Abstract
Fluorescence resonance energy transfer (FRET) is a non-invasive characterization method for studying molecular structures and dynamics, providing high spatial resolution at nanometer scale. Over the past decades, FRET-based measurements are developed and widely implemented in synthetic polymer systems for understanding and detecting a variety of nanoscale phenomena, enabling significant advances in polymer science. In this review, the basic principles of fluorescence and FRET are briefly discussed. Several representative research areas are highlighted, where FRET spectroscopy and imaging can be employed to reveal polymer morphology and kinetics. These examples include understanding polymer micelle formation and stability, detecting guest molecule release from polymer host, characterizing supramolecular assembly, imaging composite interfaces, and determining polymer chain conformations and their diffusion kinetics. Finally, a perspective on the opportunities of FRET-based measurements is provided for further allowing their greater contributions in this exciting area.
Collapse
Affiliation(s)
- Sara Valdez
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Mark Robertson
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Zhe Qiang
- School of Polymer Science and Engineering, University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| |
Collapse
|
20
|
Vu H, Woodcock JW, Krishnamurthy A, Obrzut J, Gilman JW, Coughlin EB. Visualization of Polymer Dynamics in Cellulose Nanocrystal Matrices Using Fluorescence Lifetime Measurements. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10793-10804. [PMID: 35179343 DOI: 10.1021/acsami.1c21906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Polymer nanocomposites containing self-assembled cellulose nanocrystals (CNCs) are ideal for advanced applications requiring both strength and toughness as the helicoidal structure of the CNCs deflects crack propagation and the polymer matrix dissipates impact energy. However, any adsorbed water layer surrounding the CNCs may compromise the interfacial adhesion between the polymer matrix and the CNCs, thus impacting stress transfer at that interface. Therefore, it is critical to study the role of water at the interface in connecting the polymer dynamics and the resulting mechanical performance of the nanocomposite. Here, we explore the effect of polymer confinement and water content on polymer dynamics in CNC nanocomposites by covalently attaching a fluorogenic water-sensitive dye to poly(diethylene glycol methyl ether methacrylate) (PMEO2MA), to provide insights into the observed mechanical performance. Utilizing fluorescence lifetime imaging microscopy (FLIM), the lifetime of dye fluorescence decay was measured to probe the polymer chain dynamics of PMEO2MA in CNC nanocomposite films. The PMEO2MA chains experienced distinct regions of differing dynamics within Bouligand structures. A correlation was observed between the average fluorescence lifetime and the mechanical performance of CNC films, indicating that polymer chains with high mobility improved the strain and toughness. These studies demonstrated FLIM as a method to investigate polymer dynamics at the nanosecond timescale that can readily be applied to other composite systems.
Collapse
Affiliation(s)
- Huyen Vu
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Jeremiah W Woodcock
- Material Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899-3460, United States
| | - Ajay Krishnamurthy
- Material Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899-3460, United States
| | - Jan Obrzut
- Material Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899-3460, United States
| | - Jeffrey W Gilman
- Material Science and Engineering Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899-3460, United States
| | - E Bryan Coughlin
- Department of Polymer Science and Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
21
|
Liu X, Wang Y, Effah CY, Wu L, Yu F, Wei J, Mao G, Xiong Y, He L. Endocytosis and intracellular RNAs imaging of nanomaterials-based fluorescence probes. Talanta 2022; 243:123377. [DOI: 10.1016/j.talanta.2022.123377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
|
22
|
Bousmah Y, Valenta H, Bertolin G, Singh U, Nicolas V, Pasquier H, Tramier M, Merola F, Erard M. tdLanYFP, a Yellow, Bright, Photostable, and pH-Insensitive Fluorescent Protein for Live-Cell Imaging and Förster Resonance Energy Transfer-Based Sensing Strategies. ACS Sens 2021; 6:3940-3947. [PMID: 34676768 DOI: 10.1021/acssensors.1c00874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Yellow fluorescent proteins (YFPs) are widely used as optical reporters in Förster resonance energy transfer (FRET)-based biosensors. Although great improvements have been done, the sensitivity of the biosensors is still limited by the low photostability and the poor fluorescence performances of YFPs at acidic pH values. Here, we characterize the yellow fluorescent protein tdLanYFP, derived from the tetrameric protein from the cephalochordate Branchiostoma lanceolatum, LanYFP. With a quantum yield of 0.92 and an extinction coefficient of 133,000 mol-1·L·cm-1, it is, to our knowledge, the brightest dimeric fluorescent protein available. Contrasting with EYFP and its derivatives, tdLanYFP has a very high photostability in vitro and in live cells. As a consequence, tdLanYFP allows imaging of cellular structures with subdiffraction resolution using STED nanoscopy and is compatible with the use of spectromicroscopies in single-molecule regimes. Its very low pK1/2 of 3.9 makes tdLanYFP an excellent tag even at acidic pH values. Finally, we show that tdLanYFP is a valuable FRET partner either as a donor or acceptor in different biosensing modalities. Altogether, these assets make tdLanYFP a very attractive yellow fluorescent protein for long-term or single-molecule live-cell imaging including FRET experiments at acidic pH.
Collapse
Affiliation(s)
- Yasmina Bousmah
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Hana Valenta
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)−UMR 6290, 35000 Rennes, France
| | - Utkarsh Singh
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Valérie Nicolas
- Microscopy Facility (MIPSIT), Ingénierie et Plateformes au Service de l’Innovation Thérapeutique−IPSIT−UMS−US31−UMS3679 (IPSIT), Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Hélène Pasquier
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Marc Tramier
- Univ Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes)−UMR 6290, 35000 Rennes, France
| | - Fabienne Merola
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| | - Marie Erard
- Université Paris-Saclay, CNRS, Institut de Chimie Physique, 91405 Orsay, France
| |
Collapse
|
23
|
Carlon-Andres I, Malinauskas T, Padilla-Parra S. Structure dynamics of HIV-1 Env trimers on native virions engaged with living T cells. Commun Biol 2021; 4:1228. [PMID: 34707229 PMCID: PMC8551276 DOI: 10.1038/s42003-021-02658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/09/2021] [Indexed: 11/29/2022] Open
Abstract
The HIV-1 envelope glycoprotein (Env) mediates viral entry into the host cell. Although the highly dynamic nature of Env intramolecular conformations has been shown with single molecule spectroscopy in vitro, the bona fide Env intra- and intermolecular mechanics when engaged with live T cells remains unknown. We used two photon fast fluorescence lifetime imaging detection of single-molecule Förster Resonance Energy Transfer occurring between fluorescent labels on HIV-1 Env on native virions. Our observations reveal Env dynamics at two levels: transitions between different intramolecular conformations and intermolecular interactions between Env within the viral membrane. Furthermore, we show that three broad neutralizing anti-Env antibodies directed to different epitopes restrict Env intramolecular dynamics and interactions between adjacent Env molecules when engaged with living T cells. Importantly, our results show that Env-Env interactions depend on efficient virus maturation, and that is disrupted upon binding of Env to CD4 or by neutralizing antibodies. Thus, this study illuminates how different intramolecular conformations and distribution of Env molecules mediate HIV-1 Env-T cell interactions in real time and therefore might control immune evasion.
