1
|
Lu X, Pritko DJ, Abravanel ME, Huggins JR, Ogunleye O, Biswas T, Ashy KC, Woods SK, Livingston MWT, Blenner MA, Birtwistle MR. Genetically Encoded Fluorescence Barcodes Allow for Single-Cell Analysis via Spectral Flow Cytometry. ACS Synth Biol 2025; 14:1533-1548. [PMID: 40326708 DOI: 10.1021/acssynbio.4c00807] [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: 05/07/2025]
Abstract
Genetically encoded, single-cell barcodes are broadly useful for experimental tasks such as lineage tracing or genetic screens. For such applications, a barcode library would ideally have high diversity (many unique barcodes), nondestructive identification (repeated measurements in the same cells or population), and fast, inexpensive readout (many cells and conditions). Current nucleic acid barcoding methods generate high diversity but require destructive and slow/expensive readout, and current fluorescence barcoding methods are nondestructive, fast, and inexpensive to readout but lack high diversity. We recently proposed a theory for how fluorescent protein combinations may generate a high-diversity barcode library with nondestructive, fast, and inexpensive identification. Here, we present an initial experimental proof-of-concept by generating a library of ∼150 barcodes from two-way combinations of 18 fluorescent proteins, 61 of which are tested experimentally. We use a pooled cloning strategy to generate a barcode library that is validated to contain every possible combination of the 18 fluorescent proteins. Experimental results using single mammalian cells and spectral flow cytometry demonstrate excellent classification performance of individual fluorescent proteins, with the exception of mTFP1, and of most evaluated barcodes, with many true positive rates >99%. The library is compatible with genetic screening for hundreds of genes (or gene pairs) and lineage tracing hundreds of clones. This work lays a foundation for greater diversity libraries (potentially ∼105 and more) generated from hundreds of spectrally resolvable tandem fluorescent protein probes.
Collapse
Affiliation(s)
- Xiaoming Lu
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Daniel J Pritko
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Megan E Abravanel
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Jonah R Huggins
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Oluwaferanmi Ogunleye
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Tirthankar Biswas
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Katia C Ashy
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Semaj K Woods
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Mariclaire W T Livingston
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Mark A Blenner
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
2
|
Galiakhmetova D, Koviarov A, Dremin V, Gric T, Stoliarov D, Gorodetsky A, Maimaris M, Shcherbakova DM, Baloban M, Verkhusha VV, Sokolovski SG, Rafailov E. Nonlinear optical properties of photosensory core modules of monomeric and dimeric bacterial phytochromes. Protein Sci 2025; 34:e70118. [PMID: 40248855 PMCID: PMC12006755 DOI: 10.1002/pro.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
Near-infrared (NIR) fluorescent proteins and optogenetic tools derived from bacterial phytochromes' photosensory core modules (PCMs) operate within the first (NIR-I) tissue transparency window under single-photon activation. Leveraging two-photon (2P) light in the second transparency window (NIR-II) for photoswitching bacterial phytochromes between Pr and Pfr absorption states offers significant advantages, including enhanced tissue penetration, spatial resolution, and signal-to-noise ratio. However, 2P photoconversion of bacterial phytochromes remains understudied. Here, we study the non-linear Pr to Pfr photoconversion's dependence on irradiation wavelength (1180-1360 nm) and energy fluence (41-339 mJ/cm2) for the PCM of DrBphP bacterial phytochrome. Our findings reveal substantially higher photoconversion efficiency for the engineered monomeric DrBphP-PCM (73%) compared to the natural dimeric DrBphP-PCM (57%). Molecular mechanical calculations, based on experimentally determined 2P absorption cross-section coefficients for the monomer (167 GM) and dimer (170 GM), further verify these results. We demonstrate both short- (SWE) and long-wavelength excitation (LWE) fluorescence of the Soret band using 405 and 810-890 nm laser sources, respectively. Under LWE, fluorescence emission (724 nm) exhibits saturation at a peak power density of 1.5 GW/cm2. For SWE, we observe linear degradation of fluorescence for both DrBphP-PCMs, decreasing by 32% as the temperature rises from 19 to 38°C. Conversely, under LWE, the monomeric DrBphP-PCM's brightness increases up to 182% (at 37°C), surpassing the dimeric form's fluorescence rise by 39%. These findings establish the monomeric DrBphP-PCM as a promising template for developing NIR imaging and optogenetic probes operating under the determined optimal parameters for its 2P photoconversion and LWE fluorescence.
Collapse
Affiliation(s)
| | | | - Viktor Dremin
- Aston Institute of Photonic TechnologiesAston UniversityBirminghamUK
| | - Tatjana Gric
- Department of Electronic SystemsVilnius Gediminas Technical UniversityVilniusLithuania
| | - Dmitrii Stoliarov
- Aston Institute of Photonic TechnologiesAston UniversityBirminghamUK
| | | | - Marios Maimaris
- Ultrafast Optoelectronics GroupImperial College LondonLondonUK
| | - Daria M. Shcherbakova
- Department of Genetics and Gruss‐Lipper Biophotonics CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Mikhail Baloban
- Department of Genetics and Gruss‐Lipper Biophotonics CenterAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Vladislav V. Verkhusha
- Department of Genetics and Gruss‐Lipper Biophotonics CenterAlbert Einstein College of MedicineBronxNew YorkUSA
- Medicum, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - Edik Rafailov
- Aston Institute of Photonic TechnologiesAston UniversityBirminghamUK
| |
Collapse
|
3
|
Inoue N, Saito T, Wada I. Noncanonical phagocytosis-like SEAL establishes mammalian fertilization. Cell Rep 2025; 44:115463. [PMID: 40138310 DOI: 10.1016/j.celrep.2025.115463] [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: 09/24/2024] [Revised: 12/01/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
In many forms of sexual reproduction, only the most robust spermatozoa, which overcome multiple physiological challenges, reach the oocyte. However, the exact mechanisms of gamete recognition and fusion are unknown. In the present study, we demonstrated that with the onset of gamete recognition, oocyte microvilli form lamellipodium-like structures, activate actin polymerization, and subsequently engulf spermatozoa to initiate gamete fusion. Gamete fusion occurred via a phagocytosis-like process we termed "sperm engulfment activated by IZUMO1-JUNO linkage and gamete fusion-related factors" (SEAL). Gamete adhesion was strictly regulated by binding of sperm IZUMO1 to oocyte JUNO, while SEAL was primarily mediated by sperm DCST1/2, SPACA6, TMEM95, FIMP, and TMEM81, the essential factors for gamete fusion. Interestingly, JUNO was almost depleted from oocyte surfaces in the region where SEAL enveloped spermatozoa by microvilli without actin polymerization. SEAL formation was recapitulated using JUNO-expressing K562 lymphocytic cells rather than oocytes. Together, these findings suggest that dynamic rearrangement of membrane components facilitates SEAL prior to successful fertilization.
Collapse
Affiliation(s)
- Naokazu Inoue
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan.
| | - Takako Saito
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan; Department of Applied Life Sciences, Faculty of Agriculture, Shizuoka University, Shizuoka 422-8529, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
4
|
Bu X, Ashby N, Vitali T, Lee S, Gottumukkala A, Yun K, Tabbara S, Latham P, Teal C, Chung I. Cell crowding activates pro-invasive mechanotransduction pathway in high-grade DCIS via TRPV4 inhibition and cell volume reduction. eLife 2025; 13:RP100490. [PMID: 40256993 PMCID: PMC12011371 DOI: 10.7554/elife.100490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025] Open
Abstract
Cell crowding is a common microenvironmental factor influencing various disease processes, but its role in promoting cell invasiveness remains unclear. This study investigates the biomechanical changes induced by cell crowding, focusing on pro-invasive cell volume reduction in ductal carcinoma in situ (DCIS). Crowding specifically enhanced invasiveness in high-grade DCIS cells through significant volume reduction compared to hyperplasia-mimicking or normal cells. Mass spectrometry revealed that crowding selectively relocated ion channels, including TRPV4, to the plasma membrane in high-grade DCIS cells. TRPV4 inhibition triggered by crowding decreased intracellular calcium levels, reduced cell volume, and increased invasion and motility. During this process, TRPV4 membrane relocation primed the channel for later activation, compensating for calcium loss. Analyses of patient-derived breast cancer tissues confirmed that plasma membrane-associated TRPV4 is specific to high-grade DCIS and indicates the presence of a pro-invasive cell volume reduction mechanotransduction pathway. Hyperosmotic conditions and pharmacologic TRPV4 inhibition mimicked crowding-induced effects, while TRPV4 activation reversed them. Silencing TRPV4 diminished mechanotransduction in high-grade DCIS cells, reducing calcium depletion, volume reduction, and motility. This study uncovers a novel pro-invasive mechanotransduction pathway driven by cell crowding and identifies TRPV4 as a potential biomarker for predicting invasion risk in DCIS patients.
Collapse
Affiliation(s)
- Xiangning Bu
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
| | - Nathanael Ashby
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
| | - Teresa Vitali
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
| | - Sulgi Lee
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
| | - Ananya Gottumukkala
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
- Thomas Jefferson High School for Science and TechnologyAlexandriaUnited States
| | - Kangsun Yun
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
| | - Sana Tabbara
- Department of Pathology, George Washington Medical Faculty AssociatesWashington, DCUnited States
| | - Patricia Latham
- Department of Pathology, George Washington Medical Faculty AssociatesWashington, DCUnited States
| | - Christine Teal
- Department of Surgery, George Washington Medical Faculty AssociatesWashington, DCUnited States
| | - Inhee Chung
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington UniversityWashington, DCUnited States
- Department of Biomedical Engineering, GW School of Engineering and Applied Science, George Washington UniversityWashington, DCUnited States
| |
Collapse
|
5
|
Hariharan V, White JA, Dragoni F, Fray EJ, Pathoulas N, Moskovljevic M, Zhang H, Singhal A, Lai J, Beg SA, Scully EP, Gilliams EA, Block DS, Keruly J, Moore RD, Siliciano JD, Simonetti FR, Siliciano RF. Superinfection with intact HIV-1 results in conditional replication of defective proviruses and nonsuppressible viremia in people living with HIV-1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647291. [PMID: 40236094 PMCID: PMC11996531 DOI: 10.1101/2025.04.04.647291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
During replication of some RNA viruses, defective particles can spontaneously arise and interfere with wild-type (WT) virus replication. Recently, engineered versions of these defective interfering particles (DIPs) have been proposed as an HIV-1 therapeutic. However, DIPs have yet to be reported in people with HIV-1 (PWH). Here, we find DIPs in PWH who have a rare, polyclonal form of non-suppressible viremia (NSV). While antiretroviral therapy (ART) rapidly reduces viremia to undetectable levels, some individuals experience sustained viremia due to virus production from cell clones harboring intact or defective proviruses. We characterized the source of NSV in two PWH who never reached undetectable viral load despite ART adherence. Remarkably, in each participant, we found a diverse set of defective viral genomes all sharing the same fatal deletions. We found that this paradoxical accumulation of mutations by viruses with fatal defects was driven by superinfection with intact viruses, resulting in mobilization of defective genomes and accumulation of additional mutations during untreated infection. We show that these defective proviruses interfere with WT virus replication, conditionally replicate, and, in one case, have an R 0 > 1, enabling in vivo spread. Despite this, clinical outcomes show no evidence of a beneficial effect of these DIPs.
