1
|
Kelly B, Boudreau JE, Beyea S, Brewer K. Molecular imaging of viral pathogenesis and opportunities for the future. NPJ IMAGING 2025; 3:3. [PMID: 39872292 PMCID: PMC11761071 DOI: 10.1038/s44303-024-00056-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/24/2024] [Indexed: 01/30/2025]
Abstract
Molecular imaging is used in clinical and research settings. Since tools to study viral pathogenesis longitudinally and systemically are limited, molecular imaging is an attractive and largely unexplored tool. This review discusses molecular imaging probes and techniques for studying viruses, particularly those currently used in oncology that are applicable to virology. Expanding the repertoire of probes to better detect viral disease may make imaging even more valuable in (pre-)clinical settings.
Collapse
Affiliation(s)
- Brianna Kelly
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
| | - Jeanette E. Boudreau
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- Department of Pathology, Dalhousie University, Halifax, NS Canada
- Beatrice Hunter Cancer Research Institute (BHCRI), Halifax, NS Canada
| | - Steven Beyea
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| | - Kimberly Brewer
- Biomedical MRI Research Laboratory (BMRL), IWK Health Centre, Halifax, NS Canada
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS Canada
- IWK Health Centre, Halifax, NS Canada
- Department of Diagnostic Radiology, Dalhousie University, Halifax, NS Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS Canada
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| |
Collapse
|
2
|
Townsend KM, Prescher JA. Recent advances in bioluminescent probes for neurobiology. NEUROPHOTONICS 2024; 11:024204. [PMID: 38390217 PMCID: PMC10883388 DOI: 10.1117/1.nph.11.2.024204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
Bioluminescence is a popular modality for imaging in living organisms. The platform relies on enzymatically (luciferase) generated light via the oxidation of small molecule luciferins. Since no external light is needed for photon production, there are no concerns with background autofluorescence or photobleaching over time-features that have historically limited other optical readouts. Bioluminescence is thus routinely used for longitudinal tracking across whole animals. Applications in the brain, though, have been more challenging due to a lack of sufficiently bioavailable, bright, and easily multiplexed probes. Recent years have seen the development of designer luciferase and luciferin pairs that address these issues, providing more sensitive and real-time readouts of biochemical features relevant to neurobiology. This review highlights many of the advances in bioluminescent probe design, with a focus on the small molecule light emitter, the luciferin. Specific efforts to improve luciferin pharmacokinetics and tissue-penetrant emission are covered, in addition to applications that such probes have enabled. The continued development of improved bioluminescent probes will aid in illuminating critical neurochemical processes in the brain.
Collapse
Affiliation(s)
- Katherine M Townsend
- University of California, Irvine, Department of Chemistry, Irvine, California, United States
| | - Jennifer A Prescher
- University of California, Irvine, Department of Chemistry, Irvine, California, United States
- University of California, Irvine, Department of Molecular Biology and Biochemistry, Irvine, California, United States
- University of California, Irvine, Department of Pharmaceutical Sciences, Irvine, California, United States
| |
Collapse
|
3
|
Shramova EI, Deyev SM, Proshkina GM. A Vector Nanoplatform for the Bioimaging of Deep-Seated Tumors. Acta Naturae 2024; 16:72-81. [PMID: 39188260 PMCID: PMC11345090 DOI: 10.32607/actanaturae.27425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 08/28/2024] Open
Abstract
Today, in preclinical studies, optical bioimaging based on luminescence and fluorescence is indispensable in studying the development of neoplastic transformations, the proliferative activity of the tumor, its metastatic potential, as well as the therapeutic effect of antitumor agents. In order to expand the capabilities of optical imaging, sensors based on the bioluminescence resonance energy transfer (BRET) mechanism and, therefore, independent of an external light source are being developed. A targeted nanoplatform based on HER2-specific liposomes whose internal environment contains a genetically encoded BRET sensor was developed in this study to visualize deep-seated tumors characterized by overexpression of human epidermal growth factor receptor type 2 (HER2). The BRET sensor is a hybrid protein consisting of the highly catalytic luciferase NanoLuc (an energy donor) and a LSSmKate1 red fluorescent protein with a large Stokes shift (an energy acceptor). During the bioimaging of disseminated intraperitoneal tumors formed by HER2-positive SKOV3.ip1cells of serous ovarian cystadenocarcinoma, it was shown that the developed system is applicable in detecting deep-seated tumors of a certain molecular profile. The developed system can become an efficient platform for optimizing preclinical studies of novel targeted drugs.
Collapse
Affiliation(s)
- E. I. Shramova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
| | - S. M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991 Russian Federation
- National Research Centre “Kurchatov Institute”, Moscow, 123098 Russian Federation
| | - G. M. Proshkina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russian Academy of science, Moscow, 117997 Russian Federation
| |
Collapse
|
4
|
Eom K, Jung J, Kim B, Hyun JH. Molecular tools for recording and intervention of neuronal activity. Mol Cells 2024; 47:100048. [PMID: 38521352 PMCID: PMC11021360 DOI: 10.1016/j.mocell.2024.100048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/12/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024] Open
Abstract
Observing the activity of neural networks is critical for the identification of learning and memory processes, as well as abnormal activities of neural circuits in disease, particularly for the purpose of tracking disease progression. Methodologies for describing the activity history of neural networks using molecular biology techniques first utilized genes expressed by active neurons, followed by the application of recently developed techniques including optogenetics and incorporation of insights garnered from other disciplines, including chemistry and physics. In this review, we will discuss ways in which molecular biological techniques used to describe the activity of neural networks have evolved along with the potential for future development.
Collapse
Affiliation(s)
- Kisang Eom
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jinhwan Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Byungsoo Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jung Ho Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea; Center for Synapse Diversity and Specificity, DGIST, Daegu 42988, Republic of Korea.
| |
Collapse
|
5
|
Yue O, Wang X, Xie L, Bai Z, Zou X, Liu X. Biomimetic Exogenous "Tissue Batteries" as Artificial Power Sources for Implantable Bioelectronic Devices Manufacturing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307369. [PMID: 38196276 PMCID: PMC10953594 DOI: 10.1002/advs.202307369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/27/2023] [Indexed: 01/11/2024]
Abstract
Implantable bioelectronic devices (IBDs) have gained attention for their capacity to conformably detect physiological and pathological signals and further provide internal therapy. However, traditional power sources integrated into these IBDs possess intricate limitations such as bulkiness, rigidity, and biotoxicity. Recently, artificial "tissue batteries" (ATBs) have diffusely developed as artificial power sources for IBDs manufacturing, enabling comprehensive biological-activity monitoring, diagnosis, and therapy. ATBs are on-demand and designed to accommodate the soft and confining curved placement space of organisms, minimizing interface discrepancies, and providing ample power for clinical applications. This review presents the near-term advancements in ATBs, with a focus on their miniaturization, flexibility, biodegradability, and power density. Furthermore, it delves into material-screening, structural-design, and energy density across three distinct categories of TBs, distinguished by power supply strategies. These types encompass innovative energy storage devices (chemical batteries and supercapacitors), power conversion devices that harness power from human-body (biofuel cells, thermoelectric nanogenerators, bio-potential devices, piezoelectric harvesters, and triboelectric devices), and energy transfer devices that receive and utilize external energy (radiofrequency-ultrasound energy harvesters, ultrasound-induced energy harvesters, and photovoltaic devices). Ultimately, future challenges and prospects emphasize ATBs with the indispensability of bio-safety, flexibility, and high-volume energy density as crucial components in long-term implantable bioelectronic devices.
Collapse
Affiliation(s)
- Ouyang Yue
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- National Demonstration Center for Experimental Light Chemistry Engineering EducationShaanxi University of Science &TechnologyXi'anShaanxi710021China
| | - Xuechuan Wang
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- College of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
| | - Long Xie
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- College of Chemistry and Chemical EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
| | - Zhongxue Bai
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- National Demonstration Center for Experimental Light Chemistry Engineering EducationShaanxi University of Science &TechnologyXi'anShaanxi710021China
| | - Xiaoliang Zou
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- National Demonstration Center for Experimental Light Chemistry Engineering EducationShaanxi University of Science &TechnologyXi'anShaanxi710021China
| | - Xinhua Liu
- College of Bioresources Chemical and Materials EngineeringShaanxi University of Science & TechnologyXi'anShaanxi710021China
- National Demonstration Center for Experimental Light Chemistry Engineering EducationShaanxi University of Science &TechnologyXi'anShaanxi710021China
| |
Collapse
|
6
|
Mujawar A, Dimri S, Palkina KA, Markina NM, Sarkisyan KS, Balakireva AV, Yampolsky IV, De A. Novel BRET combination for detection of rapamycin-induced protein dimerization using luciferase from fungus Neonothopanusnambi. Heliyon 2024; 10:e25553. [PMID: 38384550 PMCID: PMC10878866 DOI: 10.1016/j.heliyon.2024.e25553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/23/2024] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is one of the most promising approaches used for noninvasive imaging of protein-protein interactions in vivo. Recently, our team has discovered a genetically encodable bioluminescent system from the fungus Neonothopanus nambi and identified a novel luciferase that represents an imaging tool orthogonal to other luciferin-luciferase systems. We demonstrated the possibility of using the fungal luciferase as a new BRET donor by creating fused pairs with acceptor red fluorescent proteins, of which tdTomato provided the highest BRET efficiency. Using this new BRET system, we also designed a mTOR pathway specific rapamycin biosensor by integrating the FRB and FKBP12 protein dimerization system. We demonstrated the specificity and efficacy of the new fungal luciferase-based BRET combination for application in mammalian cell culture that will provide the unique opportunity to perform multiplexed BRET assessment in the future.