Collapse
Affiliation(s)
- Irene Carlon-Andres
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, United Kingdom.
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sergi Padilla-Parra
- Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London, United Kingdom.
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
24
|
Cevheroğlu O, Murat M, Mingu-Akmete S, Son ÇD. Ste2p Under the Microscope: the Investigation of Oligomeric States of a Yeast G Protein-Coupled Receptor. J Phys Chem B 2021; 125:9526-9536. [PMID: 34433281 DOI: 10.1021/acs.jpcb.1c05872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Oligomerization of G protein-coupled receptors (GPCRs) may play important roles in maturation, internalization, signaling, and pharmacology of these receptors. However, the nature and extent of their oligomerization is still under debate. In our study, Ste2p, a yeast mating pheromone GPCR, was tagged with enhanced green fluorescent protein (EGFP), mCherry, and with split florescent protein fragments at the receptor C-terminus. The Förster resonance energy transfer (FRET) technique was used to detect receptors' oligomerization by calculating the energy transfer from EGFP to mCherry. Stimulation of Ste2p oligomers with the receptor ligand did not result in any significant change on observed FRET values. The bimolecular fluorescence complementation (BiFC) assay was combined with FRET to further investigate the tetrameric complexes of Ste2p. Our results suggest that in its quiescent (nonligand-activated) state, Ste2p is found at least as a tetrameric complex on the plasma membrane. Intriguingly, receptor tetramers in their active form showed a significant increase in FRET. This study provides a direct in vivo visualization of Ste2p tetramers and the pheromone effect on the extent of the receptor oligomerization.
Collapse
Affiliation(s)
- Orkun Cevheroğlu
- Stem Cell Institute, Ankara University, Cankaya, 06520 Ankara, Turkey
| | - Merve Murat
- Stem Cell Institute, Ankara University, Cankaya, 06520 Ankara, Turkey.,Department of Biological Sciences, Middle East Technical University, Cankaya, 06800 Ankara, Turkey
| | - Sara Mingu-Akmete
- Stem Cell Institute, Ankara University, Cankaya, 06520 Ankara, Turkey.,Department of Biological Sciences, Middle East Technical University, Cankaya, 06800 Ankara, Turkey
| | - Çağdaş D Son
- Department of Biological Sciences, Middle East Technical University, Cankaya, 06800 Ankara, Turkey
| |
Collapse
|
25
|
Cellular context shapes cyclic nucleotide signaling in neurons through multiple levels of integration. J Neurosci Methods 2021; 362:109305. [PMID: 34343574 DOI: 10.1016/j.jneumeth.2021.109305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023]
Abstract
Intracellular signaling with cyclic nucleotides are ubiquitous signaling pathways, yet the dynamics of these signals profoundly differ in different cell types. Biosensor imaging experiments, by providing direct measurements in intact cellular environment, reveal which receptors are activated by neuromodulators and how the coincidence of different neuromodulators is integrated at various levels in the signaling cascade. Phosphodiesterases appear as one important determinant of cross-talk between different signaling pathways. Finally, analysis of signal dynamics reveal that striatal medium-sized spiny neuron obey a different logic than other brain regions such as cortex, probably in relation with the function of this brain region which efficiently detects transient dopamine.
Collapse
|
26
|
Torricella F, Bonucci A, Polykretis P, Cencetti F, Banci L. Rapid protein delivery to living cells for biomolecular investigation. Biochem Biophys Res Commun 2021; 570:82-88. [PMID: 34274850 DOI: 10.1016/j.bbrc.2021.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 12/24/2022]
Abstract
The lack of a simple, fast and efficient method for protein delivery is limiting the widespread application of in-cell experiments, which are crucial for understanding the cellular function. We present here an innovative strategy to deliver proteins into both prokaryotic and eukaryotic cells, exploiting thermal vesiculation. This method allows to internalize substantial amounts of proteins, with different molecular weight and conformation, without compromising the structural properties and cell viability. Characterizing proteins in a physiological environment is essential as the environment can dramatically affect the conformation and dynamics of biomolecules as shown by in-cell EPR spectra vs those acquired in buffer solution. Considering its versatility, this method opens the possibility to scientists to study proteins directly in living cells through a wide range of techniques.
Collapse
Affiliation(s)
- Francesco Torricella
- Magnetic Resonance Center - CERM, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Alessio Bonucci
- Aix Marseille, Univ, CNRS, BIP, Laboratoire de Bioénergétique et Ingégnerie des protéines, Marseille, France
| | - Panagis Polykretis
- Interuniversity Consortium for Magnetic Resonance of Metallo Proteins (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, v.le GB Morgagni 50, 50134, Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center - CERM, University of Florence, via Luigi Sacconi 6, 50019, Sesto Fiorentino, Florence, Italy; Interuniversity Consortium for Magnetic Resonance of Metallo Proteins (CIRMMP), via Luigi Sacconi 6, 50019, Sesto Fiorentino, Florence, Italy; Department of Chemistry, University of Florence, via della Lastruccia 3, 50019, Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
27
|
Bertran A, Henbest KB, De Zotti M, Gobbo M, Timmel CR, Di Valentin M, Bowen AM. Light-Induced Triplet-Triplet Electron Resonance Spectroscopy. J Phys Chem Lett 2021; 12:80-85. [PMID: 33306382 PMCID: PMC8016185 DOI: 10.1021/acs.jpclett.0c02884] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
We present a new technique, light-induced triplet-triplet electron resonance spectroscopy (LITTER), which measures the dipolar interaction between two photoexcited triplet states, enabling both the distance and angular distributions between the two triplet moieties to be determined on a nanometer scale. This is demonstrated for a model bis-porphyrin peptide that renders dipolar traces with strong orientation selection effects. Using simulations and density functional theory calculations, we extract distance distributions and relative orientations of the porphyrin moieties, allowing the dominant conformation of the peptide in a frozen solution to be identified. LITTER removes the requirement of current light-induced electron spin resonance pulse dipolar spectroscopy techniques to have a permanent paramagnetic moiety, becoming more suitable for in-cell applications and facilitating access to distance determination in unmodified macromolecular systems containing photoexcitable moieties. LITTER also has the potential to enable direct comparison with Förster resonance energy transfer and combination with microscopy inside cells.