Collapse
|
6
|
Lu X, Pritko DJ, Abravanel ME, Huggins JR, Ogunleye O, Biswas T, Ashy KC, Woods SK, Livingston MW, Blenner MA, Birtwistle MR. Genetically-Encoded Fluorescence Barcodes Allow for Single-Cell Analysis via Spectral Flow Cytometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.23.619855. [PMID: 39484616 PMCID: PMC11526929 DOI: 10.1101/2024.10.23.619855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Genetically-encoded, single-cell barcodes are broadly useful for experimental tasks such as lineage tracing or genetic screens. For such applications, a barcode library would ideally have high diversity (many unique barcodes), non-destructive identification (repeated measurements in the same cells or population), and fast, inexpensive readout (many cells and conditions). Current nucleic acid barcoding methods generate high diversity but require destructive and slow/expensive readout, and current fluorescence barcoding methods are non-destructive, fast, and inexpensive to readout but lack high diversity. We recently proposed theory for how fluorescent protein combinations may generate a high-diversity barcode library with non-destructive, fast and inexpensive identification. Here, we present an initial experimental proof-of-concept by generating a library of ~150 barcodes from two-way combinations of 18 fluorescent proteins, 61 of which are tested experimentally. We use a pooled cloning strategy to generate a barcode library that is validated to contain every possible combination of the 18 fluorescent proteins. Experimental results using single mammalian cells and spectral flow cytometry demonstrate excellent classification performance of individual fluorescent proteins, with the exception of mTFP1, and of most evaluated barcodes, with many true positive rates >99%. The library is compatible with genetic screening for hundreds of genes (or gene pairs) and lineage tracing hundreds of clones. This work lays a foundation for greater diversity libraries (potentially ~105 and more) generated from hundreds of spectrally-resolvable tandem fluorescent protein probes.
Collapse
Affiliation(s)
- Xiaoming Lu
- Department of Chemical and Biomolecular Engineering, Clemson University
| | - Daniel J. Pritko
- Department of Chemical and Biomolecular Engineering, Clemson University
| | | | - Jonah R. Huggins
- Department of Chemical and Biomolecular Engineering, Clemson University
| | | | - Tirthankar Biswas
- Department of Chemical and Biomolecular Engineering, Clemson University
| | - Katia C. Ashy
- Department of Chemical and Biomolecular Engineering, Clemson University
| | - Semaj K. Woods
- Department of Chemical and Biomolecular Engineering, Clemson University
| | | | - Mark A. Blenner
- Department of Chemical and Biomolecular Engineering, University of Delaware
| | | |
Collapse
|
7
|
Mao S, Wu R, Luo W, Qin J, Chen A. Spuriously transcribed RNAs from CRISPR-sgRNA expression plasmids scaffold biomolecular condensate formation and hamper accurate genomic imaging. Nucleic Acids Res 2025; 53:gkaf192. [PMID: 40119729 PMCID: PMC11928936 DOI: 10.1093/nar/gkaf192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/13/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-based imaging tools that utilize fluorescently tagged single-guide RNAs (sgRNAs) have enabled versatile analysis of the dynamics of single genomic loci, but the accuracy may be hindered by nonspecific subnuclear probe accumulation, generating false-positive foci in cell nuclei. By examining the subcellular localizations of sgRNA expression plasmids, their RNA transcripts, and several RNA-binding proteins, we found that spuriously transcribed (cryptic) transcripts, produced by sgRNA expression plasmids, are the major contributors of false-positive signals, independent of sgRNA scaffold design or effector probe (i.e. RNA aptamer- or oligonucleotide-based probes) used. These transcripts interact with the paraspeckle core proteins, but not with the sgRNA expression plasmids or the paraspeckle RNA scaffold NEAT1_2, to form nuclear bodies that display liquid-like properties including sphericality, fusion competence, and sensitivity to 1,6-hexanediol. Transfecting sgRNA transcription units (i.e. sgRNA expression cassettes), lacking the plasmid backbones, reduces false-positive signals and enhances genomic imaging accuracy. Overall, this study unveils previously undescribed activities of cryptic plasmid transcripts and presents an easy-to-adapt strategy that can potentially improve the precision of CRISPR-based imaging systems that implement fluorescently tagged sgRNAs.
Collapse
Affiliation(s)
- Shiqi Mao
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Ruonan Wu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Weibang Luo
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Jinshan Qin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Antony K Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
8
|
El Hajji L, Bunel B, Joliot O, Li C, Tebo AG, Rampon C, Volovitch M, Fischer E, Pietrancosta N, Perez F, Morin X, Vriz S, Gautier A. A tunable and versatile chemogenetic near-infrared fluorescent reporter. Nat Commun 2025; 16:2594. [PMID: 40091099 PMCID: PMC11911394 DOI: 10.1038/s41467-025-58017-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Near-infrared (NIR) fluorescent reporters open interesting perspectives for multiplexed imaging with higher contrast and depth using less toxic light. Here, we propose nirFAST, a small (14 kDa) chemogenetic NIR fluorescent reporter, displaying higher cellular brightness compared to top-performing NIR fluorescent proteins. nirFAST binds and stabilizes the fluorescent state of synthetic cell permeant fluorogenic chromophores (so-called fluorogens), otherwise dark when free. nirFAST displays tunable NIR, far-red or red emission through change of fluorogen. nirFAST allows imaging and spectral multiplexing in live cultured mammalian cells, chicken embryo tissues and zebrafish larvae. Its suitability for stimulated emission depletion nanoscopy enabled protein imaging with subdiffraction resolution in live cells. nirFAST enabled the design of a two-color cell cycle indicator for monitoring the different phases of the cell cycle. Finally, bisection of nirFAST allowed the design of a chemically induced dimerization technology with NIR fluorescence readout, enabling the control and visualization of protein proximity.
Collapse
Affiliation(s)
- Lina El Hajji
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
- Institut Curie, INSERM, CNRS, Chemical Biology of Cancer (CBC), 75005, Paris, France
| | - Benjamin Bunel
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Octave Joliot
- Institut Curie, Université PSL, CNRS UMR144, Paris, France
| | - Chenge Li
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
| | - Alison G Tebo
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
- Howard Hughes Medical Institute - Janelia Research Campus, Ashburn, VA, USA
| | - Christine Rampon
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
- Université Paris Cité, 75006, Paris, France
| | - Michel Volovitch
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
| | - Evelyne Fischer
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Nicolas Pietrancosta
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Franck Perez
- Institut Curie, Université PSL, CNRS UMR144, Paris, France
| | - Xavier Morin
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Sophie Vriz
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France
- Université Paris Cité, 75006, Paris, France
| | - Arnaud Gautier
- Sorbonne Université, École Normale Supérieure, Université PSL, CNRS, Chimie Physique et Chimie du Vivant (CPCV), 75005, Paris, France.
- Institut Curie, INSERM, CNRS, Chemical Biology of Cancer (CBC), 75005, Paris, France.
- Institut Universitaire de France, Paris, France.
| |
Collapse
|
9
|
Chen Z, Chen W, Xu C, Song H, Ji X, Jiang H, Duan H, Li Z, Gao W, Yao T, Zhang Z, He L, Yin Y, Yang N, Tian W, Wu J, Li X. Near-infrared fluorogenic RNA for in vivo imaging and sensing. Nat Commun 2025; 16:518. [PMID: 39788937 PMCID: PMC11718054 DOI: 10.1038/s41467-024-55093-1] [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: 01/07/2024] [Accepted: 11/29/2024] [Indexed: 01/12/2025] Open
Abstract
Fluorogenic RNA aptamers have various applications, including use as fluorescent tags for imaging RNA trafficking and as indicators of RNA-based sensors that exhibit fluorescence upon binding small-molecule fluorophores in living cells. Current fluorogenic RNA:fluorophore complexes typically emit visible fluorescence. However, it is challenging to develop fluorogenic RNA with near-infrared (NIR) fluorescence for in vivo imaging and sensing studies. To address this issue, we identify and modulate red fluorescent protein-like fluorophores to bind Squash, a highly folded fluorogenic RNA. One of these fluorophores, DFQL-1T, exhibits photostable NIR fluorescence when bound to Squash, enabling RNA visualization in living mammalian cells and mice. With Squash:DFQL-1T complexes, we generate RNA-based sensors for detecting non-coding RNAs and small molecule targets in living mammalian cells and in mice. These studies reveal a fluorogenic RNA:fluorophore complex that can be readily developed into NIR fluorescent RNA tags for in vivo imaging and sensing.
Collapse
Affiliation(s)
- Zhenyin Chen
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- Department of Pulmonary and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, Sichuan, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wei Chen
- Centre for Medical Genetics, School of Life Sciences, Central South University, 410078, Changsha, China
- Hengyang Key Laboratory of Cellular Stress Biology, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Cun Xu
- Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012, Changchun, China
| | - Haozhi Song
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xin Ji
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Haodong Jiang
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Hongtao Duan
- College of Life Sciences, Hebei University, 071002, Baoding, Hebei, China
| | - Zehao Li
- College of Life Sciences, Hebei University, 071002, Baoding, Hebei, China
| | - Wankai Gao
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Tuoxin Yao
- College of Life Sciences, Hunan Normal University, 410081, Changsha, China
| | - Zhongxuan Zhang
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Liuqin He
- College of Life Sciences, Hunan Normal University, 410081, Changsha, China
| | - Yulong Yin
- Yuelushan Laboratory, 410128, Changsha, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Nanyang Yang
- Centre for Medical Genetics, School of Life Sciences, Central South University, 410078, Changsha, China
| | - Wenjing Tian
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012, Changchun, China
| | - Jiahui Wu
- Department of Chemistry, University of Massachusetts, 01003, Amherst, MA, USA
| | - Xing Li
- Interdisciplinary Science Center, State Key Laboratory of Integrated Management of Pest Insects and Rodents,Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- Department of Pulmonary and Critical Care Medicine, the Affiliated Hospital of Southwest Medical University, 646000, Luzhou, Sichuan, China.
- College of Life Sciences, Hebei University, 071002, Baoding, Hebei, China.
| |
Collapse
|
10
|
Cheetham-Wilkinson IJ, Sivalingam B, Flitton C, Flottmann F, Vehling L, Drechsler M, Stojchevska M, Raimondi A, Paululat A, Fröhlich F, Swan LE, Stagi M. RpH-ILV: Probe for lysosomal pH and acute LLOMe-induced membrane permeabilization in cell lines and Drosophila. SCIENCE ADVANCES 2025; 11:eadr7325. [PMID: 39752501 PMCID: PMC11698090 DOI: 10.1126/sciadv.adr7325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025]
Abstract
Lysosomal pH dysregulation is a critical element of the pathophysiology of neurodegenerative diseases, cancers, and lysosomal storage disorders (LSDs). To study the role of lysosomes in pathophysiology, probes to analyze lysosomal size, positioning, and pH are indispensable tools. Here, we developed and characterized a ratiometric genetically encoded lysosomal pH probe, RpH-ILV, targeted to a subpopulation of lysosomal intraluminal vesicles. This subpopulation behaves similarly to the general population of LAMP1-positive vesicles in terms of pH response to pharmacological stresses. In addition, RpH-ILV, which is trafficked to the lysosome via a different cytosolic motif than our previous ratiometric sensor, RpH-LAMP1, is well tolerated by the model organism Drosophila melanogaster, exhibits minimal plasma membrane fluorescence, and reveals sensitivity to the lysosomal damaging agent LLOMe, adding a valuable tool to our repertoire of lysosomal pH sensors.