Collapse
Affiliation(s)
- Aaiyas Mujawar
- Advanced Center for Treatment Research and Education in Cancer (ACTREC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Life Science, Homi Bhabha National Institute, Mumbai, India
| | - Shalini Dimri
- Advanced Center for Treatment Research and Education in Cancer (ACTREC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Life Science, Homi Bhabha National Institute, Mumbai, India
| | - Ksenia A. Palkina
- Planta LLC, Bolshoi Boulevard, 42 Str 1, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, Russia
| | - Nadezhda M. Markina
- Planta LLC, Bolshoi Boulevard, 42 Str 1, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, Russia
| | - Karen S. Sarkisyan
- Planta LLC, Bolshoi Boulevard, 42 Str 1, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, Russia
- Synthetic Biology Group, MRC London Institute of Medical Sciences, London W12 0NN, UK
| | - Anastasia V. Balakireva
- Planta LLC, Bolshoi Boulevard, 42 Str 1, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, Russia
| | - Ilia V. Yampolsky
- Planta LLC, Bolshoi Boulevard, 42 Str 1, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya, 16/10, Moscow, Russia
| | - Abhijit De
- Advanced Center for Treatment Research and Education in Cancer (ACTREC), Sector-22, Kharghar, Navi Mumbai 410210, India
- Life Science, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
7
|
Navarro MX, Brennan CK, Love AC, Prescher JA. Caged luciferins enable rapid multicomponent bioluminescence imaging. Photochem Photobiol 2024; 100:67-74. [PMID: 37259257 PMCID: PMC10687313 DOI: 10.1111/php.13814] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 06/02/2023]
Abstract
Bioluminescence is a sensitive technique for imaging biological features over time. Historically, though, the modality has been challenging to employ for multiplexed tracking due to a lack of resolvable luciferase-luciferin pairs. Recent years have seen the development of numerous orthogonal probes for multi-parameter imaging. While successful, generating such tools often requires complex syntheses and lengthy enzyme evolution campaigns. This work showcases an alternative strategy for multiplexed bioluminescence that takes advantage of already-orthogonal caged luciferins and established uncaging enzymes. These probes generate unique bioluminescent signals that can be distinguished via a linear unmixing algorithm. Caged luciferins enabled two- and three-component imaging on the minutes time scale. We further showed that the tools can be used in conjunction with endogenous enzymes for multiplexed studies. Collectively, this approach lowers the barrier to multicomponent bioluminescence imaging and can be readily adopted by the broader community.
Collapse
Affiliation(s)
- Mariana X. Navarro
- Department of Chemistry, University of California, Irvine 1120 Natural Science II, Irvine, CA 92617 (USA)
| | - Caroline K. Brennan
- Department of Chemistry, University of California, Irvine 1120 Natural Science II, Irvine, CA 92617 (USA)
| | - Anna C. Love
- Department of Chemistry, University of California, Irvine 1120 Natural Science II, Irvine, CA 92617 (USA)
| | - Jennifer A. Prescher
- Department of Chemistry, University of California, Irvine 1120 Natural Science II, Irvine, CA 92617 (USA)
- Department of Molecular Biology and Biochemistry, University of California, Irvine, 3205 McGaugh Hall, Irvine, CA 92716 (USA)
- Department of Pharmaceutical Sciences, University of California, Irvine, 101 Theory, Suite 100, Irvine, CA 92617 (USA)
| |
Collapse
|
8
|
Calabretta MM, Michelini E. Current advances in the use of bioluminescence assays for drug discovery: an update of the last ten years. Expert Opin Drug Discov 2024; 19:85-95. [PMID: 37814480 DOI: 10.1080/17460441.2023.2266989] [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: 05/24/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION Bioluminescence is a well-established optical detection technique widely used in several bioanalytical applications, including high-throughput and high-content screenings. Thanks to advances in synthetic biology techniques and deep learning, a wide portfolio of luciferases is now available with tuned emission wavelengths, kinetics, and high stability. These luciferases can be implemented in the drug discovery and development pipeline, allowing high sensitivity and multiplexing capability. AREAS COVERED This review summarizes the latest advancements of bioluminescent systems as toolsets in drug discovery programs for in vitro applications. Particular attention is paid to the most advanced bioluminescence-based technologies for drug screening over the past 10 years (from 2013 to 2023) such as cell-free assays, cell-based assays based on genetically modified cells, bioluminescence resonance energy transfer, and protein complementation assays in 2D and 3D cell models. EXPERT OPINION The availability of tuned bioluminescent proteins with improved emission and stability properties is vital for the development of bioluminescence assays for drug discovery, spanning from reporter gene technology to protein-protein techniques. Further studies, combining machine learning with synthetic biology, will be necessary to obtain new tools for sustainable and highly predictive bioluminescent drug discovery platforms.
Collapse
Affiliation(s)
- Maria Maddalena Calabretta
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), IRCCS St. Orsola Hospital, Bologna, Italy
| | - Elisa Michelini
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), IRCCS St. Orsola Hospital, Bologna, Italy
- Health Sciences and Technologies Interdepartmental Center for Industrial Research (HSTICIR), University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Karasev MM, Verkhusha VV, Shcherbakova DM. Near-Infrared Optogenetic Module for Conditional Protein Splicing. J Mol Biol 2023; 435:168360. [PMID: 37949312 PMCID: PMC10842711 DOI: 10.1016/j.jmb.2023.168360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Optogenetics has emerged as a powerful tool for spatiotemporal control of biological processes. Near-infrared (NIR) light, with its low phototoxicity and deep tissue penetration, holds particular promise. However, the optogenetic control of polypeptide bond formation has not yet been developed. In this study, we introduce a NIR optogenetic module for conditional protein splicing (CPS) based on the gp41-1 intein. We optimized the module to minimize background signals in the darkness and to maximize the contrast between light and dark conditions. Next, we engineered a NIR CPS gene expression system based on the protein ligation of a transcription factor. We applied the NIR CPS for light-triggered protein cleavage to activate gasdermin D, a pore-forming protein that induces pyroptotic cell death. Our NIR CPS optogenetic module represents a promising tool for controlling molecular processes through covalent protein linkage and cleavage.
Collapse
Affiliation(s)
- Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland; Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
10
|
Beyond luciferase-luciferin system: Modification, improved imaging and biomedical application. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
11
|
Kamiya G, Kitada N, Maki S, Kim SB. Multiplex quadruple bioluminescent assay system. Sci Rep 2022; 12:17485. [PMID: 36261452 PMCID: PMC9581999 DOI: 10.1038/s41598-022-20468-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/13/2022] [Indexed: 01/12/2023] Open
Abstract
Bioluminescence (BL) is unique cold body radiation of light, generated by luciferin-luciferase reactions and commonly used in various bioassays and molecular imaging. However, most of the peak emissions of BL populate the blue-yellow region and have broad spectral bandwidths and thus superimpose each other, causing optical cross-leakages in multiplex assays. This study synthesized a new series of coelenterazine (CTZ) analogues, named K-series, that selectively illuminates marine luciferases with unique, blue-shifted spectral properties. The optical property and specificity of the K-series CTZ analogues were characterized by marine luciferases, with K2 and K5 found to specifically luminesce with ALuc- and RLuc-series marine luciferases, respectively. The results confirmed that the luciferase specificity and color variation of the CTZ analogues minimize the cross-leakages of BL signals and enable high-throughput screening of specific ligands in the mixture. The specificity and color variation of the substrates were further tailored to marine luciferases (or single-chain bioluminescent probes) to create a multiplex quadruple assay system with four integrated, single-chain bioluminescent probes, with each probe designed to selectively luminesce only with its specific ligand (first authentication) and a specific CTZ analogue (second authentication). This unique multiplex quadruple bioluminescent assay system is an efficient optical platform for specific and high-throughput imaging of multiple optical markers in bioassays without optical cross-leakages.
Collapse
Affiliation(s)
- Genta Kamiya
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Nobuo Kitada
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Shojiro Maki
- grid.266298.10000 0000 9271 9936Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Tokyo 182-8585 Japan
| | - Sung Bae Kim
- grid.208504.b0000 0001 2230 7538Research Institute for Environmental Management Technology, National Institute of Advanced Industrial Science and Technology (AIST), 16-1 Onogawa, Tsukuba, 305-8569 Japan
| |
Collapse
|
12
|
A Luciferase Mutant with Improved Brightness and Stability for Whole-Cell Bioluminescent Biosensors and In Vitro Biosensing. BIOSENSORS 2022; 12:bios12090742. [PMID: 36140127 PMCID: PMC9496056 DOI: 10.3390/bios12090742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022]
Abstract
The availability of new bioluminescent proteins with tuned properties, both in terms of emission wavelength, kinetics and protein stability, is highly valuable in the bioanalytical field, with the potential to improve the sensitivity and analytical performance of the currently used methods for ATP detection, whole-cell biosensors, and viability assays among others. We present a new luciferase mutant, called BgLuc, suitable for developing whole-cell biosensors and in vitro biosensors characterized by a bioluminescence maximum of 548 nm, narrow emission bandwidth, favorable kinetic properties, and excellent pH- and thermo-stabilities at 37 and 45 °C and pH from 5.0 to 8.0. We assessed the suitability of this new luciferase for whole-cell biosensing with a cell-based bioreporter assay for Nuclear Factor-kappa B (NF-kB) signal transduction pathway using 2D and 3D human embryonic kidney (HEK293T) cells, and for ATP detection with the purified enzyme. In both cases the luciferase showed suitable for sensitive detection of the target analytes, with better or similar performance than the commercial counterparts.