Collapse
Affiliation(s)
- Arnau Bertran
- Centre
for Advanced Electron Spin Resonance and Inorganic Chemistry Laboratory,
Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Kevin B. Henbest
- Centre
for Advanced Electron Spin Resonance and Inorganic Chemistry Laboratory,
Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Marta De Zotti
- Department
of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Marina Gobbo
- Department
of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Christiane R. Timmel
- Centre
for Advanced Electron Spin Resonance and Inorganic Chemistry Laboratory,
Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
| | - Marilena Di Valentin
- Department
of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Alice M. Bowen
- Centre
for Advanced Electron Spin Resonance and Inorganic Chemistry Laboratory,
Department of Chemistry, University of Oxford, South Parks Road, Oxford OX1 3QR, United Kingdom
- Department
of Chemistry and Photon Science Institute, University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| |
Collapse
|
28
|
Feruglio PF, Vinegoni C, Weissleder R. Extended dynamic range imaging for noise mitigation in fluorescence anisotropy imaging. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200159R. [PMID: 32820624 PMCID: PMC7439791 DOI: 10.1117/1.jbo.25.8.086003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 06/11/2023]
Abstract
SIGNIFICANCE Fluorescence polarization (FP) and fluorescence anisotropy (FA) microscopy are powerful imaging techniques that allow to translate the common FP assay capabilities into the in vitro and in vivo cellular domain. As a result, they have found potential for mapping drug-protein or protein-protein interactions. Unfortunately, these imaging modalities are ratiometric in nature and as such they suffer from excessive noise even under regular imaging conditions, preventing accurate image-feature analysis of fluorescent molecules behaviors. AIM We present a high dynamic range (HDR)-based FA imaging modality for improving image quality in FA microscopy. APPROACH The method exploits ad hoc acquisition schemes to extend the dynamic range of individual FP channels, allowing to obtain FA images with increased signal-to-noise ratio. RESULTS A direct comparison between FA images obtained with our method and the standard, clearly indicates how an HDR-based FA imaging approach allows to obtain high-quality images, with the ability to correctly resolve image features at different values of FA and over a substantially higher range of fluorescence intensities. CONCLUSION The method presented is shown to outperform standard FA imaging microscopy narrowing the spread of the propagated error and yielding higher quality images. The method can be effectively and routinely used on any commercial imaging system and could be also translated to other microscopy ratiometric imaging modalities.
Collapse
Affiliation(s)
- Paolo Fumene Feruglio
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
- University of Verona, Department of Neuroscience, Biomedicine, and Movement Sciences, Verona, Italy
- ITS Meccatronico Veneto, Vicenza, Italy
| | - Claudio Vinegoni
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
| | - Ralph Weissleder
- Massachusetts General Hospital, Harvard Medical School, Center for Systems Biology, Boston, Massachusetts, United States
- Harvard Medical School, Department of Systems Biology, Boston, Massachusetts, United States
| |
Collapse
|
29
|
Rozbesky D, Verhagen MG, Karia D, Nagy GN, Alvarez L, Robinson RA, Harlos K, Padilla‐Parra S, Pasterkamp RJ, Jones EY. Structural basis of semaphorin-plexin cis interaction. EMBO J 2020; 39:e102926. [PMID: 32500924 PMCID: PMC7327498 DOI: 10.15252/embj.2019102926] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 01/05/2023] Open
Abstract
Semaphorin ligands interact with plexin receptors to contribute to functions in the development of myriad tissues including neurite guidance and synaptic organisation within the nervous system. Cell-attached semaphorins interact in trans with plexins on opposing cells, but also in cis on the same cell. The interplay between trans and cis interactions is crucial for the regulated development of complex neural circuitry, but the underlying molecular mechanisms are uncharacterised. We have discovered a distinct mode of interaction through which the Drosophila semaphorin Sema1b and mouse Sema6A mediate binding in cis to their cognate plexin receptors. Our high-resolution structural, biophysical and in vitro analyses demonstrate that monomeric semaphorins can mediate a distinctive plexin binding mode. These findings suggest the interplay between monomeric vs dimeric states has a hereto unappreciated role in semaphorin biology, providing a mechanism by which Sema6s may balance cis and trans functionalities.
Collapse
Affiliation(s)
- Daniel Rozbesky
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Marieke G Verhagen
- Department of Translational NeuroscienceUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Dimple Karia
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Gergely N Nagy
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Luis Alvarez
- Cellular ImagingWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Ross A Robinson
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Present address:
Immunocore LtdAbingdonUK
| | - Karl Harlos
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Sergi Padilla‐Parra
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Cellular ImagingWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
- Present address:
Department of Infectious DiseasesFaculty of Life Sciences & MedicineKing's College LondonLondonUK
- Present address:
Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - R Jeroen Pasterkamp
- Department of Translational NeuroscienceUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Edith Yvonne Jones
- Division of Structural BiologyWellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| |
Collapse
|
30
|
Rozbesky D, Monistrol J, Jain V, Hillier J, Padilla-Parra S, Jones EY. Drosophila OTK Is a Glycosaminoglycan-Binding Protein with High Conformational Flexibility. Structure 2020; 28:507-515.e5. [PMID: 32187531 PMCID: PMC7203548 DOI: 10.1016/j.str.2020.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/29/2020] [Accepted: 02/27/2020] [Indexed: 11/25/2022]
Abstract
The transmembrane protein OTK plays an essential role in plexin and Wnt signaling during Drosophila development. We have determined a crystal structure of the last three domains of the OTK ectodomain and found that OTK shows high conformational flexibility resulting from mobility at the interdomain interfaces. We failed to detect direct binding between Drosophila Plexin A (PlexA) and OTK, which was suggested previously. We found that, instead of PlexA, OTK directly binds semaphorin 1a. Our binding analyses further revealed that glycosaminoglycans, heparin and heparan sulfate, are ligands for OTK and thus may play a role in the Sema1a-PlexA axon guidance system.