Collapse
Affiliation(s)
- Izaak J. Cheetham-Wilkinson
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Bhavya Sivalingam
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Chloe Flitton
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Franziska Flottmann
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Luisa Vehling
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Maik Drechsler
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Marija Stojchevska
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrea Raimondi
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
- Università della Svizzera Italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland
| | - Achim Paululat
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück – CellNanOs, Osnabrück University, 49076 Osnabrück, Germany
| | - Florian Fröhlich
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück – CellNanOs, Osnabrück University, 49076 Osnabrück, Germany
| | - Laura E. Swan
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Massimiliano Stagi
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
11
|
Yamashita K, Shimane K, Muramoto T. Optogenetic control of cAMP oscillations reveals frequency-selective transcription factor dynamics in Dictyostelium. Development 2025; 152:dev204403. [PMID: 39775856 PMCID: PMC11829771 DOI: 10.1242/dev.204403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Oscillatory dynamics and their modulation are crucial for cellular decision-making; however, analysing these dynamics remains challenging. Here, we present a tool that combines the light-activated adenylate cyclase mPAC with the cAMP biosensor Pink Flamindo, enabling precise manipulation and real-time monitoring of cAMP oscillation frequencies in Dictyostelium. High-frequency modulation of cAMP oscillations induced cell aggregation and multicellular formation, even at low cell densities, such as a few dozen cells. At the population level, chemotactic aggregation is driven by modulated frequency signals. Additionally, modulation of cAMP frequency significantly reduced the amplitude of the shuttling behaviour of the transcription factor GtaC, demonstrating low-pass filter characteristics capable of converting subtle oscillation changes, such as from 6 min to 4 min, into gene expression. These findings enhance our understanding of frequency-selective cellular decoding and its role in cellular signalling and development.
Collapse
Affiliation(s)
- Kensuke Yamashita
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Kazuya Shimane
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Tetsuya Muramoto
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
12
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
13
|
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Nagy G, Leite W, Heller W, Stanley C, Perrone B, Kriwacki R. p14 ARF forms meso-scale assemblies upon phase separation with NPM1. Nat Commun 2024; 15:9531. [PMID: 39528457 PMCID: PMC11555371 DOI: 10.1038/s41467-024-53904-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14ARF). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF-NPM1 complex were elusive. Here we show that p14ARF assembles into a gel-like meso-scale network upon phase separation with NPM1. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal portion of p14ARF. These hydrophobic interactions promote phase separation with NPM1, enhance p14ARF nucleolar partitioning, restrict NPM1 diffusion within condensates and nucleoli, and reduce cellular proliferation. Our structural analysis provides insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1.
Collapse
Affiliation(s)
- Eric Gibbs
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Qi Miao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mylene Ferrolino
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aila Hassan
- Bruker Switzerland AG, Fällanden, Switzerland
| | - Aaron H Phillips
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Shondra Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gergely Nagy
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Wellington Leite
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - William Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Chris Stanley
- Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA.
| |
Collapse
|
14
|
Nguyen HN, Huynh U, Zastrow ML. Fluorescent protein-based Zn 2+ sensors reveal distinct responses of aerobic and anaerobic Escherichia coli cultures to excess Zn 2. J Biol Chem 2024; 300:107840. [PMID: 39357830 PMCID: PMC11550654 DOI: 10.1016/j.jbc.2024.107840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Zinc ions are required by all known organisms. Maintaining zinc homeostasis by preventing toxic overload while ensuring sufficient acquisition for cellular functions is crucial for survival and growth of bacteria. Bacteria, however, frequently encounter and must survive in various environments. During infection in host animals, for example, bacteria are exposed to acidic conditions in the stomach and anaerobic conditions in the intestines, but the effects of oxygen on zinc homeostasis in Escherichia coli have not been well-studied. Previously, we reported a flavin-binding fluorescent protein-based zinc sensor, CreiLOVN41C, which can respond to changes in labile Zn2+ levels in bacteria under both aerobic and anaerobic conditions. Here, we combined the use of CreiLOVN41C with established oxygen-dependent fluorescent protein-based sensors, inductively coupled plasma-mass spectrometry, and growth curves to evaluate how oxygen levels affect zinc uptake in E. coli. Inductively coupled plasma-mass spectrometry results showed that cells grown aerobically with added zinc acquired more zinc, but no additional zinc was accumulated when cells were grown anaerobically. Using oxygen-independent CreiLOVN41C and the oxygen-dependent ZapCY series of sensors, intracellular labile zinc was detected in E. coli grown with varied zinc under varied conditions. Although little to no endogenous zinc was detected by any sensor in E. coli cells grown with up to 2 mM added zinc, CreiLOVN41C revealed that when Zn2+ was added and detected by cells in real-time, anaerobic cells required more Zn2+ to similarly saturate the sensor. Overall, this work reveals that zinc uptake in E. coli is impacted by oxygen levels during cell growth.
Collapse
Affiliation(s)
- Hazel N Nguyen
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Uyen Huynh
- Department of Chemistry, University of Houston, Houston, Texas, United States
| | - Melissa L Zastrow
- Department of Chemistry, University of Houston, Houston, Texas, United States.
| |
Collapse
|
15
|
Das S, Thompson W, Papoutsakis ET. Engineered and hybrid human megakaryocytic extracellular vesicles for targeted non-viral cargo delivery to hematopoietic (blood) stem and progenitor cells. Front Bioeng Biotechnol 2024; 12:1435228. [PMID: 39386042 PMCID: PMC11461334 DOI: 10.3389/fbioe.2024.1435228] [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: 05/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Native and engineered extracellular vesicles generated from human megakaryocytes (huMkEVs) or from the human megakaryocytic cell line CHRF (CHEVs) interact with tropism delivering their cargo to both human and murine hematopoietic stem and progenitor cells (HSPCs). To develop non-viral delivery vectors to HSPCs based on MkEVs, we first confirmed, using NOD-scid IL2Rγnull (NSG™) mice, the targeting potential of the large EVs, enriched in microparticles (huMkMPs), chosen for their large cargo capacity. 24 h post intravenous infusion into NSG mice, huMkEVs induced a nearly 50% increase in murine platelet counts. PKH26-labeled huMkEVs or CHEVs localized to the HSPC-rich bone marrow preferentially interacting with murine HSPCs, thus confirming their receptor-mediated tropism for NSG HSPCs, and their potential to treat thromobocytopenias. We explored this tropism to functionally deliver synthetic cargo, notably plasmid DNA coding for a fluorescent reporter, to NSG HSPCs both in vitro and in vivo. We loaded huMkEVs with plasmid DNA either through electroporation or by generating hybrid particles with preloaded liposomes. Both methods facilitated successful functional targeted delivery of pDNA, as tissue weight-normalized fluorescence intensity of the expressed fluorescent reporter was significantly higher in bone marrow than other tissues. Furthermore, the fraction of fluorescent CD117+ HSPCs was nearly 19-fold higher than other cell types within the bone marrow 72-h following administration of the hybrid particles, further supporting that HSPC tropism is retained when using hybrid particles. These data demonstrate the potential of these EVs as a non-viral, HSPC-specific cargo vehicle for gene therapy applications to treat hematological diseases.
Collapse
Affiliation(s)
- Samik Das
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Will Thompson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
| | - Eleftherios Terry Papoutsakis
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, United States
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
16
|
Cook A, Kaydanov N, Ugarte-Uribe B, Boffi JC, Kamm GB, Prevedel R, Deo C. Chemigenetic Far-Red Labels and Ca 2+ Indicators Optimized for Photoacoustic Imaging. J Am Chem Soc 2024; 146:23963-23971. [PMID: 39158696 PMCID: PMC11363013 DOI: 10.1021/jacs.4c07080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Photoacoustic imaging is an emerging modality with significant promise for biomedical applications such as neuroimaging, owing to its capability to capture large fields of view deep inside complex scattering tissue. However, widespread adoption of this technique has been hindered by a lack of suitable molecular reporters for this modality. In this work, we introduce chemigenetic labels and calcium sensors specifically tailored for photoacoustic imaging, using a combination of synthetic dyes and HaloTag-based self-labeling proteins. We rationally design and engineer far-red "acoustogenic" dyes, showing high photoacoustic turn-ons upon binding to HaloTag, and develop a suite of tunable calcium indicators based on these scaffolds. These first-generation photoacoustic reporters show excellent performance in tissue-mimicking phantoms, with the best variants outperforming existing sensors in terms of signal intensity, sensitivity, and photostability. We demonstrate the application of these ligands for labeling HaloTag-expressing neurons in mouse brain tissue, producing strong, specifically targeted photoacoustic signal, and provide a first example of in vivo labeling with these chemigenetic photoacoustic probes. Together, this work establishes a new approach for the design of photoacoustic reporters, paving the way toward deep tissue functional imaging.
Collapse
Affiliation(s)
- Alexander Cook
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Nikita Kaydanov
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Begoña Ugarte-Uribe
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Juan Carlos Boffi
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Gretel B. Kamm
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Robert Prevedel
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Claire Deo
- European Molecular Biology
Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| |
Collapse
|
17
|
Wong VC, Houlihan PR, Liu H, Walpita D, DeSantis MC, Liu Z, O'Shea EK. Plasticity-induced actin polymerization in the dendritic shaft regulates intracellular AMPA receptor trafficking. eLife 2024; 13:e80622. [PMID: 39146380 PMCID: PMC11326776 DOI: 10.7554/elife.80622] [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: 05/27/2022] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
AMPA-type receptors (AMPARs) are rapidly inserted into synapses undergoing plasticity to increase synaptic transmission, but it is not fully understood if and how AMPAR-containing vesicles are selectively trafficked to these synapses. Here, we developed a strategy to label AMPAR GluA1 subunits expressed from their endogenous loci in cultured rat hippocampal neurons and characterized the motion of GluA1-containing vesicles using single-particle tracking and mathematical modeling. We find that GluA1-containing vesicles are confined and concentrated near sites of stimulation-induced structural plasticity. We show that confinement is mediated by actin polymerization, which hinders the active transport of GluA1-containing vesicles along the length of the dendritic shaft by modulating the rheological properties of the cytoplasm. Actin polymerization also facilitates myosin-mediated transport of GluA1-containing vesicles to exocytic sites. We conclude that neurons utilize F-actin to increase vesicular GluA1 reservoirs and promote exocytosis proximal to the sites of synaptic activity.
Collapse
Affiliation(s)
- Victor C Wong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Patrick R Houlihan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Hui Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Deepika Walpita
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael C DeSantis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Erin K O'Shea
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
18
|
Szelenyi ER, Navarrete JS, Murry AD, Zhang Y, Girven KS, Kuo L, Cline MM, Bernstein MX, Burdyniuk M, Bowler B, Goodwin NL, Juarez B, Zweifel LS, Golden SA. An arginine-rich nuclear localization signal (ArgiNLS) strategy for streamlined image segmentation of single cells. Proc Natl Acad Sci U S A 2024; 121:e2320250121. [PMID: 39074275 PMCID: PMC11317604 DOI: 10.1073/pnas.2320250121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/25/2024] [Indexed: 07/31/2024] Open
Abstract
High-throughput volumetric fluorescent microscopy pipelines can spatially integrate whole-brain structure and function at the foundational level of single cells. However, conventional fluorescent protein (FP) modifications used to discriminate single cells possess limited efficacy or are detrimental to cellular health. Here, we introduce a synthetic and nondeleterious nuclear localization signal (NLS) tag strategy, called "Arginine-rich NLS" (ArgiNLS), that optimizes genetic labeling and downstream image segmentation of single cells by restricting FP localization near-exclusively in the nucleus through a poly-arginine mechanism. A single N-terminal ArgiNLS tag provides modular nuclear restriction consistently across spectrally separate FP variants. ArgiNLS performance in vivo displays functional conservation across major cortical cell classes and in response to both local and systemic brain-wide AAV administration. Crucially, the high signal-to-noise ratio afforded by ArgiNLS enhances machine learning-automated segmentation of single cells due to rapid classifier training and enrichment of labeled cell detection within 2D brain sections or 3D volumetric whole-brain image datasets, derived from both staining-amplified and native signal. This genetic strategy provides a simple and flexible basis for precise image segmentation of genetically labeled single cells at scale and paired with behavioral procedures.