Collapse
|
13
|
Karasev MM, Baloban M, Verkhusha VV, Shcherbakova DM. Nuclear Localization Signals for Optimization of Genetically Encoded Tools in Neurons. Front Cell Dev Biol 2022; 10:931237. [PMID: 35927988 PMCID: PMC9344056 DOI: 10.3389/fcell.2022.931237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/24/2022] [Indexed: 12/15/2022] Open
Abstract
Nuclear transport in neurons differs from that in non-neuronal cells. Here we developed a non-opsin optogenetic tool (OT) for the nuclear export of a protein of interest induced by near-infrared (NIR) light. In darkness, nuclear import reverses the OT action. We used this tool for comparative analysis of nuclear transport dynamics mediated by nuclear localization signals (NLSs) with different importin specificities. We found that widely used KPNA2-binding NLSs, such as Myc and SV40, are suboptimal in neurons. We identified uncommon NLSs mediating fast nuclear import and demonstrated that the performance of the OT for nuclear export can be adjusted by varying NLSs. Using these NLSs, we optimized the NIR OT for light-controlled gene expression for lower background and higher contrast in neurons. The selected NLSs binding importins abundant in neurons could improve performance of genetically encoded tools in these cells, including OTs and gene-editing tools.
Collapse
Affiliation(s)
- Maksim M. Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikhail Baloban
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Vladislav V. Verkhusha
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Daria M. Shcherbakova
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
14
|
Liu F, Hu H, Deng M, Xiang Z, Guo Y, Guan X, Li D, Hu Q, Lei W, Peng H, Chu J. A Bright Monomeric Near-Infrared Fluorescent Protein with an Excitation Peak at 633 nm for Labeling Cellular Protein and Reporting Protein-Protein Interaction. ACS Sens 2022; 7:1855-1866. [PMID: 35775925 DOI: 10.1021/acssensors.2c00286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bright monomeric near-infrared fluorescent proteins (NIR-FPs) are useful as markers for labeling proteins and cells and as sensors for reporting molecular activities in living cells and organisms. However, current monomeric NIR-FPs are dim under excitation with common 633/635/640 nm lasers, limiting their broad use in cellular/subcellular level imaging. Here, we report a bright monomeric NIR-FP with maximum excitation at 633 nm, named mIFP663, engineered from Xanthomonas campestris pv Campestris phytochrome (XccBphP). mIFP663 has high molecular brightness with a large extinction coefficient (86,600 M-1 cm-1) and a decent quantum yield (19.4%), and high cellular brightness that is 3-6 times greater than those of spectrally similar NIR-FPs in HEK293T cells in the presence of exogenous BV. Moreover, we demonstrate that mIFP663 is able to label critical cellular and viral proteins without perturbing subcellular localization and virus replication, respectively. Finally, with mIFP663, we engineer improved bimolecular fluorescence complementation (BiFC) and new bioluminescent resonance energy transfer (BRET) systems to detect protein-protein interactions in living cells.
Collapse
Affiliation(s)
- Feng Liu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huimin Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengying Deng
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zongqin Xiang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, China
| | - Yuting Guo
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xinmeng Guan
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China.,Institute for Infection and Immunity, St George's, University of London, London SW17 0RE, United Kingdom
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510630, China
| | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging & CAS Key Laboratory of Health Informatics, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| |
Collapse
|
15
|
Barykina NV, Karasev MM, Verkhusha VV, Shcherbakova DM. Technologies for large-scale mapping of functional neural circuits active during a user-defined time window. Prog Neurobiol 2022; 216:102290. [PMID: 35654210 DOI: 10.1016/j.pneurobio.2022.102290] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/25/2022]
Abstract
The mapping of neural circuits activated during behavior down to individual neurons is crucial for decoding how the brain processes information. Technologies allowing activity-dependent labeling of neurons during user-defined restricted time windows are rapidly developing. Precise marking of the time window with light, in addition to chemicals, is now possible. In these technologies, genetically encoded molecules integrate molecular events resulting from neuronal activity with light/drug-dependent events. The outputs are either changes in fluorescence or activation of gene expression. Molecular reporters allow labeling of activated neurons for visualization and cell-type identification. The transcriptional readout also allows further control of activated neuronal populations using optogenetic tools as reporters. Here we review the design of these technologies and discuss their demonstrated applications to reveal previously unknown connections in the mammalian brain. We also consider the strengths and weaknesses of the current approaches and provide a perspective for the future.
Collapse
Affiliation(s)
- Natalia V Barykina
- P.K. Anokhin Institute of Normal Physiology, Moscow 125315, Russia; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
16
|
Zhao H, Zastrow ML. Transition Metals Induce Quenching of Monomeric Near-Infrared Fluorescent Proteins. Biochemistry 2022; 61:494-504. [PMID: 35289592 DOI: 10.1021/acs.biochem.1c00705] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transition metals such as zinc and copper are essential in numerous life processes, and both deficiency and toxic overload of these metals are associated with various diseases. Fluorescent metal sensors are powerful tools for studying the roles of metal ions in the physiology and pathology of biological systems. Green fluorescent protein (GFP) and its derivatives are highly utilized for protein-based sensor design, but application to anaerobic systems is limited because these proteins require oxygen to become fluorescent. Bacteriophytochrome-based monomeric near-infrared fluorescent proteins (miRFPs) covalently bind a bilin cofactor, which can be added exogenously for anaerobic cells. miRFPs can also have emission wavelengths extending to >700 nm, which is valuable for imaging applications. Here, we evaluated the suitability of miRFP670 and miRFP709 as platforms for single fluorescent protein metal ion sensors. We found that divalent metal ions like Zn2+, Co2+, Ni2+, and Cu2+ can quench from ∼6-20% (Zn2+, Co2+, and Ni2+) and up to nearly 90% (Cu2+) of the fluorescence intensity of pure miRFPs and have similar impacts in live Escherichia coli cells expressing miRFPs. The presence of a 6× histidine tag for purification influences metal quenching, but significant Cu2+-induced quenching and a picomolar binding affinity are retained in the absence of the His6 tag in both cuvettes and live bacterial cells. By comparing the Cu2+ and Cu+-induced quenching results for miRFP670 and miRFP709 and through examining absorption spectra and previously reported crystal structures, we propose a surface metal binding site near the biliverdin IXα chromophore.
Collapse
Affiliation(s)
- Haowen Zhao
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
17
|
Tsuboi S, Jin T. BRET-Based Dual-Color (Visible/Near-Infrared) Molecular Imaging Using a Quantum Dot/EGFP-Luciferase Conjugate. Methods Mol Biol 2022; 2525:47-59. [PMID: 35836060 DOI: 10.1007/978-1-0716-2473-9_5] [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: 06/15/2023]
Abstract
By virtue of its high sensitivity, bioluminescence imaging (BLI) is an important tool for biosensing and bioimaging in life sciences. Compared to fluorescence imaging (FLI), BLI has a superior advantage that the background signals resulting from autofluorescence are almost zero due to the unnecessity of external excitation. In addition, BLI can permit a long-term observation of living cells because BL results in very low photocytotoxicity toward the host cells. Although BLI has such superior properties over FLI, the available wavelengths in BLI are mostly limited to the visible region. Here we present bioluminescence resonance energy transfer (BRET)-based visible and near-infrared dual-color molecular imaging using a quantum dot (QD) and luciferase-protein conjugate.
Collapse
Affiliation(s)
- Setsuko Tsuboi
- RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Takashi Jin
- RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan.
| |
Collapse
|
18
|
Liu S, Su Y, Lin MZ, Ronald JA. Brightening up Biology: Advances in Luciferase Systems for in Vivo Imaging. ACS Chem Biol 2021; 16:2707-2718. [PMID: 34780699 PMCID: PMC8689642 DOI: 10.1021/acschembio.1c00549] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Bioluminescence imaging
(BLI) using luciferase reporters is an
indispensable method for the noninvasive visualization of cell populations
and biochemical events in living animals. BLI is widely performed
with preclinical rodent models to understand disease processes and
evaluate potential cell- or gene-based therapies. However, in vivo BLI remains constrained by low photon production
and tissue attenuation, limiting the sensitivity of reporting from
small numbers of cells in deep locations and hindering its application
to larger animal models. This Review highlights recent advances in
the development of luciferase systems that improve the sensitivity
of in vivo BLI and discusses the expanding array
of biological applications.
Collapse
Affiliation(s)
- Shirley Liu
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A3K7, Canada
- Department of Medical Biophysics, The University of Western Ontario, London, Ontario N6A3K7, Canada
| | - Yichi Su
- Department of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Michael Z. Lin
- Department of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - John A. Ronald
- Robarts Research Institute, The University of Western Ontario, London, Ontario N6A3K7, Canada
- Department of Medical Biophysics, The University of Western Ontario, London, Ontario N6A3K7, Canada
| |
Collapse
|
19
|
Yang SY, Sencadas V, You SS, Jia NZX, Srinivasan SS, Huang HW, Ahmed AE, Liang JY, Traverso G. Powering Implantable and Ingestible Electronics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2009289. [PMID: 34720792 PMCID: PMC8553224 DOI: 10.1002/adfm.202009289] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Indexed: 05/28/2023]
Abstract
Implantable and ingestible biomedical electronic devices can be useful tools for detecting physiological and pathophysiological signals, and providing treatments that cannot be done externally. However, one major challenge in the development of these devices is the limited lifetime of their power sources. The state-of-the-art of powering technologies for implantable and ingestible electronics is reviewed here. The structure and power requirements of implantable and ingestible biomedical electronics are described to guide the development of powering technologies. These powering technologies include novel batteries that can be used as both power sources and for energy storage, devices that can harvest energy from the human body, and devices that can receive and operate with energy transferred from exogenous sources. Furthermore, potential sources of mechanical, chemical, and electromagnetic energy present around common target locations of implantable and ingestible electronics are thoroughly analyzed; energy harvesting and transfer methods befitting each energy source are also discussed. Developing power sources that are safe, compact, and have high volumetric energy densities is essential for realizing long-term in-body biomedical electronics and for enabling a new era of personalized healthcare.