Collapse
Affiliation(s)
- Daniel Rozbesky
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| | - Jim Monistrol
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Vitul Jain
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - James Hillier
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sergi Padilla-Parra
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Cellular imaging, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Infectious Diseases, King's College London, Faculty of Life Sciences & Medicine, London SE1 9RT, UK; Randall Centre for Cell and Molecular Biology, King's College London, London SE1 1UL, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.
| |
Collapse
|
31
|
Jonkman J, Brown CM, Wright GD, Anderson KI, North AJ. Tutorial: guidance for quantitative confocal microscopy. Nat Protoc 2020. [PMID: 32235926 DOI: 10.1038/s41596-020-0313-319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
When used appropriately, a confocal fluorescence microscope is an excellent tool for making quantitative measurements in cells and tissues. The confocal microscope's ability to block out-of-focus light and thereby perform optical sectioning through a specimen allows the researcher to quantify fluorescence with very high spatial precision. However, generating meaningful data using confocal microscopy requires careful planning and a thorough understanding of the technique. In this tutorial, the researcher is guided through all aspects of acquiring quantitative confocal microscopy images, including optimizing sample preparation for fixed and live cells, choosing the most suitable microscope for a given application and configuring the microscope parameters. Suggestions are offered for planning unbiased and rigorous confocal microscope experiments. Common pitfalls such as photobleaching and cross-talk are addressed, as well as several troubling instrumentation problems that may prevent the acquisition of quantitative data. Finally, guidelines for analyzing and presenting confocal images in a way that maintains the quantitative nature of the data are presented, and statistical analysis is discussed. A visual summary of this tutorial is available as a poster (https://doi.org/10.1038/s41596-020-0307-7).
Collapse
Affiliation(s)
- James Jonkman
- Advanced Optical Microscopy Facility (AOMF), University Health Network, Toronto, Ontario, Canada.
| | - Claire M Brown
- Advanced BioImaging Facility (ABIF), McGill University, Montreal, Quebec, Canada
| | - Graham D Wright
- A*STAR Microscopy Platform (AMP), Skin Research Institute of Singapore, A*STAR, Singapore, Singapore
| | - Kurt I Anderson
- Crick Advanced Light Microscopy Facility (CALM), The Francis Crick Institute, London, UK
| | - Alison J North
- Bio-Imaging Resource Center, The Rockefeller University, New York, NY, USA
| |
Collapse
|
32
|
Hepatitis B Virus Core Protein Domains Essential for Viral Capsid Assembly in a Cellular Context. J Mol Biol 2020; 432:3802-3819. [PMID: 32371046 DOI: 10.1016/j.jmb.2020.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/25/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Hepatitis B virus (HBV) core protein (HBc) is essential to the formation of the HBV capsid. HBc contains two domains: the N-terminal domain corresponding to residues 1-140 essential to form the icosahedral shell and the C-terminal domain corresponding to a basic and phosphorylated peptide, and required for DNA replication. The role of these two domains for HBV capsid assembly was essentially studied in vitro with HBc purified from mammalian or non-mammalian cell lysates, but their respective role in living cells remains to be clarified. We therefore investigated the assembly of the HBV capsid in Huh7 cells by combining fluorescence lifetime imaging microscopy/Förster's resonance energy transfer, fluorescence correlation spectroscopy and transmission electron microscopy approaches. We found that wild-type HBc forms oligomers early after transfection and at a sub-micromolar concentration. These oligomers are homogeneously diffused throughout the cell. We quantified a stoichiometry ranging from ~170 to ~230 HBc proteins per oligomer, consistent with the visualization of eGFP-containingHBV capsid shaped as native capsid particles by transmission electron microscopy. In contrast, no assembly was observed when HBc-N-terminal domain was expressed. This highlights the essential role of the C-terminal domain to form capsid in mammalian cells. Deletion of either the third helix or of the 124-135 residues of HBc had a dramatic impact on the assembly of the HBV capsid, inducing the formation of mis-assembled oligomers and monomers, respectively. This study shows that our approach using fluorescent derivatives of HBc is an innovative method to investigate HBV capsid formation.
Collapse
|
33
|
QuanTI-FRET: a framework for quantitative FRET measurements in living cells. Sci Rep 2020; 10:6504. [PMID: 32300110 PMCID: PMC7162988 DOI: 10.1038/s41598-020-62924-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/17/2020] [Indexed: 12/15/2022] Open
Abstract
Förster Resonance Energy Transfer (FRET) allows for the visualization of nanometer-scale distances and distance changes. This sensitivity is regularly achieved in single-molecule experiments in vitro but is still challenging in biological materials. Despite many efforts, quantitative FRET in living samples is either restricted to specific instruments or limited by the complexity of the required analysis. With the recent development and expanding utilization of FRET-based biosensors, it becomes essential to allow biologists to produce quantitative results that can directly be compared. Here, we present a new calibration and analysis method allowing for quantitative FRET imaging in living cells with a simple fluorescence microscope. Aside from the spectral crosstalk corrections, two additional correction factors were defined from photophysical equations, describing the relative differences in excitation and detection efficiencies. The calibration is achieved in a single step, which renders the Quantitative Three-Image FRET (QuanTI-FRET) method extremely robust. The only requirement is a sample of known stoichiometry donor:acceptor, which is naturally the case for intramolecular FRET constructs. We show that QuanTI-FRET gives absolute FRET values, independent of the instrument or the expression level. Through the calculation of the stoichiometry, we assess the quality of the data thus making QuanTI-FRET usable confidently by non-specialists.
Collapse
|
34
|
Jonkman J, Brown CM, Wright GD, Anderson KI, North AJ. Tutorial: guidance for quantitative confocal microscopy. Nat Protoc 2020; 15:1585-1611. [DOI: 10.1038/s41596-020-0313-9] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/10/2020] [Indexed: 01/04/2023]
|
35
|
Juliusson HY, Sigurdsson ST. Reduction Resistant and Rigid Nitroxide Spin-Labels for DNA and RNA. J Org Chem 2020; 85:4036-4046. [PMID: 32103670 DOI: 10.1021/acs.joc.9b02988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Electron paramagnetic resonance (EPR) spectroscopy, coupled with site-directed spin labeling (SDSL), is a useful method for studying conformational changes of biomolecules in cells. To employ in-cell EPR using nitroxide-based spin labels, the structure of the nitroxides must confer reduction resistance to withstand the reductive environment within cells. Here, we report the synthesis of two new spin labels, EÇ and EÇm, both of which possess the rigidity and the reduction resistance needed for extracting detailed structural information by EPR spectroscopy. EÇ and EÇm were incorporated into DNA and RNA, respectively, by oligonucleotide synthesis. Both labels were shown to be nonperturbing of the duplex structure. The partial reduction of EÇm during RNA synthesis was circumvented by the protection of the nitroxide as a benzoylated hydroxylamine.