Collapse
Affiliation(s)
- Eric R. Szelenyi
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Jovana S. Navarrete
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | - Alexandria D. Murry
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Yizhe Zhang
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Kasey S. Girven
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195
| | - Lauren Kuo
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Undergraduate Program in Biochemistry, University of Washington, Seattle, WA98195
| | - Marcella M. Cline
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | - Mollie X. Bernstein
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | | | - Bryce Bowler
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Nastacia L. Goodwin
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
- Graduate Program in Neuroscience, University of Washington, Seattle, WA98195
| | - Barbara Juarez
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Pharmacology, University of Washington, Seattle, WA98195
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA98195
| | - Larry S. Zweifel
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Pharmacology, University of Washington, Seattle, WA98195
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA98195
| | - Sam A. Golden
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA98195
- Department of Biological Structure, University of Washington, Seattle, WA98195
| |
Collapse
|
19
|
Ajiki M, Yoshikawa M, Miyazaki T, Kawasaki A, Aoki K, Nakatsu F, Tsukiji S. ORP9-PH domain-based fluorescent reporters for visualizing phosphatidylinositol 4-phosphate dynamics in living cells. RSC Chem Biol 2024; 5:544-555. [PMID: 38846081 PMCID: PMC11151866 DOI: 10.1039/d3cb00232b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 06/09/2024] Open
Abstract
Fluorescent reporters that visualize phosphatidylinositol 4-phosphate (PI4P) in living cells are indispensable to elucidate the roles of this fundamental lipid in cell physiology. However, currently available PI4P reporters have limitations, such as Golgi-biased localization and low detection sensitivity. Here, we present a series of fluorescent PI4P reporters based on the pleckstrin homology (PH) domain of oxysterol-binding protein-related protein 9 (ORP9). We show that the green fluorescent protein AcGFP1-tagged ORP9-PH domain can be used as a fluorescent PI4P reporter to detect cellular PI4P across its wide distribution at multiple cellular locations, including the plasma membrane (PM), Golgi, endosomes, and lysosomes with high specificity and contrast. We also developed blue, red, and near-infrared fluorescent PI4P reporters suitable for multicolor fluorescence imaging experiments. Finally, we demonstrate the utility of the ORP9-PH domain-based reporter to visualize dynamic changes in the PI4P distribution and level in living cells upon synthetic ER-PM membrane contact manipulation and GPCR stimulation. This work offers a new set of genetically encoded fluorescent PI4P reporters that are practically useful for the study of PI4P biology.
Collapse
Affiliation(s)
- Moeka Ajiki
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Masaru Yoshikawa
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Tomoki Miyazaki
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University 1-757 Asahimachi, Chuo-ku Niigata 951-8510 Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
- Department of Basic Biology, Faculty of Life Science, SOKENDAI (The Graduate University for Advanced Studies) 5-1 Higashiyama, Myodaiji-cho Okazaki Aichi 444-8787 Japan
| | - Fubito Nakatsu
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University 1-757 Asahimachi, Chuo-ku Niigata 951-8510 Japan
| | - Shinya Tsukiji
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology Gokiso-cho, Showa-ku Nagoya 466-8555 Japan
| |
Collapse
|
20
|
Reicher A, Reiniš J, Ciobanu M, Růžička P, Malik M, Siklos M, Kartysh V, Tomek T, Koren A, Rendeiro AF, Kubicek S. Pooled multicolour tagging for visualizing subcellular protein dynamics. Nat Cell Biol 2024; 26:745-756. [PMID: 38641660 PMCID: PMC11098740 DOI: 10.1038/s41556-024-01407-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/18/2024] [Indexed: 04/21/2024]
Abstract
Imaging-based methods are widely used for studying the subcellular localization of proteins in living cells. While routine for individual proteins, global monitoring of protein dynamics following perturbation typically relies on arrayed panels of fluorescently tagged cell lines, limiting throughput and scalability. Here, we describe a strategy that combines high-throughput microscopy, computer vision and machine learning to detect perturbation-induced changes in multicolour tagged visual proteomics cell (vpCell) pools. We use genome-wide and cancer-focused intron-targeting sgRNA libraries to generate vpCell pools and a large, arrayed collection of clones each expressing two different endogenously tagged fluorescent proteins. Individual clones can be identified in vpCell pools by image analysis using the localization patterns and expression level of the tagged proteins as visual barcodes, enabling simultaneous live-cell monitoring of large sets of proteins. To demonstrate broad applicability and scale, we test the effects of antiproliferative compounds on a pool with cancer-related proteins, on which we identify widespread protein localization changes and new inhibitors of the nuclear import/export machinery. The time-resolved characterization of changes in subcellular localization and abundance of proteins upon perturbation in a pooled format highlights the power of the vpCell approach for drug discovery and mechanism-of-action studies.
Collapse
Affiliation(s)
- Andreas Reicher
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jiří Reiniš
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Maria Ciobanu
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Pavel Růžička
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Monika Malik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Marton Siklos
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Victoria Kartysh
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tatjana Tomek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Anna Koren
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André F Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
21
|
Hu X, Xu Y, Yi J, Wang C, Zhu Z, Yue T, Zhang H, Wang X, Wu F, Xue L, Bai L, Liu H, Chen Q. Using Protein Design and Directed Evolution to Monomerize a Bright Near-Infrared Fluorescent Protein. ACS Synth Biol 2024; 13:1177-1190. [PMID: 38552148 DOI: 10.1021/acssynbio.3c00643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
The small ultrared fluorescent protein (smURFP) is a bright near-infrared (NIR) fluorescent protein (FP) that forms a dimer and binds its fluorescence chromophore, biliverdin, at its dimer interface. To engineer a monomeric NIR FP based on smURFP potentially more suitable for bioimaging, we employed protein design to extend the protein backbone with a new segment of two helices that shield the original dimer interface while covering the biliverdin binding pocket in place of the second chain in the original dimer. We experimentally characterized 13 designs and obtained a monomeric protein with a weak fluorescence. We enhanced the fluorescence of this designed protein through two rounds of directed evolution and obtained designed monomeric smURFP (DMsmURFP), a bright, stable, and monomeric NIR FP with a molecular weight of 19.6 kDa. We determined the crystal structures of DMsmURFP both in the apo state and in complex with biliverdin, which confirmed the designed structure. The use of DMsmURFP in in vivo imaging of mammalian systems was demonstrated. The backbone design-based strategy used here can also be applied to monomerize other naturally multimeric proteins with intersubunit functional sites.
Collapse
Affiliation(s)
- Xiuhong Hu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yang Xu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Junxi Yi
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chenchen Wang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhongliang Zhu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ting Yue
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiyan Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xinyu Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Fan Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Lin Xue
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Li Bai
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Haiyan Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China
- School of Data Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Quan Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Interdisciplinary Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
22
|
Lu K, Wazawa T, Matsuda T, Shcherbakova DM, Verkhusha VV, Nagai T. Near-infrared PAINT localization microscopy via chromophore replenishment of phytochrome-derived fluorescent tag. Commun Biol 2024; 7:473. [PMID: 38637683 PMCID: PMC11026395 DOI: 10.1038/s42003-024-06169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
Bacterial phytochromes are attractive molecular templates for engineering fluorescent proteins (FPs) because their near-infrared (NIR) emission significantly extends the spectral coverage of GFP-like FPs. Existing phytochrome-based FPs covalently bind heme-derived tetrapyrrole chromophores and exhibit constitutive fluorescence. Here we introduce Rep-miRFP, an NIR imaging probe derived from bacterial phytochrome, which interacts non-covalently and reversibly with biliverdin chromophore. In Rep-miRFP, the photobleached non-covalent adduct can be replenished with fresh biliverdin, restoring fluorescence. By exploiting this chromophore renewal capability, we demonstrate NIR PAINT nanoscopy in mammalian cells using Rep-miRFP.
Collapse
Affiliation(s)
- Kai Lu
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Tetsuichi Wazawa
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Tomoki Matsuda
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Daria M Shcherbakova
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - 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
| | - Takeharu Nagai
- SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan.
| |
Collapse
|
23
|
Jiang XX, Hou YN, Lu LW, Zhao KH. Monomeric Far-red and Near-infrared Fluorescent Biliproteins of Ultrahigh Brightness. Chembiochem 2024:e202400068. [PMID: 38623786 DOI: 10.1002/cbic.202400068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 04/17/2024]
Abstract
Far-red and near-infrared fluorescent proteins have regions of maximum transmission in most tissues and can be widely used as fluorescent biomarkers. We report that fluorescent phycobiliproteins originating from the phycobilisome core subunit ApcF2 can covalently bind biliverdin, named BDFPs. To further improve BDFPs, we conducted a series of studies. Firstly, we mutated K53Q and T144A of BDFPs to increase their effective brightness up to 190 % in vivo. Secondly, by homochromatic tandem fusion of high-brightness BDFPs to achieve monomerization, which increases the effective brightness by up to 180 % in vivo, and can effectively improve the labeling effect. By combining the above two approaches, the brightness of the tandem BDFPs was much improved compared with that of the previously reported fluorescent proteins in a similar spectral range. The tandem BDFPs were expressed stably while maintaining fluorescence in mammalian cells and Caenorhabditis elegans. They were also photostable and resistant to high temperature, low pH, and chemical denaturation. The tandem BDFPs advantages were proved in applications as biomarkers for imaging in super-resolution microscopy.
Collapse
Affiliation(s)
- Xiang-Xiang Jiang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Ya-Nan Hou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Li-Wen Lu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Kai-Hong Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| |
Collapse
|
24
|
Xu J, Zhu N, Du Y, Han T, Zheng X, Li J, Zhu S. Biomimetic NIR-II fluorescent proteins created from chemogenic protein-seeking dyes for multicolor deep-tissue bioimaging. Nat Commun 2024; 15:2845. [PMID: 38565859 PMCID: PMC10987503 DOI: 10.1038/s41467-024-47063-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Near-infrared-I/II fluorescent proteins (NIR-I/II FPs) are crucial for in vivo imaging, yet the current NIR-I/II FPs face challenges including scarcity, the requirement for chromophore maturation, and limited emission wavelengths (typically < 800 nm). Here, we utilize synthetic protein-seeking NIR-II dyes as chromophores, which covalently bind to tag proteins (e.g., human serum albumin, HSA) through a site-specific nucleophilic substitution reaction, thereby creating proof-of-concept biomimetic NIR-II FPs. This chemogenic protein-seeking strategy can be accomplished under gentle physiological conditions without catalysis. Proteomics analysis identifies specific binding site (Cys 477 on DIII). NIR-II FPs significantly enhance chromophore brightness and photostability, while improving biocompatibility, allowing for high-performance NIR-II lymphography and angiography. This strategy is universal and applicable in creating a wide range of spectrally separated NIR-I/II FPs for real-time visualization of multiple biological events. Overall, this straightforward biomimetic approach holds the potential to transform fluorescent protein-based bioimaging and enables in-situ albumin targeting to create NIR-I/II FPs for deep-tissue imaging in live organisms.
Collapse
Affiliation(s)
- Jiajun Xu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding, 071002, P.R. China
| | - Ningning Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Yijing Du
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Tianyang Han
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Xue Zheng
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China
| | - Jia Li
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Changchun, 130021, P.R. China.