Collapse
Affiliation(s)
- So-Yoon Yang
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vitor Sencadas
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; School of Mechanical, Materials & Mechatronics Engineering, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Siheng Sean You
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Neil Zi-Xun Jia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shriya Sruthi Srinivasan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hen-Wei Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abdelsalam Elrefaey Ahmed
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jia Ying Liang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
Abstract
Near-infrared (NIR) luminescent materials have emerged as a growing field of interest, particularly for imaging and optics applications in biology, chemistry, and physics. However, the development of materials for this and other use cases has been hindered by a range of issues that prevents their widespread use beyond benchtop research. This review explores emerging trends in some of the most promising NIR materials and their applications. In particular, we focus on how a more comprehensive understanding of intrinsic NIR material properties might allow researchers to better leverage these traits for innovative and robust applications in biological and physical sciences.
Collapse
Affiliation(s)
- Christopher T. Jackson
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Sanghwa Jeong
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | | | - Markita P. Landry
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
- Innovative Genomics Institute (IGI), Berkeley, CA, USA
- California Institute for Quantitative Biosciences, QB3, University of California, Berkeley, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
21
|
Abstract
Bioluminescence imaging with luciferase-luciferin pairs is routinely used to monitor cellular functions. Multiple targets can be visualized in tandem using luciferases that process unique substrates, but only a handful of such orthogonal probes are known. Multiplexed studies require additional robust, light-emitting molecules. In this work, we report new luciferins for orthogonal imaging that comprise disubstituted cores. These probes were found to be bright emitters with various engineered luciferases. The unique patterns of light output also provided insight into enzyme-substrate interactions necessary for productive emission. Screening studies identified mutant luciferases that could preferentially process the disubstituted analogues, enabling orthogonal imaging with existing bioluminescent reporters. Further mutational analyses revealed the origins of substrate selectivity. Collectively, this work provides insights into luciferase-luciferin features relevant to bioluminescence and expands the number of probes for multicomponent tracking.
Collapse
Affiliation(s)
- Sierra J. Williams
- Department of Chemistry, University of California, Irvine; Irvine, California 92697, United States
| | - Clare S. Hwang
- Department of Public Health, University of California, Irvine; Irvine, California 92697, United States
| | - Jennifer A. Prescher
- Department of Chemistry, University of California, Irvine; Irvine, California 92697, United States
- Department of Molecular Biology & Biochemistry, University of California, Irvine; Irvine, California 92697, United States
- Department of Pharmaceutical Sciences, University of California, Irvine; Irvine, California 92697, United States
| |
Collapse
|
22
|
Gagnot G, Hervin V, Coutant EP, Goyard S, Jacob Y, Rose T, Hibti FE, Quatela A, Janin YL. Core-Modified Coelenterazine Luciferin Analogues: Synthesis and Chemiluminescence Properties. Chemistry 2021; 27:2112-2123. [PMID: 33137225 DOI: 10.1002/chem.202004311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/26/2020] [Indexed: 11/10/2022]
Abstract
In this work on the design and studies of luciferins related to the blue-hued coelenterazine, the synthesis of heterocyclic analogues susceptible to produce a photon, possibly at a different wavelength, is undertaken. Here, the synthesis of O-acetylated derivatives of imidazo[1,2-b]pyridazin-3(5 H)-one, imidazo[2,1-f][1,2,4]triazin-7(1 H)-one, imidazo[1,2-a]pyridin-3-ol, imidazo[1,2-a]quinoxalin-1(5 H)-one, benzo[f]imidazo[1,2-a]quinoxalin-3(11 H)-one, imidazo[1',2':1,6]pyrazino[2,3-c]quinolin-3(11 H)-one, and 5,11-dihydro-3 H-chromeno[4,3-e]imidazo[1,2-a]pyrazin-3-one is described thanks to extensive use of the Buchwald-Hartwig N-arylation reaction. The acidic hydrolysis of these derivatives then gave solutions of the corresponding luciferin analogues, which were studied. Not too unexpectedly, even if these were "dressed" with substituents found in actual substrates of the nanoKAZ/NanoLuc luciferase, no bioluminescence was observed with these compounds. However, in a phosphate buffer, all produced a light signal, by chemiluminescence, with extensive variations in their respective intensity and this could be increased by adding a quaternary ammonium salt in the buffer. This aspect was actually instrumental to determine the emission spectra of many of these luciferin analogues.
Collapse
Affiliation(s)
- Glwadys Gagnot
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France.,Université de Paris, 12 rue de l'école de Médecine, 75006, Paris, France
| | - Vincent Hervin
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Eloi P Coutant
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Sophie Goyard
- Center for Innovation and Technological Research, Institut Pasteur, 25 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Yves Jacob
- Unité de Génétique Moléculaire des Virus à ARN, Institut Pasteur, UMR 3569, CNRS, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Thierry Rose
- Center for Innovation and Technological Research, Institut Pasteur, 25 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Fatima Ezzahra Hibti
- HORIBA FRANCE SAS, 14 Boulevard Thomas Gobert, Passage Jobin Yvon CS45002, 91120, Palaiseau, France
| | - Alessia Quatela
- HORIBA FRANCE SAS, 14 Boulevard Thomas Gobert, Passage Jobin Yvon CS45002, 91120, Palaiseau, France
| | - Yves L Janin
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| |
Collapse
|
23
|
Nishihara R, Suzuki K, Kim SB, Paulmurugan R. Highly Bright NIR-BRET System for Imaging Molecular Events in Live Cells. Methods Mol Biol 2021; 2274:247-259. [PMID: 34050477 DOI: 10.1007/978-1-0716-1258-3_21] [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: 06/12/2023]
Abstract
The present protocol demonstrates a novel mammalian cell imaging platform exerting a bioluminescence resonance energy transfer (BRET) system. This platform achieves a ~300 nm blue-to-near infrared shift of the emission (NIR-BRET) with the development of a unique coelenterazine (CTZ) derivative named BBlue2.3 and a fusion reporter protein probe named iRFP-RLuc8.6-535SG. The best NIR-BRET shift was achieved by tuning the blue emission peak of BBlue2.3 to a Soret band of the iRFP. In mammalian cells, BBlue2.3 emits light that is ~50-fold brighter than DeepBlueC in cell imaging when combined with RLuc8.6-535SG. This NIR-BRET platform is sufficiently brighter to be used for imaging live mammalian cells at single-cell level, and also for imaging metastases in deep tissues in live mice without generating considerable autoluminescence. This unique optical platform provides the brightest NIR-BLI template that can be used for imaging a diverse group of cellular events in living subjects.
Collapse
Affiliation(s)
- Ryo Nishihara
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | - Koji Suzuki
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, Yokohama, Kanagawa, Japan
| | - Sung-Bae Kim
- Research Institute for Environmental Management Technology, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
24
|
Zhan Z, Dai Y, Li Q, Lv Y. Small molecule-based bioluminescence and chemiluminescence probes for sensing and imaging of reactive species. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2020.116129] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Zambito G, Hall MP, Wood MG, Gaspar N, Ridwan Y, Stellari FF, Shi C, Kirkland TA, Encell LP, Löwik C, Mezzanotte L. Red-shifted click beetle luciferase mutant expands the multicolor bioluminescent palette for deep tissue imaging. iScience 2020; 24:101986. [PMID: 33490896 PMCID: PMC7811125 DOI: 10.1016/j.isci.2020.101986] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 12/21/2022] Open
Abstract
For in vivo multicolor bioluminescence applications, red and near-infrared signals are desirable over shorter wavelength signals because they are not as susceptible to light attenuation by blood and tissue. Herein, we describe the development of a new click beetle luciferase mutant, CBG2, with a red-shifted color emission. When paired with NH2-NpLH2 luciferin, CBG2 (λ = 660 nm) and CBR2 (λ = 730 nm) luciferases can be used for simultaneous dual-color bioluminescence imaging in deep tissue. Using a spectral unmixing algorithm tool it is possible to distinguish each spectral contribution. Ultimately, this enzyme pair can expand the near-infrared bioluminescent toolbox to enable rapid visualization of multiple biological processes in deep tissue using a single substrate. CBG2 is a new click beetle mutant derived from CBG99 luciferase It has a far red-shifted bioluminescent emission with NH2-NpLH2 luciferin (λ = 660 nm) CBG2 with CBR2 luciferase and NH2-NpLH2 allows fast acquisition of data in deep tissue
Collapse
Affiliation(s)
- Giorgia Zambito
- Erasmus Medical Center, Radiology and Nuclear Medicine, Rotterdam, 3015 CE, the Netherlands.,Erasmus Medical Center, Molecular Genetics, Rotterdam, 3015 CE, the Netherlands.,Medres Medical Research GmBH, Cologne 50931, Germany
| | | | | | - Natasa Gaspar
- Erasmus Medical Center, Radiology and Nuclear Medicine, Rotterdam, 3015 CE, the Netherlands.,Erasmus Medical Center, Molecular Genetics, Rotterdam, 3015 CE, the Netherlands.,Percuros B.V., Leiden, 1333 CL, the Netherlands
| | - Yanto Ridwan
- Erasmus Medical Center, Radiology and Nuclear Medicine, Rotterdam, 3015 CE, the Netherlands.,Erasmus Medical Center, Molecular Genetics, Rotterdam, 3015 CE, the Netherlands
| | | | - Ce Shi
- Promega Biosciences Incorporated, San Luis Obispo, CA 93401, USA
| | | | | | - Clemens Löwik
- Erasmus Medical Center, Radiology and Nuclear Medicine, Rotterdam, 3015 CE, the Netherlands.,Erasmus Medical Center, Molecular Genetics, Rotterdam, 3015 CE, the Netherlands.,CHUV Department of Oncology, University of Lausanne, 461011 Lausanne, Switzerland
| | - Laura Mezzanotte
- Erasmus Medical Center, Radiology and Nuclear Medicine, Rotterdam, 3015 CE, the Netherlands.,Erasmus Medical Center, Molecular Genetics, Rotterdam, 3015 CE, the Netherlands
| |
Collapse
|
26
|
Endo M, Ozawa T. Advanced Bioluminescence System for In Vivo Imaging with Brighter and Red-Shifted Light Emission. Int J Mol Sci 2020; 21:E6538. [PMID: 32906768 PMCID: PMC7555964 DOI: 10.3390/ijms21186538] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 01/04/2023] Open
Abstract
In vivo bioluminescence imaging (BLI), which is based on luminescence emitted by the luciferase-luciferin reaction, has enabled continuous monitoring of various biochemical processes in living animals. Bright luminescence with a high signal-to-background ratio, ideally red or near-infrared light as the emission maximum, is necessary for in vivo animal experiments. Various attempts have been undertaken to achieve this goal, including genetic engineering of luciferase, chemical modulation of luciferin, and utilization of bioluminescence resonance energy transfer (BRET). In this review, we overview a recent advance in the development of a bioluminescence system for in vivo BLI. We also specifically examine the improvement in bioluminescence intensity by mutagenic or chemical modulation on several beetle and marine luciferase bioluminescence systems. We further describe that intramolecular BRET enhances luminescence emission, with recent attempts for the development of red-shifted bioluminescence system, showing great potency in in vivo BLI. Perspectives for future improvement of bioluminescence systems are discussed.