Collapse
Affiliation(s)
- Haraldur Y Juliusson
- Department of Chemistry, Science Institute, University of Iceland, Dunhaga 3, 107 Reykjavik, Iceland
| | - Snorri Th Sigurdsson
- Department of Chemistry, Science Institute, University of Iceland, Dunhaga 3, 107 Reykjavik, Iceland
| |
Collapse
|
36
|
Sizaire F, Le Marchand G, Pécréaux J, Bouchareb O, Tramier M. Automated screening of AURKA activity based on a genetically encoded FRET biosensor using fluorescence lifetime imaging microscopy. Methods Appl Fluoresc 2020; 8:024006. [PMID: 32032967 DOI: 10.1088/2050-6120/ab73f5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fluorescence Lifetime Imaging Microscopy (FLIM) is a robust tool to measure Förster Resonance Energy Transfer (FRET) between two fluorescent proteins, mainly when using genetically-encoded FRET biosensors. It is then possible to monitor biological processes such as kinase activity with a good spatiotemporal resolution and accuracy. Therefore, it is of interest to improve this methodology for future high content screening purposes. We here implement a time-gated FLIM microscope that can image and quantify fluorescence lifetime with a higher speed than conventional techniques such as Time-Correlated Single Photon Counting (TCSPC). We then improve our system to perform automatic screen analysis in a 96-well plate format. Moreover, we use a FRET biosensor of AURKA activity, a mitotic kinase involved in several epithelial cancers. Our results show that our system is suitable to measure FRET within our biosensor paving the way to the screening of novel compounds, potentially allowing to find new inhibitors of AURKA activity.
Collapse
Affiliation(s)
- Florian Sizaire
- Univ Rennes, CNRS, IGDR (Genetics and Development Institute of Rennes), UMR 6290, F-35000 Rennes, France
| | | | | | | | | |
Collapse
|
37
|
Carlon-Andres I, Padilla-Parra S. Quantitative FRET-FLIM-BlaM to Assess the Extent of HIV-1 Fusion in Live Cells. Viruses 2020; 12:E206. [PMID: 32059513 PMCID: PMC7077196 DOI: 10.3390/v12020206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 11/16/2022] Open
Abstract
The first steps of human immunodeficiency virus (HIV) infection go through the engagement of HIV envelope (Env) with CD4 and coreceptors (CXCR4 or CCR5) to mediate viral membrane fusion between the virus and the host. New approaches are still needed to better define both the molecular mechanistic underpinnings of this process but also the point of fusion and its kinetics. Here, we have developed a new method able to detect and quantify HIV-1 fusion in single live cells. We present a new approach that employs fluorescence lifetime imaging microscopy (FLIM) to detect Förster resonance energy transfer (FRET) when using the β-lactamase (BlaM) assay. This novel approach allows comparing different populations of single cells regardless the concentration of CCF2-AM FRET reporter in each cell, and more importantly, is able to determine the relative amount of viruses internalized per cell. We have applied this approach in both reporter TZM-bl cells and primary T cell lymphocytes.
Collapse
Affiliation(s)
| | - Sergi Padilla-Parra
- Division of Structural Biology, University of Oxford, Wellcome Centre for Human Genetics, Headington, Oxford OX3 7BN, UK;
| |
Collapse
|
38
|
Gökerküçük EB, Tramier M, Bertolin G. Imaging Mitochondrial Functions: from Fluorescent Dyes to Genetically-Encoded Sensors. Genes (Basel) 2020; 11:E125. [PMID: 31979408 PMCID: PMC7073610 DOI: 10.3390/genes11020125] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria are multifunctional organelles that are crucial to cell homeostasis. They constitute the major site of energy production for the cell, they are key players in signalling pathways using secondary messengers such as calcium, and they are involved in cell death and redox balance paradigms. Mitochondria quickly adapt their dynamics and biogenesis rates to meet the varying energy demands of the cells, both in normal and in pathological conditions. Therefore, understanding simultaneous changes in mitochondrial functions is crucial in developing mitochondria-based therapy options for complex pathological conditions such as cancer, neurological disorders, and metabolic syndromes. To this end, fluorescence microscopy coupled to live imaging represents a promising strategy to track these changes in real time. In this review, we will first describe the commonly available tools to follow three key mitochondrial functions using fluorescence microscopy: Calcium signalling, mitochondrial dynamics, and mitophagy. Then, we will focus on how the development of genetically-encoded fluorescent sensors became a milestone for the understanding of these mitochondrial functions. In particular, we will show how these tools allowed researchers to address several biochemical activities in living cells, and with high spatiotemporal resolution. With the ultimate goal of tracking multiple mitochondrial functions simultaneously, we will conclude by presenting future perspectives for the development of novel genetically-encoded fluorescent biosensors.
Collapse
Affiliation(s)
| | | | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR [Institut de génétique et développement de Rennes] UMR 6290, F-35000 Rennes, France
| |
Collapse
|
39
|
A Software Tool for High-Throughput Real-Time Measurement of Intensity-Based Ratio-Metric FRET. Cells 2019; 8:cells8121541. [PMID: 31795419 PMCID: PMC6952787 DOI: 10.3390/cells8121541] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Förster resonance energy transfer (FRET) is increasingly used for non-invasive measurement of fluorescently tagged molecules in live cells. In this study, we have developed a freely available software tool MultiFRET, which, together with the use of a motorised microscope stage, allows multiple single cells to be studied in one experiment. MultiFRET is a Java plugin for Micro-Manager software, which provides real-time calculations of ratio-metric signals during acquisition and can simultaneously record from multiple cells in the same experiment. It can also make other custom-determined live calculations that can be easily exported to Excel at the end of the experiment. It is flexible and can work with multiple spectral acquisition channels. We validated this software by comparing the output of MultiFRET to that of a previously established and well-documented method for live ratio-metric FRET experiments and found no significant difference between the data produced with the use of the new MultiFRET and other methods. In this validation, we used several cAMP FRET sensors and cell models: i) isolated adult cardiomyocytes from transgenic mice expressing the cytosolic epac1-camps and targeted pmEpac1 and Epac1-PLN sensors, ii) isolated neonatal mouse cardiomyocytes transfected with the AKAP79-CUTie sensor, and iii) human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) transfected with the Epac-SH74 sensor. The MultiFRET plugin is an open source freely available package that can be used in a wide area of live cell imaging when live ratio-metric calculations are required.