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, P.R. China.
| |
Collapse
|
25
|
Song C, Matlashov ME, Shcherbakova DM, Antic SD, Verkhusha VV, Knöpfel T. Characterization of two near-infrared genetically encoded voltage indicators. NEUROPHOTONICS 2024; 11:024201. [PMID: 38090225 PMCID: PMC10712888 DOI: 10.1117/1.nph.11.2.024201] [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: 07/25/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024]
Abstract
Significance Efforts starting more than 20 years ago led to increasingly well performing genetically encoded voltage indicators (GEVIs) for optical imaging at wavelengths < 600 nm . Although optical imaging in the > 600 nm wavelength range has many advantages over shorter wavelength approaches for mesoscopic in vivo monitoring of neuronal activity in the mammalian brain, the availability and evaluation of well performing near-infrared GEVIs are still limited. Aim Here, we characterized two recent near-infrared GEVIs, Archon1 and nirButterfly, to support interested tool users in selecting a suitable near-infrared GEVI for their specific research question requirements. Approach We characterized side-by-side the brightness, sensitivity, and kinetics of both near-infrared GEVIs in a setting focused on population imaging. Results We found that nirButterfly shows seven-fold higher brightness than Archon1 under the same conditions and faster kinetics than Archon1 for population imaging without cellular resolution. But Archon1 showed larger signals than nirButterfly. Conclusions Neither GEVI characterized here surpasses in all three key parameters (brightness, kinetics, and sensitivity), so there is no unequivocal preference for one of the two. Our side-by-side characterization presented here provides new information for future in vitro and ex vivo experimental designs.
Collapse
Affiliation(s)
- Chenchen Song
- Imperial College, Laboratory for Neuronal Circuit Dynamics, London, United Kingdom
- Nanyang Technological University, Singapore
| | - Mikhail E. Matlashov
- Albert Einstein College of Medicine, Gruss-Lipper Biophotonics Center, Department of Genetics, Bronx, New York, United States
| | - Daria M. Shcherbakova
- Albert Einstein College of Medicine, Gruss-Lipper Biophotonics Center, Department of Genetics, Bronx, New York, United States
| | - Srdjan D. Antic
- Institute for Systems Genomics, UConn Health, Department of Neuroscience, Farmington, Connecticut, United States
| | - Vladislav V. Verkhusha
- Albert Einstein College of Medicine, Gruss-Lipper Biophotonics Center, Department of Genetics, Bronx, New York, United States
- University of Helsinki, Medicum, Faculty of Medicine, Helsinki, Finland
| | - Thomas Knöpfel
- Imperial College, Laboratory for Neuronal Circuit Dynamics, London, United Kingdom
- Hong Kong Baptist University, Laboratory for Neuronal Circuit Dynamics, Hong Kong, China
| |
Collapse
|
26
|
Pulfer A, Pizzagalli DU, Gagliardi PA, Hinderling L, Lopez P, Zayats R, Carrillo-Barberà P, Antonello P, Palomino-Segura M, Grädel B, Nicolai M, Giusti A, Thelen M, Gambardella LM, Murooka TT, Pertz O, Krause R, Gonzalez SF. Transformer-based spatial-temporal detection of apoptotic cell death in live-cell imaging. eLife 2024; 12:RP90502. [PMID: 38497754 PMCID: PMC10948145 DOI: 10.7554/elife.90502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Intravital microscopy has revolutionized live-cell imaging by allowing the study of spatial-temporal cell dynamics in living animals. However, the complexity of the data generated by this technology has limited the development of effective computational tools to identify and quantify cell processes. Amongst them, apoptosis is a crucial form of regulated cell death involved in tissue homeostasis and host defense. Live-cell imaging enabled the study of apoptosis at the cellular level, enhancing our understanding of its spatial-temporal regulation. However, at present, no computational method can deliver robust detection of apoptosis in microscopy timelapses. To overcome this limitation, we developed ADeS, a deep learning-based apoptosis detection system that employs the principle of activity recognition. We trained ADeS on extensive datasets containing more than 10,000 apoptotic instances collected both in vitro and in vivo, achieving a classification accuracy above 98% and outperforming state-of-the-art solutions. ADeS is the first method capable of detecting the location and duration of multiple apoptotic events in full microscopy timelapses, surpassing human performance in the same task. We demonstrated the effectiveness and robustness of ADeS across various imaging modalities, cell types, and staining techniques. Finally, we employed ADeS to quantify cell survival in vitro and tissue damage in mice, demonstrating its potential application in toxicity assays, treatment evaluation, and inflammatory dynamics. Our findings suggest that ADeS is a valuable tool for the accurate detection and quantification of apoptosis in live-cell imaging and, in particular, intravital microscopy data, providing insights into the complex spatial-temporal regulation of this process.
Collapse
Affiliation(s)
- Alain Pulfer
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Department of Information Technology and Electrical Engineering, ETH ZurichZürichSwitzerland
| | - Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Euler Institute, USILuganoSwitzerland
| | | | | | | | | | - Pau Carrillo-Barberà
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universitat de ValènciaValenciaSpain
| | - Paola Antonello
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | - Benjamin Grädel
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | - Alessandro Giusti
- Dalle Molle Institute for Artificial Intelligence, IDSIALuganoSwitzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USILuganoSwitzerland
| | | | | | - Olivier Pertz
- Institute of Cell Biology, University of BernBernSwitzerland
| | | | | |
Collapse
|
27
|
Popgeorgiev N, Gil C, Berthenet K, Bertolin G, Ichim G. Shedding light on mitochondrial outer-membrane permeabilization and membrane potential: State of the art methods and biosensors. Semin Cell Dev Biol 2024; 156:58-65. [PMID: 37438211 DOI: 10.1016/j.semcdb.2023.07.003] [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: 04/02/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 07/14/2023]
Abstract
Membrane structural integrity is essential for optimal mitochondrial function. These organelles produce the energy needed for all vital processes, provided their outer and inner membranes are intact. This prevents the release of mitochondrial apoptogenic factors into the cytosol and ensures intact mitochondrial membrane potential (ΔΨm) to sustain ATP production. Cell death by apoptosis is generally triggered by outer mitochondrial membrane permeabilization (MOMP), tightly coupled with loss of ΔΨ m. As these two processes are essential for both mitochondrial function and cell death, researchers have devised various techniques to assess them. Here, we discuss current methods and biosensors available for detecting MOMP and measuring ΔΨ m, focusing on their advantages and limitations and discuss what new imaging tools are needed to improve our knowledge of mitochondrial function.
Collapse
Affiliation(s)
- Nikolay Popgeorgiev
- Cancer Cell Death laboratory, part of LabEX DEVweCAN, Cancer Initiation and Tumoral Cell Identity Department, CRCL, U1052 INSERM, UMR CNRS 5286, Centre Léon Bérard, Université Lyon I, Institut Convergence PLAsCAN Lyon, France; Institut Universitaire de France (IUF), Paris, France
| | - Clara Gil
- Cancer Cell Death laboratory, part of LabEX DEVweCAN, Cancer Initiation and Tumoral Cell Identity Department, CRCL, U1052 INSERM, UMR CNRS 5286, Centre Léon Bérard, Université Lyon I, Institut Convergence PLAsCAN Lyon, France
| | - Kevin Berthenet
- Cancer Cell Death laboratory, part of LabEX DEVweCAN, Cancer Initiation and Tumoral Cell Identity Department, CRCL, U1052 INSERM, UMR CNRS 5286, Centre Léon Bérard, Université Lyon I, Institut Convergence PLAsCAN Lyon, France
| | - Giulia Bertolin
- CNRS, Univ Rennes, IGDR (Institute of Genetics and Development of Rennes), Rennes, France.
| | - Gabriel Ichim
- Cancer Cell Death laboratory, part of LabEX DEVweCAN, Cancer Initiation and Tumoral Cell Identity Department, CRCL, U1052 INSERM, UMR CNRS 5286, Centre Léon Bérard, Université Lyon I, Institut Convergence PLAsCAN Lyon, France.
| |
Collapse
|
28
|
Hu Y, Li CY, Lu Q, Kuang Y. Multiplex miRNA reporting platform for real-time profiling of living cells. Cell Chem Biol 2024; 31:150-162.e7. [PMID: 38035883 DOI: 10.1016/j.chembiol.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/15/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
Accurately characterizing cell types within complex cell structures provides invaluable information for comprehending the cellular status during biological processes. In this study, we have developed an miRNA-switch cocktail platform capable of reporting and tracking the activities of multiple miRNAs (microRNAs) at the single-cell level, while minimizing disruption to the cell culture. Drawing on the principles of traditional miRNA-sensing mRNA switches, our platform incorporates subcellular tags and employs intelligent engineering to segment three subcellular regions using two fluorescent proteins. These designs enable the quantification of multiple miRNAs within the same cell. Through our experiments, we have demonstrated the platform's ability to track marker miRNA levels during cell differentiation and provide spatial information of heterogeneity on outlier cells exhibiting extreme miRNA levels. Importantly, this platform offers real-time and in situ miRNA reporting, allowing for multidimensional evaluation of cell profile and paving the way for a comprehensive understanding of cellular events during biological processes.
Collapse
Affiliation(s)
- Yaxin Hu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Qiuyu Lu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
29
|
Gebhardt CM, Niopek D. Anti-CRISPR Proteins and Their Application to Control CRISPR Effectors in Mammalian Systems. Methods Mol Biol 2024; 2774:205-231. [PMID: 38441767 DOI: 10.1007/978-1-0716-3718-0_14] [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: 03/07/2024]
Abstract
CRISPR-Cas effectors are powerful tools for genome and transcriptome targeting and editing. Naturally, these protein-RNA complexes are part of the microbial innate immune system, which emerged from the evolutionary arms race between microbes and phages. This coevolution has also given rise to so-called anti-CRISPR (Acr) proteins that counteract the CRISPR-Cas adaptive immunity. Acrs constitutively block cognate CRISPR-Cas effectors, e.g., by interfering with guide RNA binding, target DNA/RNA recognition, or target cleavage. In addition to their important role in microbiology and evolution, Acrs have recently gained particular attention for being useful tools and switches to regulate or fine-tune the activity of CRISPR-Cas effectors. Due to their commonly small size, high inhibition potency, and structural and mechanistic versatility, Acrs offer a wide range of potential applications for controlling CRISPR effectors in heterologous systems, including mammalian cells.Here, we review the diverse applications of Acrs in mammalian cells and organisms and discuss the underlying engineering strategies. These applications include (i) persistent blockage of CRISPR-Cas function to create write-protected cells, (ii) reduction of CRISPR-Cas off-target editing, (iii) focusing CRISPR-Cas activity to specific cell types and tissues, (iv) spatiotemporal control of CRISPR effectors based on engineered, opto-, or chemogenetic Acrs, and (v) the use of Acrs for selective binding and detection of CRISPR-Cas effectors in complex samples. We will also highlight potential future applications of Acrs in a biomedical context and point out present challenges that need to be overcome on the way.
Collapse
Affiliation(s)
- Carolin Maja Gebhardt
- Centre for Synthetic Biology, Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - Dominik Niopek
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
30
|
Jeong S, Koh D, Gwak E, Srambickal CV, Seo D, Widengren J, Lee JC. Pushing the Resolution Limit of Stimulated Emission Depletion Optical Nanoscopy. Int J Mol Sci 2023; 25:26. [PMID: 38203197 PMCID: PMC10779414 DOI: 10.3390/ijms25010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Optical nanoscopy, also known as super-resolution optical microscopy, has provided scientists with the means to surpass the diffraction limit of light microscopy and attain new insights into nanoscopic structures and processes that were previously inaccessible. In recent decades, numerous studies have endeavored to enhance super-resolution microscopy in terms of its spatial (lateral) resolution, axial resolution, and temporal resolution. In this review, we discuss recent efforts to push the resolution limit of stimulated emission depletion (STED) optical nanoscopy across multiple dimensions, including lateral resolution, axial resolution, temporal resolution, and labeling precision. We introduce promising techniques and methodologies building on the STED concept that have emerged in the field, such as MINSTED, isotropic STED, and event-triggered STED, and evaluate their respective strengths and limitations. Moreover, we discuss trade-off relationships that exist in far-field optical microscopy and how they come about in STED optical nanoscopy. By examining the latest developments addressing these aspects, we aim to provide an updated overview of the current state of STED nanoscopy and its potential for future research.