Collapse
Affiliation(s)
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan;
| |
Collapse
|
27
|
Love AC, Prescher JA. Seeing (and Using) the Light: Recent Developments in Bioluminescence Technology. Cell Chem Biol 2020; 27:904-920. [PMID: 32795417 PMCID: PMC7472846 DOI: 10.1016/j.chembiol.2020.07.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 07/24/2020] [Indexed: 02/08/2023]
Abstract
Bioluminescence has long been used to image biological processes in vivo. This technology features luciferase enzymes and luciferin small molecules that produce visible light. Bioluminescent photons can be detected in tissues and live organisms, enabling sensitive and noninvasive readouts on physiological function. Traditional applications have focused on tracking cells and gene expression patterns, but new probes are pushing the frontiers of what can be visualized. The past few years have also seen the merger of bioluminescence with optogenetic platforms. Luciferase-luciferin reactions can drive light-activatable proteins, ultimately triggering signal transduction and other downstream events. This review highlights these and other recent advances in bioluminescence technology, with an emphasis on tool development. We showcase how new luciferins and engineered luciferases are expanding the scope of optical imaging. We also highlight how bioluminescent systems are being leveraged not just for sensing-but also controlling-biological processes.
Collapse
Affiliation(s)
- Anna C Love
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jennifer A Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
28
|
Hypoxia-Inducible Factor 1α (HIF-1α) Counteracts the Acute Death of Cells Transplanted into the Injured Spinal Cord. eNeuro 2020; 7:ENEURO.0092-19.2019. [PMID: 31488552 PMCID: PMC7215587 DOI: 10.1523/eneuro.0092-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/10/2019] [Accepted: 08/19/2019] [Indexed: 01/13/2023] Open
Abstract
Cellular transplantation is in clinical testing for a number of central nervous system disorders, including spinal cord injury (SCI). One challenge is acute transplanted cell death. To prevent this death, there is a need to both establish when the death occurs and develop approaches to mitigate its effects. Here, using luciferase (luc) and green fluorescent protein (GFP) expressing Schwann cell (SC) transplants in the contused thoracic rat spinal cord 7 d postinjury, we establish via in vivo bioluminescent (IVIS) imaging and stereology that cell death occurs prior to 2–3 d postimplantation. We then test an alternative approach to the current paradigm of enhancing transplant survival by including multiple factors along with the cells. To stimulate multiple cellular adaptive pathways concurrently, we activate the hypoxia-inducible factor 1α (HIF-1α) transcriptional pathway. Retroviral expression of VP16-HIF-1α in SCs increased HIF-α by 5.9-fold and its target genes implicated in oxygen transport and delivery (VEGF, 2.2-fold) and cellular metabolism (enolase, 1.7-fold). In cell death assays in vitro, HIF-1α protected cells from H2O2-induced oxidative damage. It also provided some protection against camptothecin-induced DNA damage, but not thapsigargin-induced endoplasmic reticulum stress or tunicamycin-induced unfolded protein response. Following transplantation, VP16-HIF-1α increased SC survival by 34.3%. The increase in cell survival was detectable by stereology, but not by in vivo luciferase or ex vivo GFP IVIS imaging. The results support the hypothesis that activating adaptive cellular pathways enhances transplant survival and identifies an alternative pro-survival approach that, with optimization, could be amenable to clinical translation.
Collapse
|
29
|
Hu H, Zhang Z, Wang R, Wang Y, Jin J, Cai L, Yang J, Duan H, Wu Z, Fang Z, Liu B. BGC823 Cell Line with the Stable Expression of iRFP720 Retains Its Primary Properties with Promising Fluorescence Imaging Ability. DNA Cell Biol 2020; 39:900-908. [PMID: 32096664 DOI: 10.1089/dna.2019.5057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Reliable animal models are required for understanding the molecular events of gastric tumor growth and metastasis. Tracing techniques based on iRFP720 may optimize the noninvasive monitoring of tumors in vivo. The present study established a human gastric adenocarcinoma cell line BGC823-iRFP720-GFP (abbreviated as BGC823-iRFP) that stably expressed iRFP720 and green fluorescent protein (GFP) by piggyBac transposon system. The monoclonal cell line BGC823-iRFP was isolated under puromycin selection. The cell morphology and proliferation ability of BGC823-iRFP cells in vitro were similar to that of the BGC823 cells. The iRFP720 and GFP expressions were confirmed by laser confocal microscopy and Cytation™ 5. Hematoxylin and eosin staining, immunohistochemical analysis, and animal experiments also revealed that BGC823-iRFP exhibited no significant changes in morphology, growth kinetics, and tumorigenicity in vivo. IVIS Lumina III imaging indicated that the iRFP720 signals of the BGC823-iRFP cells could be used to evaluate the antitumor efficacy of oncolytic viruses and chemotherapy drugs. Therefore, the BGC823-iRFP cells would be a useful tool for gastric cancer research and antitumor drug evaluation.
Collapse
Affiliation(s)
- Han Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Ziyi Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Runyang Wang
- Wuhan Binhui Biotechnology Co. Ltd., Wuhan, China
| | - Yang Wang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Jing Jin
- Wuhan Binhui Biotechnology Co. Ltd., Wuhan, China
| | - Linkang Cai
- Wuhan Binhui Biotechnology Co. Ltd., Wuhan, China
| | - Junhan Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Haixiao Duan
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China
| | - Zhen Wu
- Wuhan Binhui Biotechnology Co. Ltd., Wuhan, China
| | | | - Binlei Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan, China.,Wuhan Binhui Biotechnology Co. Ltd., Wuhan, China
| |
Collapse
|
30
|
Coutant EP, Gagnot G, Hervin V, Baatallah R, Goyard S, Jacob Y, Rose T, Janin YL. Bioluminescence Profiling of NanoKAZ/NanoLuc Luciferase Using a Chemical Library of Coelenterazine Analogues. Chemistry 2020; 26:948-958. [PMID: 31765054 DOI: 10.1002/chem.201904844] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Indexed: 12/11/2022]
Abstract
We describe here an extensive structure-bioluminescence relationship study of a chemical library of analogues of coelenterazine, using nanoKAZ/NanoLuc, a mutated luciferase originated from the catalytic subunit of the deep-sea shrimp Oplophorus gracilirostris. Out of the 135 O-acetylated precursors that were prepared by using our recently reported synthesis and following their hydrolysis to give solutions of the corresponding luciferins, notable bioluminescence improvements were achieved in comparison with furimazine, which is currently amongst the best substrates of nanoKAZ/NanoLuc. For instance, the rather more lipophilic analogue 8-(2,3-difluorobenzyl)-2-((5-methylfuran-2-yl)methyl)-6-phenylimidazo[1,2-a]pyrazin-3(7H)-one provided a 1.5-fold improvement of the total light output over a 2 h period, a close to threefold increase of the initial signal intensity and a signal-to-background ratio five times greater than furimazine. The kinetic parameters for the enzymatic reaction were obtained for a selection of luciferin analogues and provided unexpected insights into the luciferase activity. Most prominently, along with a general substrate-dependent and irreversible inactivation of this enzyme, in the case of the optimized luciferin mentioned above, the consumption of 2664 molecules was found to be required for the detection of a single Relative Light Unit (RLU; a luminometer-dependent fraction of a photon).