Collapse
|
40
|
Skruzny M, Pohl E, Abella M. FRET Microscopy in Yeast. BIOSENSORS 2019; 9:E122. [PMID: 31614546 PMCID: PMC6956097 DOI: 10.3390/bios9040122] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Förster resonance energy transfer (FRET) microscopy is a powerful fluorescence microscopy method to study the nanoscale organization of multiprotein assemblies in vivo. Moreover, many biochemical and biophysical processes can be followed by employing sophisticated FRET biosensors directly in living cells. Here, we summarize existing FRET experiments and biosensors applied in yeasts Saccharomyces cerevisiae and Schizosaccharomyces pombe, two important models of fundamental biomedical research and efficient platforms for analyses of bioactive molecules. We aim to provide a practical guide on suitable FRET techniques, fluorescent proteins, and experimental setups available for successful FRET experiments in yeasts.
Collapse
Affiliation(s)
- Michal Skruzny
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany.
- LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043 Marburg, Germany.
| | - Emma Pohl
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany
- LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043 Marburg, Germany
| | - Marc Abella
- Department of Systems and Synthetic Microbiology, Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany
- LOEWE Center for Synthetic Microbiology (SYNMIKRO), 35043 Marburg, Germany
| |
Collapse
|
41
|
Qin Q, Laub S, Shi Y, Ouyang M, Peng Q, Zhang J, Wang Y, Lu S. Fluocell for Ratiometric and High-Throughput Live-Cell Image Visualization and Quantitation. FRONTIERS IN PHYSICS 2019; 7:154. [PMID: 33163483 PMCID: PMC7646842 DOI: 10.3389/fphy.2019.00154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Spatiotemporal regulation of molecular activities dictates cellular function and fate. Investigation of dynamic molecular activities in live cells often requires the visualization and quantitation of fluorescent ratio image sequences with subcellular resolution and in high throughput. Hence, there is a great need for convenient software tools specifically designed with these capabilities. Here we describe a well-characterized open-source software package, Fluocell, customized to visualize pixelwise ratiometric images and calculate ratio time courses with subcellular resolution and in high throughput. Fluocell also provides group statistics and kinetic analysis functions for the quantified time courses, as well as 3D structure and function visualization for ratio images. The application of Fluocell is demonstrated by the ratiometric analysis of intensity images for several single-chain Förster (or fluorescence) resonance energy transfer (FRET)-based biosensors, allowing efficient quantification of dynamic molecular activities in a heterogeneous population of single live cells. Our analysis revealed distinct activation kinetics of Fyn kinase in the cytosolic and membrane compartments, and visualized a 4D spatiotemporal distribution of epigenetic signals in mitotic cells. Therefore, Fluocell provides an integrated environment for ratiometric live-cell image visualization and analysis, which generates high-quality single-cell dynamic data and allows the quantitative machine-learning of biophysical and biochemical computational models for molecular regulations in cells and tissues.
Collapse
Affiliation(s)
- Qin Qin
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Shannon Laub
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Yiwen Shi
- Department of Mathematics, Center of Computational Mathematics, University of California, San Diego, San Diego, CA, United State
| | - Mingxing Ouyang
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou, China
| | - Qin Peng
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
| | - Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, CA, United States
- Department of Mathematics, Center of Computational Mathematics, University of California, San Diego, San Diego, CA, United State
| |
Collapse
|
42
|
Doll F, Keckeis P, Scheel P, Cölfen H. Visualizing Cholesterol Uptake by Self-Assembling Rhodamine B-Labeled Polymer Inside Living Cells via FLIM-FRET Microscopy. Macromol Biosci 2019; 20:e1900081. [PMID: 31222918 DOI: 10.1002/mabi.201900081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/04/2019] [Indexed: 01/25/2023]
Abstract
Atherosclerosis is a widespread and hazardous disease characterized by the formation of arterial plaques mostly composed of fat, cholesterol, and calcium ions. The direct solubilization of cholesterol represents a promising, atheroprotective strategy to subside lipid blood levels and reverse atherosclerosis. This study deals with the in-depth analysis of polymer-mediated cholesterol dissolution inside living human cells. To this end, a recently described multifunctional block-polymer is labeled with Rhodamine B (RhoB) to investigate its interaction with cells via fluorescence microscopy. This gives insight into the cellular internalization process of the polymer, which appears to be clathrin- and caveolae/raft-dependent endocytosis. In cell single particle tracking reveals an active transport of RhoB polymer including structures. Förster resonance energy transfer (FRET) measurements of cells treated with a fluorophore-tagged cholesterol derivative and the RhoB polymer indicates the uptake of cholesterol by the polymeric particles. Hence, these results present a first step toward possible applications of cholesterol-absorbing polymers for treating atherosclerosis.
Collapse
Affiliation(s)
- Franziska Doll
- Physical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Philipp Keckeis
- Physical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Patricia Scheel
- Physical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| | - Helmut Cölfen
- Physical Chemistry, Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78457, Konstanz, Germany
| |
Collapse
|
43
|
Liu X, Zhan S, Hu X, Zhang X. A ratio-metric fluorescent sensor design platform with red emission of Europium(III) as built-in fluorescence correction. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Lin T, Scott BL, Hoppe AD, Chakravarty S. FRETting about the affinity of bimolecular protein-protein interactions. Protein Sci 2019; 27:1850-1856. [PMID: 30052312 DOI: 10.1002/pro.3482] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 01/19/2023]
Abstract
Fluorescence resonance energy transfer (FRET) is a powerful tool to study macromolecular interactions such as protein-protein interactions (PPIs). Fluorescent protein (FP) fusions enable FRET-based PPI analysis of signaling pathways and molecular structure in living cells. Despite FRET's importance in PPI studies, FRET has seen limited use in quantifying the affinities of PPIs in living cells. Here, we have explored the relationship between FRET efficiency and PPI affinity over a wide range when expressed from a single plasmid system in Escherichia coli. Using live-cell microscopy and a set of 20 pairs of small interacting proteins, belonging to different structural folds and interaction affinities, we demonstrate that FRET efficiency can reliably measure the dissociation constant (KD ) over a range of mM to nM. A 10-fold increase in the interaction affinity results in 0.05 unit increase in FRET efficiency, providing sufficient resolution to quantify large affinity differences (> 10-fold) using live-cell FRET. This approach provides a rapid and simple strategy for assessment of PPI affinities over a wide range and will have utility for high-throughput analysis of protein interactions.