Collapse
Affiliation(s)
- Sejoo Jeong
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Dongbin Koh
- School of Undergraduate Studies, DGIST, Daegu 42988, Republic of Korea
| | - Eunha Gwak
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Chinmaya V. Srambickal
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu 42988, Republic of Korea
| | - Jerker Widengren
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Jong-Chan Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
- New Biology Research Center, DGIST, Daegu 42988, Republic of Korea
| |
Collapse
|
31
|
Pennacchietti F, Alvelid J, Morales RA, Damenti M, Ollech D, Oliinyk OS, Shcherbakova DM, Villablanca EJ, Verkhusha VV, Testa I. Blue-shift photoconversion of near-infrared fluorescent proteins for labeling and tracking in living cells and organisms. Nat Commun 2023; 14:8402. [PMID: 38114484 PMCID: PMC10730883 DOI: 10.1038/s41467-023-44054-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Photolabeling of intracellular molecules is an invaluable approach to studying various dynamic processes in living cells with high spatiotemporal precision. Among fluorescent proteins, photoconvertible mechanisms and their products are in the visible spectrum (400-650 nm), limiting their in vivo and multiplexed applications. Here we report the phenomenon of near-infrared to far-red photoconversion in the miRFP family of near infrared fluorescent proteins engineered from bacterial phytochromes. This photoconversion is induced by near-infrared light through a non-linear process, further allowing optical sectioning. Photoconverted miRFP species emit fluorescence at 650 nm enabling photolabeling entirely performed in the near-infrared range. We use miRFPs as photoconvertible fluorescent probes to track organelles in live cells and in vivo, both with conventional and super-resolution microscopy. The spectral properties of miRFPs complement those of GFP-like photoconvertible proteins, allowing strategies for photoconversion and spectral multiplexed applications.
Collapse
Affiliation(s)
- Francesca Pennacchietti
- Department of Applied Physics and SciLifeLab, KTH Royal Institute of Technology, Stockholm, 17165, Sweden.
| | - Jonatan Alvelid
- Department of Applied Physics and SciLifeLab, KTH Royal Institute of Technology, Stockholm, 17165, Sweden
| | - Rodrigo A Morales
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, 17176, Sweden
- Center for Molecular Medicine (CMM), Stockholm, 17176, Sweden
| | - Martina Damenti
- Department of Applied Physics and SciLifeLab, KTH Royal Institute of Technology, Stockholm, 17165, Sweden
| | - Dirk Ollech
- Department of Applied Physics and SciLifeLab, KTH Royal Institute of Technology, Stockholm, 17165, Sweden
| | | | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, 17176, Sweden
- Center for Molecular Medicine (CMM), Stockholm, 17176, Sweden
| | - Vladislav V Verkhusha
- Medicum, University of Helsinki, Helsinki, 00290, Finland
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ilaria Testa
- Department of Applied Physics and SciLifeLab, KTH Royal Institute of Technology, Stockholm, 17165, Sweden.
| |
Collapse
|
32
|
Szelenyi ER, Navarrete JS, Murry AD, Zhang Y, Girven KS, Kuo L, Cline MM, Bernstein MX, Burdyniuk M, Bowler B, Goodwin NL, Juarez B, Zweifel LS, Golden SA. An arginine-rich nuclear localization signal (ArgiNLS) strategy for streamlined image segmentation of single-cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568319. [PMID: 38045271 PMCID: PMC10690249 DOI: 10.1101/2023.11.22.568319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
High-throughput volumetric fluorescent microscopy pipelines can spatially integrate whole-brain structure and function at the foundational level of single-cells. However, conventional fluorescent protein (FP) modifications used to discriminate single-cells possess limited efficacy or are detrimental to cellular health. Here, we introduce a synthetic and non-deleterious nuclear localization signal (NLS) tag strategy, called 'Arginine-rich NLS' (ArgiNLS), that optimizes genetic labeling and downstream image segmentation of single-cells by restricting FP localization near-exclusively in the nucleus through a poly-arginine mechanism. A single N-terminal ArgiNLS tag provides modular nuclear restriction consistently across spectrally separate FP variants. ArgiNLS performance in vivo displays functional conservation across major cortical cell classes, and in response to both local and systemic brain wide AAV administration. Crucially, the high signal-to-noise ratio afforded by ArgiNLS enhances ML-automated segmentation of single-cells due to rapid classifier training and enrichment of labeled cell detection within 2D brain sections or 3D volumetric whole-brain image datasets, derived from both staining-amplified and native signal. This genetic strategy provides a simple and flexible basis for precise image segmentation of genetically labeled single-cells at scale and paired with behavioral procedures.
Collapse
Affiliation(s)
- Eric R. Szelenyi
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
| | - Jovana S. Navarrete
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
- University of Washington, Graduate Program in Neuroscience, Seattle, WA, USA
| | - Alexandria D. Murry
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
| | - Yizhe Zhang
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
| | - Kasey S. Girven
- University of Washington, Department of Anesthesiology and Pain Medicine
| | - Lauren Kuo
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington Undergraduate Program in Biochemistry
- Allen Institute for Cell Science, Seattle, WA, USA
| | - Marcella M. Cline
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Pharmacology, Seattle, WA, USA
- Cajal Neuroscience, Seattle, WA, USA
| | - Mollie X. Bernstein
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Pharmacology, Seattle, WA, USA
| | | | - Bryce Bowler
- University of Washington, Department of Biological Structure, Seattle, WA, USA
| | - Nastacia L. Goodwin
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
- University of Washington, Graduate Program in Neuroscience, Seattle, WA, USA
| | - Barbara Juarez
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Psychiatry and Behavioral Sciences, Seattle, WA, USA
- University of Washington, Department of Pharmacology, Seattle, WA, USA
- University of Maryland School of Medicine, Department of Neurobiology, Baltimore, MD, USA
| | - Larry S. Zweifel
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Psychiatry and Behavioral Sciences, Seattle, WA, USA
- University of Washington, Department of Pharmacology, Seattle, WA, USA
| | - Sam A. Golden
- University of Washington Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), Seattle, WA, USA
- University of Washington, Department of Biological Structure, Seattle, WA, USA
| |
Collapse
|
33
|
Zhang G, Dong M, Yao X, Xia Y, Yu H, Zhou Y, Lian C, Zhang Y, Cui Y. Advancing breast cancer diagnosis with a near-infrared fluorescence imaging smart sensor for estrogen/progesterone receptor detection. Sci Rep 2023; 13:21086. [PMID: 38030755 PMCID: PMC10687265 DOI: 10.1038/s41598-023-48556-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023] Open
Abstract
Molecular-genetic imaging has greatly advanced clinical diagnosis and prognosis monitoring. However, the specific visualization of intracellular proteins such as estrogen receptor (ER) and progesterone receptor (PR) remains an elusive goal. Here, we highlight a novel method for selectively detecting ER/PR positive tumors using genetically engineered responsive elements. Our study demonstrates that the double responsive elements of ER/PR exhibit the most sensitivity to the steroid receptors in breast cancers. By utilizing a cationic polymer vector, we constructed a responsive element-fluorescence protein system that can selectively image ER/PR positive breast cancers in murine models under a near-infrared laser. This non-invasive imaging achieved high-resolution detection without death or serious anaphylactic activity in the animals. Our findings suggest that the reporter system consisting of steroid receptor response elements and near-infrared proteins provides a practical system for identifying biomarkers and advancing cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Gong Zhang
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Min Dong
- Department of Comparative Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China
| | - Xiulei Yao
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Yuke Xia
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Han Yu
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Yu Zhou
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Chao Lian
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China
| | - Yunlei Zhang
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China.
- The Key Laboratory of Clinical and Medical Engineering, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211100, China.
| | - Yiyao Cui
- Department of Thyroid and Breast Surgery, Department of Ultrasound, Central Laboratory, Translational Medicine Research Center, The Affiliated JiangNing Hospital of NanJing Medical University, Nanjing, 211100, China.
| |
Collapse
|
34
|
Lyons AC, Mehta S, Zhang J. Fluorescent biosensors illuminate the spatial regulation of cell signaling across scales. Biochem J 2023; 480:1693-1717. [PMID: 37903110 PMCID: PMC10657186 DOI: 10.1042/bcj20220223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/01/2023]
Abstract
As cell signaling research has advanced, it has become clearer that signal transduction has complex spatiotemporal regulation that goes beyond foundational linear transduction models. Several technologies have enabled these discoveries, including fluorescent biosensors designed to report live biochemical signaling events. As genetically encoded and live-cell compatible tools, fluorescent biosensors are well suited to address diverse cell signaling questions across different spatial scales of regulation. In this review, methods of examining spatial signaling regulation and the design of fluorescent biosensors are introduced. Then, recent biosensor developments that illuminate the importance of spatial regulation in cell signaling are highlighted at several scales, including membranes and organelles, molecular assemblies, and cell/tissue heterogeneity. In closing, perspectives on how fluorescent biosensors will continue enhancing cell signaling research are discussed.
Collapse
Affiliation(s)
- Anne C. Lyons
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
35
|
Jensen GC, Janis MK, Jara J, Abbasi N, Zastrow ML. Zinc-Induced Fluorescence Turn-On in Native and Mutant Phycoerythrobilin-Binding Orange Fluorescent Proteins. Biochemistry 2023; 62:2828-2840. [PMID: 37699411 PMCID: PMC11057272 DOI: 10.1021/acs.biochem.3c00183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Cyanobacteriochrome (CBCR)-derived fluorescent proteins are a class of reporters that can bind bilin cofactors and fluoresce across the ultraviolet to the near-infrared spectrum. Derived from phytochrome-related photoreceptor proteins in cyanobacteria, many of these proteins use a single small GAF domain to autocatalytically bind a bilin and fluoresce. The second GAF domain of All1280 (All1280g2) from Nostoc sp. PCC7120 is a DXCF motif-containing protein that exhibits blue-light-responsive photochemistry when bound to its native cofactor, phycocyanobilin. All1280g2 can also bind non-photoswitching phycoerythrobilin (PEB), resulting in a highly fluorescent protein. Given the small size, high quantum yield, and that unlike green fluorescent proteins, bilin-binding proteins can be used in anaerobic organisms, the orange fluorescent All1280g2-PEB protein is a promising platform for designing new genetically encoded metal ion sensors. Here, we show that All1280g2-PEB undergoes a ∼5-fold reversible zinc-induced fluorescence enhancement with a blue-shifted emission maximum (572 to 517 nm), which is not observed for a related PEB-bound GAF from Synechocystis sp. PCC6803 (Slr1393g3). Zn2+ significantly enhances All1280g2-PEB fluorescence across a biologically relevant pH range from 6.0 to 9.0, with pH-dependent dissociation constants from 1 μM to ∼20-80 nM. Site-directed mutants aiming to sterically decrease and increase access to PEB show a decreased and similar amount of zinc-induced fluorescence enhancement. Mutation of the cysteine residue within the DXCF motif to alanine abolishes the zinc-induced fluorescence enhancement. Collectively, these results support the presence of a unique fluorescence-enhancing Zn2+ binding site in All1280g2-PEB likely involving coordination to the bilin cofactor and requiring a nearby cysteine residue.