Collapse
Affiliation(s)
- Eloi P Coutant
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Glwadys Gagnot
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France.,Université Paris Descartes, Sorbonne Paris Cité, 12 rue de l'Ecole de Médecine, 75006, Paris, France
| | - Vincent Hervin
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Racha Baatallah
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Sophie Goyard
- Center for Innovation and Technological Research, Institut Pasteur, 25 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Yves Jacob
- Unité de Génétique Moléculaire des Virus à ARN, Institut Pasteur, UMR 3569, CNRS, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Thierry Rose
- Center for Innovation and Technological Research, Institut Pasteur, 25 rue du Dr. Roux, 75724, Paris Cedex 15, France
| | - Yves L Janin
- Institut Pasteur, UMR 3523, CNRS, Unité de Chimie et Biocatalyse, 28 rue du Dr. Roux, 75724, Paris Cedex 15, France
| |
Collapse
|
31
|
Matlashov ME, Shcherbakova DM, Alvelid J, Baloban M, Pennacchietti F, Shemetov AA, Testa I, Verkhusha VV. A set of monomeric near-infrared fluorescent proteins for multicolor imaging across scales. Nat Commun 2020; 11:239. [PMID: 31932632 PMCID: PMC6957686 DOI: 10.1038/s41467-019-13897-6] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 12/03/2019] [Indexed: 11/09/2022] Open
Abstract
Bright monomeric near-infrared (NIR) fluorescent proteins (FPs) are in high demand as protein tags for multicolor microscopy and in vivo imaging. Here we apply rational design to engineer a complete set of monomeric NIR FPs, which are the brightest genetically encoded NIR probes. We demonstrate that the enhanced miRFP series of NIR FPs, which combine high effective brightness in mammalian cells and monomeric state, perform well in both nanometer-scale imaging with diffraction unlimited stimulated emission depletion (STED) microscopy and centimeter-scale imaging in mice. In STED we achieve ~40 nm resolution in live cells. In living mice we detect ~105 fluorescent cells in deep tissues. Using spectrally distinct monomeric NIR FP variants, we perform two-color live-cell STED microscopy and two-color imaging in vivo. Having emission peaks from 670 nm to 720 nm, the next generation of miRFPs should become versatile NIR probes for multiplexed imaging across spatial scales in different modalities.
Collapse
Affiliation(s)
- Mikhail E Matlashov
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Jonatan Alvelid
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mikhail Baloban
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Francesca Pennacchietti
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anton A Shemetov
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, 10461, USA. .,Medicum, Faculty of Medicine, University of Helsinki, 00029, Helsinki, Finland.
| |
Collapse
|
32
|
Deng H, Liu H, Kang W, Lei C, Nie Z, Huang Y, Yao S. Biomineralization synthesis of a near-infrared fluorescent nanoprobe for direct glucose sensing in whole blood. NANOSCALE 2020; 12:864-870. [PMID: 31833533 DOI: 10.1039/c9nr06691h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A near-infrared (NIR) fluorescent nanoprobe that enables to circumvent the interference of background absorption and fluorescence in whole blood was developed for the direct sensing of blood glucose. Here, NIR fluorescent protein (iRFP) and glucose oxidase (GOx) were collectively deployed as the templates for the biomineralization of Mn2+ to prepare a NIR fluorescent nanoprobe (iRFP-GOx-MnO2 nanoparticles, iRGMs), in which the fluorescence of iRFP was effectively quenched by MnO2via energy transfer. When the iRGMs were mixed with whole blood samples, GOx can convert blood glucose into gluconic acid, as well as H2O2, which will reduce MnO2 and decompose the iRGMs. As a result, the NIR fluorescence of iRFPs was restored, providing a fluorometric assay for the direct detection of blood glucose. Owing to the high efficiency of the cascade reaction and the low background interference of the NIR fluorescence signal, accurate and rapid analysis of the glucose levels in whole blood samples was achieved using the iRGMs. Moreover, an iRGM-based paper device that only requires 5 microliters of samples was also demonstrated in the direct assay of blood glucose without any pretreatment, affording an alternative approach for the accurate monitoring of blood glucose levels.
Collapse
Affiliation(s)
- Honghua Deng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China..
| | | | | | | | | | | | | |
Collapse
|
33
|
Stowe CL, Burley TA, Allan H, Vinci M, Kramer-Marek G, Ciobota DM, Parkinson GN, Southworth TL, Agliardi G, Hotblack A, Lythgoe MF, Branchini BR, Kalber TL, Anderson JC, Pule MA. Near-infrared dual bioluminescence imaging in mouse models of cancer using infraluciferin. eLife 2019; 8:e45801. [PMID: 31610848 PMCID: PMC6828332 DOI: 10.7554/elife.45801] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 09/25/2019] [Indexed: 02/01/2023] Open
Abstract
Bioluminescence imaging (BLI) is ubiquitous in scientific research for the sensitive tracking of biological processes in small animal models. However, due to the attenuation of visible light by tissue, and the limited set of near-infrared bioluminescent enzymes, BLI is largely restricted to monitoring single processes in vivo. Here we show, that by combining stabilised colour mutants of firefly luciferase (FLuc) with the luciferin (LH2) analogue infraluciferin (iLH2), near-infrared dual BLI can be achieved in vivo. The X-ray crystal structure of FLuc with a high-energy intermediate analogue, 5'-O-[N-(dehydroinfraluciferyl)sulfamoyl] adenosine (iDLSA) provides insight into the FLuc-iLH2 reaction leading to near-infrared light emission. The spectral characterisation and unmixing validation studies reported here established that iLH2 is superior to LH2 for the spectral unmixing of bioluminescent signals in vivo; which led to this novel near-infrared dual BLI system being applied to monitor both tumour burden and CAR T cell therapy within a systemically induced mouse tumour model.
Collapse
Affiliation(s)
- Cassandra L Stowe
- Cancer InstituteUniversity College LondonLondonUnited Kingdom
- Centre for Advanced Biomedical ImagingUniversity College LondonLondonUnited Kingdom
| | | | - Helen Allan
- Department of ChemistryUniversity College LondonLondonUnited Kingdom
| | - Maria Vinci
- The Institute of Cancer ResearchLondonUnited Kingdom
| | | | | | | | | | - Giulia Agliardi
- Cancer InstituteUniversity College LondonLondonUnited Kingdom
| | | | - Mark F Lythgoe
- Centre for Advanced Biomedical ImagingUniversity College LondonLondonUnited Kingdom
| | | | - Tammy L Kalber
- Centre for Advanced Biomedical ImagingUniversity College LondonLondonUnited Kingdom
| | - James C Anderson
- Department of ChemistryUniversity College LondonLondonUnited Kingdom
| | - Martin A Pule
- Cancer InstituteUniversity College LondonLondonUnited Kingdom
| |
Collapse
|
34
|
Nomura N, Nishihara R, Nakajima T, Kim SB, Iwasawa N, Hiruta Y, Nishiyama S, Sato M, Citterio D, Suzuki K. Biothiol-Activatable Bioluminescent Coelenterazine Derivative for Molecular Imaging in Vitro and in Vivo. Anal Chem 2019; 91:9546-9553. [PMID: 31291724 DOI: 10.1021/acs.analchem.9b00694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There is a high demand for sensitive biothiol probes targeting cysteine, glutathione, and homocysteine. These biothiols are known as playing essential roles to maintain homeostasis and work as indicators of many diseases. This work presents a bioluminescent probe (named AMCM) to detect biothiols in live mammalian cells and in vivo with a limit of detection of 0.11 μM for cysteine in solution and high selectivity for biothiols, making it suitable for real-time biothiol detection in biological systems. Upon application to live cells, AMCM showed low cytotoxicity and sensitively reported bioluminescence in response to changes of biothiol levels. Furthermore, a bioluminescence resonance energy transfer system consisting of AMCM combined with the near-infrared fluorescent protein iRFP713 was applied to in vivo imaging, with emitted tissue-permeable luminescence in living mice. In summary, this work demonstrates that AMCM is of high practical value for the detection of biothiols in living cells and for deep tissue imaging in living animals.
Collapse
Affiliation(s)
| | | | - Takahiro Nakajima
- Graduate School of Arts and Sciences , The University of Tokyo , 3-8-1 Komaba , Meguro , Tokyo , Japan
| | - Sung Bae Kim
- National Institute of Advanced Industrial Science and Technology , 1-1-1 Umezono , Tsukuba , Ibaraki 305-8560 , Japan
| | | | | | | | - Moritoshi Sato
- Graduate School of Arts and Sciences , The University of Tokyo , 3-8-1 Komaba , Meguro , Tokyo , Japan
| | | | | |
Collapse
|
35
|
Red-shifted bioluminescence Resonance Energy Transfer: Improved tools and materials for analytical in vivo approaches. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.04.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Karasev MM, Stepanenko OV, Rumyantsev KA, Turoverov KK, Verkhusha VV. Near-Infrared Fluorescent Proteins and Their Applications. BIOCHEMISTRY (MOSCOW) 2019; 84:S32-S50. [PMID: 31213194 DOI: 10.1134/s0006297919140037] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
High transparency, low light-scattering, and low autofluorescence of mammalian tissues in the near-infrared (NIR) spectral range (~650-900 nm) open a possibility for in vivo imaging of biological processes at the micro- and macroscales to address basic and applied problems in biology and biomedicine. Recently, probes that absorb and fluoresce in the NIR optical range have been engineered using bacterial phytochromes - natural NIR light-absorbing photoreceptors that regulate metabolism in bacteria. Since the chromophore in all these proteins is biliverdin, a natural product of heme catabolism in mammalian cells, they can be used as genetically encoded fluorescent probes, similarly to GFP-like fluorescent proteins. In this review, we discuss photophysical and biochemical properties of NIR fluorescent proteins, reporters, and biosensors and analyze their characteristics required for expression of these molecules in mammalian cells. Structural features and molecular engineering of NIR fluorescent probes are discussed. Applications of NIR fluorescent proteins and biosensors for studies of molecular processes in cells, as well as for tissue and organ visualization in whole-body imaging in vivo, are described. We specifically focus on the use of NIR fluorescent probes in advanced imaging technologies that combine fluorescence and bioluminescence methods with photoacoustic tomography.