Collapse
Affiliation(s)
- Tao Lin
- Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota, 57007
| | - Brandon L Scott
- Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota, 57007
| | - Adam D Hoppe
- Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota, 57007.,BioSNTR, Brookings, South Dakota, 57007
| | - Suvobrata Chakravarty
- Chemistry and Biochemistry, South Dakota State University, Brookings, South Dakota, 57007.,BioSNTR, Brookings, South Dakota, 57007
| |
Collapse
|
45
|
Cost A, Khalaji S, Grashoff C. Genetically Encoded FRET‐Based Tension Sensors. ACTA ACUST UNITED AC 2019; 83:e85. [DOI: 10.1002/cpcb.85] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anna‐Lena Cost
- Group of Molecular Mechanotransduction, Max Planck Institute of Biochemistry Martinsried Germany
- Department of Quantitative Cell Biology, Institute of Molecular Cell BiologyUniversity of Münster Münster Germany
| | - Samira Khalaji
- Group of Molecular Mechanotransduction, Max Planck Institute of Biochemistry Martinsried Germany
- Department of Quantitative Cell Biology, Institute of Molecular Cell BiologyUniversity of Münster Münster Germany
| | - Carsten Grashoff
- Group of Molecular Mechanotransduction, Max Planck Institute of Biochemistry Martinsried Germany
- Department of Quantitative Cell Biology, Institute of Molecular Cell BiologyUniversity of Münster Münster Germany
| |
Collapse
|
46
|
Chapnick DA, Bunker E, Liu X, Old WM. Temporal Metabolite, Ion, and Enzyme Activity Profiling Using Fluorescence Microscopy and Genetically Encoded Biosensors. Methods Mol Biol 2019; 1978:343-353. [PMID: 31119673 DOI: 10.1007/978-1-4939-9236-2_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Living cells employ complex and highly dynamic signaling networks and transcriptional circuits to maintain homeostasis and respond appropriately to constantly changing environments. These networks enable cells to maintain tight control on intracellular concentrations of ions, metabolites, proteins, and other biomolecules and ensure a careful balance between a cell's energetic needs and catabolic processes required for growth. Establishing molecular mechanisms of genetic and pharmacological perturbations remains challenging, due to the interconnected nature of these networks and the extreme sensitivity of cellular systems to their external environment. Live cell imaging with genetically encoded fluorescent biosensors provides a powerful new modality for nondestructive spatiotemporal tracking of ions, small molecules, enzymatic activities, and molecular interactions in living systems, from cells, tissues, and even living organisms. By deploying large panels of cell lines, each with distinct biosensors, many critical biochemical pathways can be monitored in a highly parallel and high-throughput fashion to identify pharmacological vulnerabilities and combination therapies unique to a given cell type or genetic background. Here we describe the experimental and analytical methods required to conduct multiplexed parallel fluorescence microscopy experiments on live cells expressing stable transgenic synthetic protein biosensors.
Collapse
Affiliation(s)
| | - Eric Bunker
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
| | - Xuedong Liu
- Department of Biochemistry, University of Colorado, Boulder, CO, USA
| | - William M Old
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
47
|
Snell NE, Rao VP, Seckinger KM, Liang J, Leser J, Mancini AE, Rizzo MA. Homotransfer of FRET Reporters for Live Cell Imaging. BIOSENSORS-BASEL 2018; 8:bios8040089. [PMID: 30314323 PMCID: PMC6316388 DOI: 10.3390/bios8040089] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/27/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023]
Abstract
Förster resonance energy transfer (FRET) between fluorophores of the same species was recognized in the early to mid-1900s, well before modern heterotransfer applications. Recently, homotransfer FRET principles have re-emerged in biosensors that incorporate genetically encoded fluorescent proteins. Homotransfer offers distinct advantages over the standard heterotransfer FRET method, some of which are related to the use of fluorescence polarization microscopy to quantify FRET between two fluorophores of identical color. These include enhanced signal-to-noise, greater compatibility with other optical sensors and modulators, and new design strategies based upon the clustering or dimerization of singly-labeled sensors. Here, we discuss the theoretical basis for measuring homotransfer using polarization microscopy, procedures for data collection and processing, and we review the existing genetically-encoded homotransfer biosensors.
Collapse
Affiliation(s)
- Nicole E Snell
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - Vishnu P Rao
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - Kendra M Seckinger
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - Junyi Liang
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - Jenna Leser
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - Allison E Mancini
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| | - M A Rizzo
- Department of Physiology, University of Maryland School of Medicine, 660 W Redwood St/HH525B, Baltimore, MD 21201, USA.
| |
Collapse
|
48
|
Bertolin G, Bulteau AL, Alves-Guerra MC, Burel A, Lavault MT, Gavard O, Le Bras S, Gagné JP, Poirier GG, Le Borgne R, Prigent C, Tramier M. Aurora kinase A localises to mitochondria to control organelle dynamics and energy production. eLife 2018; 7:38111. [PMID: 30070631 PMCID: PMC6140714 DOI: 10.7554/elife.38111] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022] Open
Abstract
Many epithelial cancers show cell cycle dysfunction tightly correlated with the overexpression of the serine/threonine kinase Aurora A (AURKA). Its role in mitotic progression has been extensively characterised, and evidence for new AURKA functions emerges. Here, we reveal that AURKA is located and imported in mitochondria in several human cancer cell lines. Mitochondrial AURKA impacts on two organelle functions: mitochondrial dynamics and energy production. When AURKA is expressed at endogenous levels during interphase, it induces mitochondrial fragmentation independently from RALA. Conversely, AURKA enhances mitochondrial fusion and ATP production when it is over-expressed. We demonstrate that AURKA directly regulates mitochondrial functions and that AURKA over-expression promotes metabolic reprogramming by increasing mitochondrial interconnectivity. Our work paves the way to anti-cancer therapeutics based on the simultaneous targeting of mitochondrial functions and AURKA inhibition. Structures called mitochondria power cells by turning oxygen and sugar into chemical energy. Each cell can have thousands of mitochondria, which work together to supply changing energy demands. They can fuse together or break apart, forming networks that change size and produce different amounts of energy. Getting the balance right is crucial; if energy levels are too low, the cell will not be able to grow and divide. If energy levels are too high, the cell can grow at a faster rate, which can contribute to the cell becoming cancerous. Although we know that mitochondria provide energy, it is not clear how they communicate to fine-tune the supply. Some clues come from cancer cells that seem dependent on their mitochondria for survival. In these cells, levels of a protein called AURKA are higher than normal. AURKA helps cells to divide, and it interacts with many different proteins. This complexity makes it difficult to work out exactly what AURKA does, but it is possible that it plays a role in energy supply. Bertolin et al. have now investigated whether mitochondria use AURKA to communicate inside human breast cancer cells. Tagging AURKA proteins with a fluorescent marker revealed that it accumulates inside mitochondria. Once it gets there, AURKA changes the shape of the mitochondria, which has dramatic effects on their capacity to produce energy. At normal levels, AURKA causes the mitochondria to fragment, breaking apart into smaller pieces. This maintains their energy output at a normal level. If AURKA levels are too high, the mitochondria fuse together and produce more energy. This means AURKA could help to fuel fast-growing cancer cells. Current drugs that aim to treat cancer by blocking the activity of AURKA show poor results. This is partly due to the fact that the protein has so many different roles in the cell. Finding that AURKA affects mitochondria is the first step in understanding one of its unknown roles. It also suggests the possibility of developing new drugs to change how mitochondria make energy in cancer cells that contain high levels of AURKA.