Collapse
Affiliation(s)
- Gary C Jensen
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Makena K Janis
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Jazzmin Jara
- Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Nasir Abbasi
- 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
|
36
|
Zhang H, Papadaki S, Sun X, Wang X, Drobizhev M, Yao L, Rehbock M, Köster RW, Wu L, Namikawa K, Piatkevich KD. Quantitative assessment of near-infrared fluorescent proteins. Nat Methods 2023; 20:1605-1616. [PMID: 37666982 PMCID: PMC11753454 DOI: 10.1038/s41592-023-01975-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/29/2023] [Indexed: 09/06/2023]
Abstract
Recent progress in fluorescent protein development has generated a large diversity of near-infrared fluorescent proteins (NIR FPs), which are rapidly becoming popular probes for a variety of imaging applications. However, the diversity of NIR FPs poses a challenge for end-users in choosing the optimal one for a given application. Here we conducted a systematic and quantitative assessment of intracellular brightness, photostability, oligomeric state, chemical stability and cytotoxicity of 22 NIR FPs in cultured mammalian cells and primary mouse neurons and identified a set of top-performing FPs including emiRFP670, miRFP680, miRFP713 and miRFP720, which can cover a majority of imaging applications. The top-performing proteins were further validated for in vivo imaging of neurons in Caenorhabditis elegans, zebrafish, and mice as well as in mice liver. We also assessed the applicability of the selected NIR FPs for multicolor imaging of fusions, expansion microscopy and two-photon imaging.
Collapse
Affiliation(s)
- Hanbin Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Stavrini Papadaki
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaoting Sun
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xinyue Wang
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Mikhail Drobizhev
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Luxia Yao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Michel Rehbock
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard W Köster
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Lianfeng Wu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Kazuhiko Namikawa
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kiryl D Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
37
|
Ramírez Martínez C, Gómez-Pérez LS, Ordaz A, Torres-Huerta AL, Antonio-Perez A. Current Trends of Bacterial and Fungal Optoproteins for Novel Optical Applications. Int J Mol Sci 2023; 24:14741. [PMID: 37834188 PMCID: PMC10572898 DOI: 10.3390/ijms241914741] [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: 08/31/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Photoproteins, luminescent proteins or optoproteins are a kind of light-response protein responsible for the conversion of light into biochemical energy that is used by some bacteria or fungi to regulate specific biological processes. Within these specific proteins, there are groups such as the photoreceptors that respond to a given light wavelength and generate reactions susceptible to being used for the development of high-novel applications, such as the optocontrol of metabolic pathways. Photoswitchable proteins play important roles during the development of new materials due to their capacity to change their conformational structure by providing/eliminating a specific light stimulus. Additionally, there are bioluminescent proteins that produce light during a heatless chemical reaction and are useful to be employed as biomarkers in several fields such as imaging, cell biology, disease tracking and pollutant detection. The classification of these optoproteins from bacteria and fungi as photoreceptors or photoresponse elements according to the excitation-emission spectrum (UV-Vis-IR), as well as their potential use in novel applications, is addressed in this article by providing a structured scheme for this broad area of knowledge.
Collapse
Affiliation(s)
| | | | | | | | - Aurora Antonio-Perez
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Campus Estado de México, Av. Lago de Guadalupe KM 3.5, Margarita Maza de Juárez, Ciudad López Mateos, Atizapán de Zaragoza 52926, Estado de México, Mexico; (C.R.M.); (L.S.G.-P.); (A.O.); (A.L.T.-H.)
| |
Collapse
|
38
|
Dan Q, Jiang X, Wang R, Dai Z, Sun D. Biogenic Imaging Contrast Agents. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207090. [PMID: 37401173 PMCID: PMC10477908 DOI: 10.1002/advs.202207090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 06/08/2023] [Indexed: 07/05/2023]
Abstract
Imaging contrast agents are widely investigated in preclinical and clinical studies, among which biogenic imaging contrast agents (BICAs) are developing rapidly and playing an increasingly important role in biomedical research ranging from subcellular level to individual level. The unique properties of BICAs, including expression by cells as reporters and specific genetic modification, facilitate various in vitro and in vivo studies, such as quantification of gene expression, observation of protein interactions, visualization of cellular proliferation, monitoring of metabolism, and detection of dysfunctions. Furthermore, in human body, BICAs are remarkably helpful for disease diagnosis when the dysregulation of these agents occurs and can be detected through imaging techniques. There are various BICAs matched with a set of imaging techniques, including fluorescent proteins for fluorescence imaging, gas vesicles for ultrasound imaging, and ferritin for magnetic resonance imaging. In addition, bimodal and multimodal imaging can be realized through combining the functions of different BICAs, which helps overcome the limitations of monomodal imaging. In this review, the focus is on the properties, mechanisms, applications, and future directions of BICAs.
Collapse
Affiliation(s)
- Qing Dan
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Xinpeng Jiang
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Run Wang
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| | - Zhifei Dai
- Department of Biomedical EngineeringCollege of Future TechnologyPeking UniversityBeijing100871P. R. China
| | - Desheng Sun
- Shenzhen Key Laboratory for Drug Addiction and Medication SafetyDepartment of UltrasoundInstitute of Ultrasonic MedicinePeking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhen518036P. R. China
| |
Collapse
|
39
|
Wang Z, Li J, Chen J, Cao Z, Li H, Cao Y, Li Q, She M, Liu P, Zhang S, Li J. A NIR fluorescent probe for imaging thiophenol in the living system and revealing thiophenol-induced oxidative stress. CHINESE CHEM LETT 2023; 34:108507. [DOI: 10.1016/j.cclet.2023.108507] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Ando R, Sakaue-Sawano A, Shoda K, Miyawaki A. Two coral fluorescent proteins of distinct colors for sharp visualization of cell-cycle progression. Cell Struct Funct 2023; 48:135-144. [PMID: 37394513 PMCID: PMC10958192 DOI: 10.1247/csf.23028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023] Open
Abstract
We cloned and characterized two new coral fluorescent proteins: h2-3 and 1-41. h2-3 formed an obligate dimeric complex and exhibited bright green fluorescence. On the other hand, 1-41 formed a highly multimeric complex and exhibited dim red fluorescence. We engineered 1-41 into AzaleaB5, a practically useful red-emitting fluorescent protein for cellular labeling applications. We fused h2-3 and AzaleaB5 to the ubiquitination domains of human Geminin and Cdt1, respectively, to generate a new color variant of Fucci (Fluorescent Ubiquitination-based Cell-Cycle Indicator): Fucci5. We found Fucci5 provided more reliable nuclear labeling for monitoring cell-cycle progression than the 1st and 2nd generations that used mAG/mKO2 and mVenus/mCherry, respectively.Key words: fluorescent protein, cell cycle, time-lapse imaging, flow cytometry.
Collapse
Affiliation(s)
- Ryoko Ando
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Asako Sakaue-Sawano
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
- Department of Optical Biomedical Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Keiko Shoda
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
- Laboratory of Bioresponse Analysis, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
41
|
Pal DS, Lin Y, Zhan H, Banerjee T, Kuhn J, Providence S, Devreotes PN. Optogenetic modulation of guanine nucleotide exchange factors of Ras superfamily proteins directly controls cell shape and movement. Front Cell Dev Biol 2023; 11:1195806. [PMID: 37492221 PMCID: PMC10363612 DOI: 10.3389/fcell.2023.1195806] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023] Open
Abstract
In this article, we provide detailed protocols on using optogenetic dimerizers to acutely perturb activities of guanine nucleotide exchange factors (GEFs) specific to Ras, Rac or Rho small GTPases of the migratory networks in various mammalian and amoeba cell lines. These GEFs are crucial components of signal transduction networks which link upstream G-protein coupled receptors to downstream cytoskeletal components and help cells migrate through their dynamic microenvironment. Conventional approaches to perturb and examine these signaling and cytoskeletal networks, such as gene knockout or overexpression, are protracted which allows networks to readjust through gene expression changes. Moreover, these tools lack spatial resolution to probe the effects of local network activations. To overcome these challenges, blue light-inducible cryptochrome- and LOV domain-based dimerization systems have been recently developed to control signaling or cytoskeletal events in a spatiotemporally precise manner. We illustrate that, within minutes of global membrane recruitment of full-length GEFs or their catalytic domains only, widespread increases or decreases in F-actin rich protrusions and cell size occur, depending on the particular node in the networks targeted. Additionally, we demonstrate localized GEF recruitment as a robust assay system to study local network activation-driven changes in polarity and directed migration. Altogether, these optical tools confirmed GEFs of Ras superfamily GTPases as regulators of cell shape, actin dynamics, and polarity. Furthermore, this optogenetic toolbox may be exploited in perturbing complex signaling interactions in varied physiological contexts including mammalian embryogenesis.
Collapse
Affiliation(s)
- Dhiman Sankar Pal
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Yiyan Lin
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Huiwang Zhan
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Tatsat Banerjee
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Jonathan Kuhn
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Stephenie Providence
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Ingenuity Research Program, Baltimore Polytechnic Institute, Baltimore, MD, United States
| | - Peter N. Devreotes
- Department of Cell Biology and Center for Cell Dynamics, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Biological Chemistry, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
42
|
Salsinha AS, Socodato R, Rodrigues A, Vale-Silva R, Relvas JB, Pintado M, Rodríguez-Alcalá LM. Potential of omega-3 and conjugated fatty acids to control microglia inflammatory imbalance elicited by obesogenic nutrients. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159331. [PMID: 37172801 DOI: 10.1016/j.bbalip.2023.159331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/05/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
High-fat diet-induced obesity detrimentally affects brain function by inducing chronic low-grade inflammation. This neuroinflammation is, at least in part, likely to be mediated by microglia, which are the main immune cell population in the brain. Microglia express a wide range of lipid-sensitive receptors and their activity can be modulated by fatty acids that cross the blood-brain barrier. Here, by combining live cell imaging and FRET technology we assessed how different fatty acids modulate microglia activity. We demonstrate that the combined action of fructose and palmitic acid induce Ikβα degradation and nuclear translocation of the p65 subunit nuclear factor kB (NF-κB) in HCM3 human microglia. Such obesogenic nutrients also lead to reactive oxygen species production and LynSrc activation (critical regulators of microglia inflammation). Importantly, short-time exposure to omega-3 (EPA and DHA), CLA and CLNA are sufficient to abolish NF-κB pathway activation, suggesting a potential neuroprotective role. Omega-3 and CLA also show an antioxidant potential by inhibiting reactive oxygen species production, and the activation of LynSrc in microglia. Furthermore, using chemical agonists (TUG-891) and antagonists (AH7614) of GPR120/FFA4, we demonstrated that omega-3, CLA and CLNA inhibition of the NF-κB pathway is mediated by this receptor, while omega-3 and CLA antioxidant potential occurs through different signaling mechanisms.
Collapse
Affiliation(s)
- A S Salsinha
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - R Socodato
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - A Rodrigues
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - R Vale-Silva
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - J B Relvas
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - M Pintado
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| | - L M Rodríguez-Alcalá
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina, Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho, 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
43
|
Xu Q, Xiao F, Xu H. Fluorescent detection of emerging virus based on nanoparticles: From synthesis to application. Trends Analyt Chem 2023; 161:116999. [PMID: 36852170 PMCID: PMC9946731 DOI: 10.1016/j.trac.2023.116999] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/26/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
The spread of COVID-19 has caused huge economic losses and irreversible social impact. Therefore, to successfully prevent the spread of the virus and solve public health problems, it is urgent to develop detection methods with high sensitivity and accuracy. However, existing detection methods are time-consuming, rely on instruments, and require skilled operators, making rapid detection challenging to implement. Biosensors based on fluorescent nanoparticles have attracted interest in the field of detection because of their advantages, such as high sensitivity, low detection limit, and simple result readout. In this review, we systematically describe the synthesis, intrinsic advantages, and applications of organic dye-doped fluorescent nanoparticles, metal nanoclusters, up-conversion particles, quantum dots, carbon dots, and others for virus detection. Furthermore, future research initiatives are highlighted, including green production of fluorescent nanoparticles with high quantum yield, speedy signal reading by integrating with intelligent information, and error reduction by coupling with numerous fluorescent nanoparticles.