Collapse
Affiliation(s)
- M M Karasev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Medicum, University of Helsinki, Helsinki, 00290, Finland
| | - O V Stepanenko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia.
| | - K A Rumyantsev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Loginov Moscow Clinical Scientific Center, Moscow, 111123, Russia
| | - K K Turoverov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia. .,Peter the Great St. Petersburg Polytechnic University, St. Petersburg, 195251, Russia
| | - V V Verkhusha
- Medicum, University of Helsinki, Helsinki, 00290, Finland. .,Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
37
|
Yeh HW, Ai HW. Development and Applications of Bioluminescent and Chemiluminescent Reporters and Biosensors. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:129-150. [PMID: 30786216 PMCID: PMC6565457 DOI: 10.1146/annurev-anchem-061318-115027] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Although fluorescent reporters and biosensors have become indispensable tools in biological and biomedical fields, fluorescence measurements require external excitation light, thereby limiting their use in thick tissues and live animals. Bioluminescent reporters and biosensors may potentially overcome this hurdle because they use enzyme-catalyzed exothermic biochemical reactions to generate excited-state emitters. This review first introduces the development of bioluminescent reporters, and next, their applications in sensing biological changes in vitro and in vivo as biosensors. Lastly, we discuss chemiluminescent sensors that produce photons in the absence of luciferases. This review aims to explore fundamentals and experimental insights and to emphasize the yet-to-be-reached potential of next-generation luminescent reporters and biosensors.
Collapse
Affiliation(s)
- Hsien-Wei Yeh
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, and Department of Chemistry, University of Virginia, Charlottesville, Virginia 22908, USA;
| | - Hui-Wang Ai
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, and Department of Chemistry, University of Virginia, Charlottesville, Virginia 22908, USA;
| |
Collapse
|
38
|
Bhuckory S, Kays JC, Dennis AM. In Vivo Biosensing Using Resonance Energy Transfer. BIOSENSORS 2019; 9:E76. [PMID: 31163706 PMCID: PMC6628364 DOI: 10.3390/bios9020076] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 05/20/2019] [Accepted: 05/27/2019] [Indexed: 01/05/2023]
Abstract
Solution-phase and intracellular biosensing has substantially enhanced our understanding of molecular processes foundational to biology and pathology. Optical methods are favored because of the low cost of probes and instrumentation. While chromatographic methods are helpful, fluorescent biosensing further increases sensitivity and can be more effective in complex media. Resonance energy transfer (RET)-based sensors have been developed to use fluorescence, bioluminescence, or chemiluminescence (FRET, BRET, or CRET, respectively) as an energy donor, yielding changes in emission spectra, lifetime, or intensity in response to a molecular or environmental change. These methods hold great promise for expanding our understanding of molecular processes not just in solution and in vitro studies, but also in vivo, generating information about complex activities in a natural, organismal setting. In this review, we focus on dyes, fluorescent proteins, and nanoparticles used as energy transfer-based optical transducers in vivo in mice; there are examples of optical sensing using FRET, BRET, and in this mammalian model system. After a description of the energy transfer mechanisms and their contribution to in vivo imaging, we give a short perspective of RET-based in vivo sensors and the importance of imaging in the infrared for reduced tissue autofluorescence and improved sensitivity.
Collapse
Affiliation(s)
- Shashi Bhuckory
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Joshua C Kays
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Allison M Dennis
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
- Division of Materials Science and Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
39
|
Nemukhin AV, Grigorenko BL, Khrenova MG, Krylov AI. Computational Challenges in Modeling of Representative Bioimaging Proteins: GFP-Like Proteins, Flavoproteins, and Phytochromes. J Phys Chem B 2019; 123:6133-6149. [DOI: 10.1021/acs.jpcb.9b00591] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Alexander V. Nemukhin
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Bella L. Grigorenko
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Maria G. Khrenova
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
- Federal Research Center of Biotechnology, Bach Institute of Biochemistry, Russian Academy of Sciences, Moscow 119071, Russian
| | - Anna I. Krylov
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-0482, United States
| |
Collapse
|
40
|
Nishihara R, Paulmurugan R, Nakajima T, Yamamoto E, Natarajan A, Afjei R, Hiruta Y, Iwasawa N, Nishiyama S, Citterio D, Sato M, Kim SB, Suzuki K. Highly bright and stable NIR-BRET with blue-shifted coelenterazine derivatives for deep-tissue imaging of molecular events in vivo. Theranostics 2019; 9:2646-2661. [PMID: 31131059 PMCID: PMC6525985 DOI: 10.7150/thno.32219] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/13/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Bioluminescence imaging (BLI) is one of the most widely used optical platforms in molecular imaging, but it suffers from severe tissue attenuation and autoluminescence in vivo. Methods: Here, we developed a novel BLI platform on the basis of bioluminescence resonance energy transfer (BRET) for achieving a ~300 nm blue-to-near infrared shift of the emission (NIR-BRET) by synthesizing an array of 18 novel coelenterazine (CTZ) derivatives, named "Bottle Blue (BBlue)" and a unique iRFP-linked RLuc8.6-535SG fusion protein as a probe. Results: The best NIR-BRET was achieved by tuning the emission peaks of the CTZ derivatives to a Soret band of the iRFP. In mammalian cells, BBlue2.3, one of the CTZ derivatives, emits light that is ~50-fold brighter than DBlueC when combined with RLuc8.6-535SG, which shows stable BL kinetics. When we used a caged version of BBLue2.3, it showed a BL half decay time of over 60 minutes while maintaining the higher signal sensitivity. This NIR BL is sufficiently brighter to be used for imaging live mammalian cells at single cell level, and also for imaging metastases in deep tissues in live mice without generating considerable autoluminescence. A single-chain probe developed based on this BLI platform allowed us to sensitively image ligand antagonist-specific activation of estrogen receptor in the NIR region. Conclusion: This unique optical platform provides the brightest NIR BLI template that can be used for imaging a diverse group of cellular events in living subjects including protein‒protein interactions and cancer metastasis.
Collapse
Affiliation(s)
- Ryo Nishihara
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Takahiro Nakajima
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba Meguro-ku, Tokyo 153-8902, Japan
| | - Eiji Yamamoto
- Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Arutselvan Natarajan
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Rayhaneh Afjei
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Yuki Hiruta
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Naoko Iwasawa
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Shigeru Nishiyama
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Daniel Citterio
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Moritoshi Sato
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba Meguro-ku, Tokyo 153-8902, Japan
| | - Sung Bae Kim
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine, Palo Alto, California 94304, United States
- Research Institute for Environmental Management Technology, National Institute of Advanced Industrial Science and Technology (AIST), 16-1 Onogawa, Tsukuba 305-8569, Japan
| | - Koji Suzuki
- Department of Applied Chemistry, Faculty of Science and Technology, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| |
Collapse
|
41
|
Dale NC, Johnstone EKM, White CW, Pfleger KDG. NanoBRET: The Bright Future of Proximity-Based Assays. Front Bioeng Biotechnol 2019; 7:56. [PMID: 30972335 PMCID: PMC6443706 DOI: 10.3389/fbioe.2019.00056] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022] Open
Abstract
Bioluminescence resonance energy transfer (BRET) is a biophysical technique used to monitor proximity within live cells. BRET exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate. This results in production of a quantifiable signal that denotes proximity between proteins and/or molecules tagged with complementary luciferase and fluorophore partners. BRET assays have been used to observe an array of biological functions including ligand binding, intracellular signaling, receptor-receptor proximity, and receptor trafficking, however, BRET assays can theoretically be used to monitor the proximity of any protein or molecule for which appropriate fusion constructs and/or fluorophore conjugates can be produced. Over the years, new luciferases and approaches have been developed that have increased the potential applications for BRET assays. In particular, the development of the small, bright and stable Nanoluciferase (NanoLuc; Nluc) and its use in NanoBRET has vastly broadened the potential applications of BRET assays. These advances have exciting potential to produce new experimental methods to monitor protein-protein interactions (PPIs), protein-ligand interactions, and/or molecular proximity. In addition to NanoBRET, Nluc has also been exploited to produce NanoBiT technology, which further broadens the scope of BRET to monitor biological function when NanoBiT is combined with an acceptor. BRET has proved to be a powerful tool for monitoring proximity and interaction, and these recent advances further strengthen its utility for a range of applications.
Collapse
Affiliation(s)
- Natasha C Dale
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Carl W White
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia.,Australian Research Council Centre for Personalised Therapeutics TechnologiesAustralia.,Dimerix Limited, Nedlands, WA, Australia
| |
Collapse
|
42
|
Integration of Nanomaterials and Bioluminescence Resonance Energy Transfer Techniques for Sensing Biomolecules. BIOSENSORS-BASEL 2019; 9:bios9010042. [PMID: 30884844 PMCID: PMC6468577 DOI: 10.3390/bios9010042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 01/11/2023]
Abstract
Bioluminescence resonance energy transfer (BRET) techniques offer a high degree of sensitivity, reliability and ease of use for their application to sensing biomolecules. BRET is a distance dependent, non-radiative energy transfer, which uses a bioluminescent protein to excite an acceptor through the resonance energy transfer. A BRET sensor can quickly detect the change of a target biomolecule quantitatively without an external electromagnetic field, e.g., UV light, which normally can damage tissue. Having been developed quite recently, this technique has evolved rapidly. Here, different bioluminescent proteins have been reviewed. In addition to a multitude of bioluminescent proteins, this manuscript focuses on the recent development of BRET sensors by utilizing quantum dots. The special size-dependent properties of quantum dots have made the BRET sensing technique attractive for the real-time monitoring of the changes of target molecules and bioimaging in vivo. This review offers a look into the basis of the technique, donor/acceptor pairs, experimental applications and prospects.