Collapse
Affiliation(s)
- Giulia Bertolin
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France
| | - Anne-Laure Bulteau
- ENS de Lyon, Lyon, France.,CNRS UMR 5242, Lyon, France.,INRA USC 1370, Lyon, France
| | - Marie-Clotilde Alves-Guerra
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Agnes Burel
- Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| | - Marie-Thérèse Lavault
- Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| | - Olivia Gavard
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France.,Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Stephanie Le Bras
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France
| | - Jean-Philippe Gagné
- Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Guy G Poirier
- Centre de recherche du CHU de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | - Roland Le Borgne
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France
| | - Claude Prigent
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Equipes labélisées Ligue Contre Le Cancer, Rennes, France
| | - Marc Tramier
- CNRS, UMR 6290, Rennes, France.,Université de Rennes 1, UBL, Genetics and Development Institute of Rennes (IGDR), Rennes, France.,Microscopy Rennes Imaging Centre, SFR Biosit, UMS CNRS 3480- US INSERM 018, Université de Rennes, Rennes, France
| |
Collapse
|
49
|
Markwardt ML, Snell NE, Guo M, Wu Y, Christensen R, Liu H, Shroff H, Rizzo MA. A Genetically Encoded Biosensor Strategy for Quantifying Non-muscle Myosin II Phosphorylation Dynamics in Living Cells and Organisms. Cell Rep 2018; 24:1060-1070.e4. [PMID: 30044973 PMCID: PMC6117825 DOI: 10.1016/j.celrep.2018.06.088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 05/25/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023] Open
Abstract
Complex cell behaviors require dynamic control over non-muscle myosin II (NMMII) regulatory light chain (RLC) phosphorylation. Here, we report that RLC phosphorylation can be tracked in living cells and organisms using a homotransfer fluorescence resonance energy transfer (FRET) approach. Fluorescent protein-tagged RLCs exhibit FRET in the dephosphorylated conformation, permitting identification and quantification of RLC phosphorylation in living cells. This approach is versatile and can accommodate several different fluorescent protein colors, thus enabling multiplexed imaging with complementary biosensors. In fibroblasts, dynamic myosin phosphorylation was observed at the leading edge of migrating cells and retracting structures where it persistently colocalized with activated myosin light chain kinase. Changes in myosin phosphorylation during C. elegans embryonic development were tracked using polarization inverted selective-plane illumination microscopy (piSPIM), revealing a shift in phosphorylated myosin localization to a longitudinal orientation following the onset of twitching. Quantitative analyses further suggested that RLC phosphorylation dynamics occur independently from changes in protein expression.
Collapse
Affiliation(s)
- Michele L Markwardt
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicole E Snell
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Min Guo
- Section on High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, US NIH, Bethesda, MD 20814, USA
| | - Yicong Wu
- Section on High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, US NIH, Bethesda, MD 20814, USA
| | - Ryan Christensen
- Section on High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, US NIH, Bethesda, MD 20814, USA
| | - Huafeng Liu
- State Key Laboratory of Modern Optical Instrumentation, College of Optical Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Hari Shroff
- Section on High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, US NIH, Bethesda, MD 20814, USA
| | - M A Rizzo
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
50
|
Hoischen C, Monajembashi S, Weisshart K, Hemmerich P. Multimodal Light Microscopy Approaches to Reveal Structural and Functional Properties of Promyelocytic Leukemia Nuclear Bodies. Front Oncol 2018; 8:125. [PMID: 29888200 PMCID: PMC5980967 DOI: 10.3389/fonc.2018.00125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/05/2018] [Indexed: 12/11/2022] Open
Abstract
The promyelocytic leukemia (pml) gene product PML is a tumor suppressor localized mainly in the nucleus of mammalian cells. In the cell nucleus, PML seeds the formation of macromolecular multiprotein complexes, known as PML nuclear bodies (PML NBs). While PML NBs have been implicated in many cellular functions including cell cycle regulation, survival and apoptosis their role as signaling hubs along major genome maintenance pathways emerged more clearly. However, despite extensive research over the past decades, the precise biochemical function of PML in these pathways is still elusive. It remains a big challenge to unify all the different previously suggested cellular functions of PML NBs into one mechanistic model. With the advent of genetically encoded fluorescent proteins it became possible to trace protein function in living specimens. In parallel, a variety of fluorescence fluctuation microscopy (FFM) approaches have been developed which allow precise determination of the biophysical and interaction properties of cellular factors at the single molecule level in living cells. In this report, we summarize the current knowledge on PML nuclear bodies and describe several fluorescence imaging, manipulation, FFM, and super-resolution techniques suitable to analyze PML body assembly and function. These include fluorescence redistribution after photobleaching, fluorescence resonance energy transfer, fluorescence correlation spectroscopy, raster image correlation spectroscopy, ultraviolet laser microbeam-induced DNA damage, erythrocyte-mediated force application, and super-resolution microscopy approaches. Since most if not all of the microscopic equipment to perform these techniques may be available in an institutional or nearby facility, we hope to encourage more researches to exploit sophisticated imaging tools for their research in cancer biology.
Collapse
|