Collapse
Affiliation(s)
- Qian Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| | - Fangbin Xiao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, PR China
| |
Collapse
|
44
|
Actin-driven chromosome clustering facilitates fast and complete chromosome capture in mammalian oocytes. Nat Cell Biol 2023; 25:439-452. [PMID: 36732633 PMCID: PMC10014578 DOI: 10.1038/s41556-022-01082-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/20/2022] [Indexed: 02/04/2023]
Abstract
Accurate chromosome segregation during meiosis is crucial for reproduction. Human and porcine oocytes transiently cluster their chromosomes before the onset of spindle assembly and subsequent chromosome segregation. The mechanism and function of chromosome clustering are unknown. Here we show that chromosome clustering is required to prevent chromosome losses in the long gap phase between nuclear envelope breakdown and the onset of spindle assembly, and to promote the rapid capture of all chromosomes by the acentrosomal spindle. The initial phase of chromosome clustering is driven by a dynamic network of Formin-2- and Spire-nucleated actin cables. The actin cables form in the disassembling nucleus and migrate towards the nuclear centre, moving the chromosomes centripetally by interacting with their arms and kinetochores as they migrate. A cage of stable microtubule loops drives the late stages of chromosome clustering. Together, our data establish a crucial role for chromosome clustering in accurate progression through meiosis.
Collapse
|
45
|
Chen YL, Xie XX, Zhong N, Sun LC, Lin D, Zhang LJ, Weng L, Jin T, Cao MJ. Research Progresses and Applications of Fluorescent Protein Antibodies: A Review Focusing on Nanobodies. Int J Mol Sci 2023; 24:4307. [PMID: 36901737 PMCID: PMC10002328 DOI: 10.3390/ijms24054307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Since the discovery of fluorescent proteins (FPs), their rich fluorescence spectra and photochemical properties have promoted widespread biological research applications. FPs can be classified into green fluorescent protein (GFP) and its derivates, red fluorescent protein (RFP) and its derivates, and near-infrared FPs. With the continuous development of FPs, antibodies targeting FPs have emerged. The antibody, a class of immunoglobulin, is the main component of humoral immunity that explicitly recognizes and binds antigens. Monoclonal antibody, originating from a single B cell, has been widely applied in immunoassay, in vitro diagnostics, and drug development. The nanobody is a new type of antibody entirely composed of the variable domain of a heavy-chain antibody. Compared with conventional antibodies, these small and stable nanobodies can be expressed and functional in living cells. In addition, they can easily access grooves, seams, or hidden antigenic epitopes on the surface of the target. This review provides an overview of various FPs, the research progress of their antibodies, particularly nanobodies, and advanced applications of nanobodies targeting FPs. This review will be helpful for further research on nanobodies targeting FPs, making FPs more valuable in biological research.
Collapse
Affiliation(s)
- Yu-Lei Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xin-Xin Xie
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ning Zhong
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Le-Chang Sun
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Duanquan Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ling-Jing Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ling Weng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Tengchuan Jin
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230007, China
| | - Min-Jie Cao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
46
|
Oliinyk OS, Ma C, Pletnev S, Baloban M, Taboada C, Sheng H, Yao J, Verkhusha VV. Deep-tissue SWIR imaging using rationally designed small red-shifted near-infrared fluorescent protein. Nat Methods 2023; 20:70-74. [PMID: 36456785 PMCID: PMC10725253 DOI: 10.1038/s41592-022-01683-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022]
Abstract
Applying rational design, we developed 17 kDa cyanobacteriochrome-based near-infrared (NIR-I) fluorescent protein, miRFP718nano. miRFP718nano efficiently binds endogenous biliverdin chromophore and brightly fluoresces in mammalian cells and tissues. miRFP718nano has maximal emission at 718 nm and an emission tail in the short-wave infrared (SWIR) region, allowing deep-penetrating off-peak fluorescence imaging in vivo. The miRFP718nano structure reveals the molecular basis of its red shift. We demonstrate superiority of miRFP718nano-enabled SWIR imaging over NIR-I imaging of microbes in the mouse digestive tract, mammalian cells injected into the mouse mammary gland and NF-kB activity in a mouse model of liver inflammation.
Collapse
Affiliation(s)
- Olena S Oliinyk
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Chenshuo Ma
- Department of Biomedical Engineering, School of Engineering, Duke University, Durham, NC, USA
| | - Sergei Pletnev
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mikhail Baloban
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carlos Taboada
- Department of Biomedical Engineering, School of Engineering, Duke University, Durham, NC, USA
| | - Huaxin Sheng
- Department of Anesthesiology, School of Medicine, Duke University, Durham, NC, USA
| | - Junjie Yao
- Department of Biomedical Engineering, School of Engineering, Duke University, Durham, NC, USA.
| | - 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, USA.
| |
Collapse
|
47
|
Saxena T, Sie C, Lin K, Ye D, Saatchi K, Häfeli UO. Potential of Nuclear Imaging Techniques to Study the Oral Delivery of Peptides. Pharmaceutics 2022; 14:2809. [PMID: 36559303 PMCID: PMC9780892 DOI: 10.3390/pharmaceutics14122809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Peptides are small biomolecules known to stimulate or inhibit important functions in the human body. The clinical use of peptides by oral delivery, however, is very limited due to their sensitive structure and physiological barriers present in the gastrointestinal tract. These barriers can be overcome with chemical and mechanical approaches protease inhibitors, permeation enhancers, and polymeric encapsulation. Studying the success of these approaches pre-clinically with imaging techniques such as fluorescence imaging (IVIS) and optical microscopy is difficult due to the lack of in-depth penetration. In comparison, nuclear imaging provides a better platform to observe the gastrointestinal transit and quantitative distribution of radiolabeled peptides. This review provides a brief background on the oral delivery of peptides and states examples from the literature on how nuclear imaging can help to observe and analyze the gastrointestinal transit of oral peptides. The review connects the fields of peptide delivery and nuclear medicine in an interdisciplinary way to potentially overcome the challenges faced during the study of oral peptide formulations.
Collapse
Affiliation(s)
- Tanya Saxena
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Claire Sie
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Kristine Lin
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Daisy Ye
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Urs O. Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
48
|
Blanch-Asensio A, Grandela C, Brandão KO, de Korte T, Mei H, Ariyurek Y, Yiangou L, Mol MP, van Meer BJ, Kloet SL, Mummery CL, Davis RP. STRAIGHT-IN enables high-throughput targeting of large DNA payloads in human pluripotent stem cells. CELL REPORTS METHODS 2022; 2:100300. [PMID: 36313798 PMCID: PMC9606106 DOI: 10.1016/j.crmeth.2022.100300] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 07/12/2022] [Accepted: 08/31/2022] [Indexed: 04/20/2023]
Abstract
Inserting large DNA payloads (>10 kb) into specific genomic sites of mammalian cells remains challenging. Applications ranging from synthetic biology to evaluating the pathogenicity of disease-associated variants for precision medicine initiatives would greatly benefit from tools that facilitate this process. Here, we merge the strengths of different classes of site-specific recombinases and combine these with CRISPR-Cas9-mediated homologous recombination to develop a strategy for stringent site-specific replacement of genomic fragments at least 50 kb in size in human induced pluripotent stem cells (hiPSCs). We demonstrate the versatility of STRAIGHT-IN (serine and tyrosine recombinase-assisted integration of genes for high-throughput investigation) by (1) inserting various combinations of fluorescent reporters into hiPSCs to assess the excitation-contraction coupling cascade in derivative cardiomyocytes and (2) simultaneously targeting multiple variants associated with inherited cardiac arrhythmic disorders into a pool of hiPSCs. STRAIGHT-IN offers a precise approach to generate genetically matched panels of hiPSC lines efficiently and cost effectively.
Collapse
Affiliation(s)
- Albert Blanch-Asensio
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Catarina Grandela
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Karina O. Brandão
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Tessa de Korte
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333RC Leiden, the Netherlands
| | - Yavuz Ariyurek
- Leiden Genome Technology Center, Leiden University Medical Center, 2333RC Leiden, the Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Mervyn P.H. Mol
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| | - Susan L. Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, 2333RC Leiden, the Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, 7500AE Enschede, the Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2300RC Leiden, the Netherlands
| |
Collapse
|
49
|
Scott L, Wigglesworth MJ, Siewers V, Davis AM, David F. Genetically Encoded Whole Cell Biosensor for Drug Discovery of HIF-1 Interaction Inhibitors. ACS Synth Biol 2022; 11:3182-3189. [PMID: 36223492 PMCID: PMC9594322 DOI: 10.1021/acssynbio.2c00274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The heterodimeric transcription factor, hypoxia inducible factor-1 (HIF-1), is an important anticancer target as it supports the adaptation and response of tumors to hypoxia. Here, we optimized the repressed transactivator yeast two-hybrid system to further develop it as part of a versatile yeast-based drug discovery platform and validated it using HIF-1. We demonstrate both fluorescence-based and auxotrophy-based selections that could detect HIF-1α/HIF-1β dimerization inhibition. The engineered genetic selection is tunable and able to differentiate between strong and weak interactions, shows a large dynamic range, and is stable over different growth phases. Furthermore, we engineered mechanisms to control for cellular activity and off-target drug effects. We thoroughly characterized all parts of the biosensor system and argue this tool will be generally applicable to a wide array of protein-protein interaction targets. We anticipate this biosensor will be useful as part of a drug discovery platform, particularly when screening DNA-encoded new modality drugs.
Collapse
Affiliation(s)
- Louis
H. Scott
- Discovery
Sciences, Biopharmaceuticals R&D, AstraZeneca, SE-41320 Gothenburg, Sweden,Department
of Biology and Biological Engineering, Division of Systems and Synthetic
Biology, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Mark J. Wigglesworth
- Discovery
Sciences, Biopharmaceuticals R&D, AstraZeneca, Alderley Park SK10 2NA, U.K.
| | - Verena Siewers
- Department
of Biology and Biological Engineering, Division of Systems and Synthetic
Biology, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Andrew M. Davis
- Discovery
Sciences, Biopharmaceutical R&D, AstraZeneca, Cambridge, CB2 0AA, U.K.
| | - Florian David
- Department
of Biology and Biological Engineering, Division of Systems and Synthetic
Biology, Chalmers University of Technology, SE-41296 Gothenburg, Sweden,
| |
Collapse
|
50
|
Ender P, Gagliardi PA, Dobrzyński M, Frismantiene A, Dessauges C, Höhener T, Jacques MA, Cohen AR, Pertz O. Spatiotemporal control of ERK pulse frequency coordinates fate decisions during mammary acinar morphogenesis. Dev Cell 2022; 57:2153-2167.e6. [PMID: 36113484 DOI: 10.1016/j.devcel.2022.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/06/2022] [Accepted: 08/20/2022] [Indexed: 12/30/2022]
Abstract
The signaling events controlling proliferation, survival, and apoptosis during mammary epithelial acinar morphogenesis remain poorly characterized. By imaging single-cell ERK activity dynamics in MCF10A acini, we find that these fates depend on the average frequency of non-periodic ERK pulses. High pulse frequency is observed during initial acinus growth, correlating with rapid cell motility and proliferation. Subsequent decrease in motility correlates with lower ERK pulse frequency and quiescence. Later, during lumen formation, coordinated multicellular ERK waves emerge, correlating with high and low ERK pulse frequencies in outer surviving and inner dying cells, respectively. Optogenetic entrainment of ERK pulses causally connects high ERK pulse frequency with inner cell survival. Acini harboring the PIK3CA H1047R mutation display increased ERK pulse frequency and inner cell survival. Thus, fate decisions during acinar morphogenesis are coordinated by different spatiotemporal modalities of ERK pulse frequency.
Collapse
Affiliation(s)
- Pascal Ender
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | | | - Maciej Dobrzyński
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Agne Frismantiene
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Coralie Dessauges
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Thomas Höhener
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Marc-Antoine Jacques
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Andrew R Cohen
- Department of Electrical and Computer Engineering, Drexel University, 3120-40 Market Street, Suite 313, Philadelphia, PA 19104, USA
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|