Collapse
|
43
|
Wang H, Mu X, Yang J, Liang Y, Zhang XD, Ming D. Brain imaging with near-infrared fluorophores. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2018.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Tsuboi S, Jin T. BRET based dual-colour (visible/near-infrared) molecular imaging using a quantum dot/EGFP–luciferase conjugate. RSC Adv 2019; 9:34964-34971. [PMID: 35530680 PMCID: PMC9074158 DOI: 10.1039/c9ra07011g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/17/2019] [Indexed: 12/25/2022] Open
Abstract
Owing to its high sensitivity, bioluminescence imaging is an important tool for biosensing and bioimaging in life sciences. Compared to fluorescence imaging, bioluminescence imaging has a superior advantage that the background signals resulting from autofluorescence are almost zero. In addition, bioluminescence imaging can permit long-term observation of living cells because external excitation is not needed, leading to no photobleaching and photocytotoxicity. Although bioluminescence imaging has such superior properties over fluorescence imaging, observation wavelengths in bioluminescence imaging are mostly limited to the visible region. Here we present bioluminescence resonance energy transfer (BRET) based dual-colour (visible/near-infrared) molecular imaging using a quantum dot (QD) and luciferase protein conjugate. This bioluminescent probe is designed to emit green and near-infrared luminescence from enhanced green fluorescent protein (EGFP) and CdSeTe/CdS (core/shell) QDs, where EGFP–Renilla luciferase (RLuc) fused proteins are conjugated to the QDs. Since the EGFP–RLuc fused protein contains an immunoglobulin binding domain (GB1) of protein G, it is possible to prepare a variety of molecular imaging probes functionalized with antibodies (IgG). We show that the BRET-based QD probe can be used for highly sensitive dual-colour (visible/near-infrared) bioluminescence molecular imaging of membrane receptors in cancer cells. A bioluminescent dual-colour molecular-imaging probe was prepared to emit green and near-infrared luminescence from a conjugate between enhanced green fluorescent protein (EGFP), Renilla luciferase (RLuc) and CdSeTe/CdS quantum dot (QD).![]()
Collapse
Affiliation(s)
- Setsuko Tsuboi
- RIKEN Center for Biosystems Dynamics Research (BDR)
- Osaka 565-0874
- Japan
| | - Takashi Jin
- RIKEN Center for Biosystems Dynamics Research (BDR)
- Osaka 565-0874
- Japan
- Graduate School of Frontier Biosciences
- Osaka University
| |
Collapse
|
45
|
Osipova ZM, Shcheglov AS, Yampolsky IV. Bioluminescent imaging: new opportunities. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2018. [DOI: 10.24075/brsmu.2018.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Modern biomedical research technologies actively use bioimaging for studying cells, tissues and whole organisms. Multicolor bioimaging is applied when simultaneous observation of different events at the molecular and cellular level is needed. Bioluminescent imaging methods are the most sensitive, however, their use for multicolor labeling is complicated due to the insufficient number of available uciferin-luciferase pairs. Having a number of advantages compared to previously studied bioluminescent systems, the new bioluminescence systems of higher fungi and marine polychaete Odontosyllis could become a useful expansion of the bioimaging toolbox.
Collapse
Affiliation(s)
- ZM Osipova
- Biomolecular Chemistry Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow; Laboratory of Natural Compounds Chemistry, Pirogov Russian National Research Medical University, Moscow
| | - AS Shcheglov
- Biomolecular Chemistry Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow; Laboratory of Natural Compounds Chemistry, Pirogov Russian National Research Medical University, Moscow
| | - IV Yampolsky
- Biomolecular Chemistry Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow; Laboratory of Natural Compounds Chemistry, Pirogov Russian National Research Medical University, Moscow
| |
Collapse
|
46
|
Yao Z, Zhang BS, Prescher JA. Advances in bioluminescence imaging: new probes from old recipes. Curr Opin Chem Biol 2018; 45:148-156. [PMID: 29879594 PMCID: PMC6076869 DOI: 10.1016/j.cbpa.2018.05.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 11/18/2022]
Abstract
Bioluminescent probes are powerful tools for visualizing biology in live tissues and whole animals. Recent years have seen a surge in the number of new luciferases, luciferins, and related tools available for bioluminescence imaging. Many were crafted using classic methods of optical probe design and engineering. Here we highlight recent advances in bioluminescent tool discovery and development, along with applications of the probes in cells, tissues, and organisms. Collectively, these tools are improving in vivo imaging capabilities and bolstering new research directions.
Collapse
Affiliation(s)
- Zi Yao
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Brendan S Zhang
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Jennifer A Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology & Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
47
|
Shcherbakova DM, Stepanenko OV, Turoverov KK, Verkhusha VV. Near-Infrared Fluorescent Proteins: Multiplexing and Optogenetics across Scales. Trends Biotechnol 2018; 36:1230-1243. [PMID: 30041828 DOI: 10.1016/j.tibtech.2018.06.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 10/28/2022]
Abstract
Since mammalian tissue is relatively transparent to near-infrared (NIR) light, NIR fluorescent proteins (FPs) engineered from bacterial phytochromes have become widely used probes for non-invasive in vivo imaging. Recently, these genetically encoded NIR probes have been substantially improved, enabling imaging experiments that were not possible previously. Here, we discuss the use of monomeric NIR FPs and NIR biosensors for multiplexed imaging with common visible GFP-based probes and blue light-activatable optogenetic tools. These NIR probes are suitable for visualization of functional activities from molecular to organismal levels. In combination with advanced imaging techniques, such as two-photon microscopy with adaptive optics, photoacoustic tomography and its recent modification reversibly switchable photoacoustic computed tomography, NIR probes allow subcellular resolution at millimeter depths.
Collapse
Affiliation(s)
- Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Olesya V Stepanenko
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg 194064, Russian Federation
| | - Konstantin K Turoverov
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg 194064, Russian Federation; Department of Biophysics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg 195251, Russian Federation
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland.
| |
Collapse
|
48
|
Dhadve A, Thakur B, Ray P. Construction of Dual Modality Optical Reporter Gene Constructs for Bioluminescent and Fluorescent Imaging. Methods Mol Biol 2018; 1790:13-27. [PMID: 29858780 DOI: 10.1007/978-1-4939-7860-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Dual modality reporter genes are powerful means of tracking cellular processes in cell culture systems and whole animals. In this chapter, we describe the methods for construction of a plasmid reporter gene vector expressing a fluorescent and a bioluminescent gene and its validation by in vitro assays in mammalian cells as well as by noninvasive imaging methods in small animal models.
Collapse
Affiliation(s)
- Ajit Dhadve
- Imaging Cell Signaling & Therapeutics Lab, Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Bhushan Thakur
- Imaging Cell Signaling & Therapeutics Lab, Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, India
| | - Pritha Ray
- Imaging Cell Signaling & Therapeutics Lab, Tata Memorial Centre (TMC), Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, Maharashtra, India. .,Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
49
|
Tsuboi S, Jin T. Recombinant Protein (Luciferase-IgG Binding Domain) Conjugated Quantum Dots for BRET-Coupled Near-Infrared Imaging of Epidermal Growth Factor Receptors. Bioconjug Chem 2018. [DOI: 10.1021/acs.bioconjchem.8b00149] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Setsuko Tsuboi
- Quantitative Biology Center, RIKEN, Furuedai 6-2-3, Suita, Osaka 565-0874, Japan
| | - Takashi Jin
- Quantitative Biology Center, RIKEN, Furuedai 6-2-3, Suita, Osaka 565-0874, Japan
- Graduate School of Frontier Biosciences, Osaka University, Yamada-oka 2-1, Suita, Osaka 565-0871, Japan
| |
Collapse
|
50
|
Hall MP, Woodroofe CC, Wood MG, Que I, Van't Root M, Ridwan Y, Shi C, Kirkland TA, Encell LP, Wood KV, Löwik C, Mezzanotte L. Click beetle luciferase mutant and near infrared naphthyl-luciferins for improved bioluminescence imaging. Nat Commun 2018; 9:132. [PMID: 29317625 PMCID: PMC5760652 DOI: 10.1038/s41467-017-02542-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
The sensitivity of bioluminescence imaging in animals is primarily dependent on the amount of photons emitted by the luciferase enzyme at wavelengths greater than 620 nm where tissue penetration is high. This area of work has been dominated by firefly luciferase and its substrate, D-luciferin, due to the system's peak emission (~ 600 nm), high signal to noise ratio, and generally favorable biodistribution of D-luciferin in mice. Here we report on the development of a codon optimized mutant of click beetle red luciferase that produces substantially more light output than firefly luciferase when the two enzymes are compared in transplanted cells within the skin of black fur mice or in deep brain. The mutant enzyme utilizes two new naphthyl-luciferin substrates to produce near infrared emission (730 nm and 743 nm). The stable luminescence signal and near infrared emission enable unprecedented sensitivity and accuracy for performing deep tissue multispectral tomography in mice.
Collapse
Affiliation(s)
- Mary P Hall
- Promega Corporation, Madison, WI, 53711, USA
| | - Carolyn C Woodroofe
- Promega Corporation, Madison, WI, 53711, USA
- Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | | | - Ivo Que
- Department of Radiology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Moniek Van't Root
- Optical molecular imaging, Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Yanto Ridwan
- Optical molecular imaging, Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Ce Shi
- Promega Biosciences Incorporated, San Luis Obispo, CA, 93401, USA
| | | | | | | | - Clemens Löwik
- Optical molecular imaging, Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Laura Mezzanotte
- Optical molecular imaging, Department of Radiology & Nuclear Medicine, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands.
- Department of Molecular Genetics, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|