1
|
Liu H, Dong J, Wu R, Dai J, Lou X, Xia F, Willner I, Huang F. Light-Triggered CRISPR/Cas12a for Genomic Editing and Tumor Regression. Angew Chem Int Ed Engl 2025:e202502892. [PMID: 40334276 DOI: 10.1002/anie.202502892] [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: 02/04/2025] [Revised: 04/23/2025] [Accepted: 05/07/2025] [Indexed: 05/09/2025]
Abstract
A photo-triggered CRISPR/Cas12a machinery for in vitro and in vivo gene editing is introduced. The system consists of a caged, inactive ortho-nitrobenzyl phosphate ester photo-responsive crRNA, which, upon light-induced deprotection, yields the active CRISPR/Cas12a gene editing machinery (LAC12aGE). The LAC12aGE system induces specific thymidine-rich (TTTN) protospacer-adjacent motif (PAM)-guided double-stranded breaks in genomic DNA, which upon non-homologous end-joining lead to gene repair. The LAC12aGE machinery is applied for gene editing of an exogenous dual fluorescent reporter gene in living cells, as well as the endogenous gene encoding DNA methyltransferase 1. In addition, the LAC12aGE is applied for in vitro gene editing and disruption of the hepatocyte growth factor (HGF) gene in HepG2 cells, where knockout of the HGF gene results in inhibited cell proliferation and migration, as well as enhanced apoptosis. Moreover, the in vivo knockout and disruption of the HGF gene in HepG2 tumors by the LAC12aGE machinery is demonstrated. The cyclic temporal development of the LAC12aGE system in tumors shows effective inhibition of tumor growth and enhanced apoptosis/necrosis of tumor tissues compared to control systems.
Collapse
Affiliation(s)
- Hong Liu
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Jiantong Dong
- The Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Renzhi Wu
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Xiaoding Lou
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Fan Xia
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Itamar Willner
- The Institute of Chemistry, The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Fujian Huang
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| |
Collapse
|
2
|
Lewash SA, McKenney VR, Wuebben C, Ludwig J, Hosni R, Radzey D, Toma MI, Bartok E, Schlee M, Zillinger T, Heckel A, Hartmann G. Immunoengineering of a Photocaged 5´-triphosphate Oligoribonucleotide Ligand for Spatiotemporal Control of RIG-I Activation in Cancer. Angew Chem Int Ed Engl 2025; 64:e202423321. [PMID: 40095771 PMCID: PMC12087816 DOI: 10.1002/anie.202423321] [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: 11/29/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/19/2025]
Abstract
Photochemical control of oligonucleotides bears great potential for the spatio-temporal control of therapeutic targets, such as immune sensing receptors. Retinoic acid-inducible gene I (RIG-I) is a cytoplasmic receptor of the innate immune system that triggers antiviral responses upon detection of viral RNA. RIG-I can be specifically activated by short double-stranded (ds) RNA with a blunt 5' end bearing a triphosphate, mimicking nascent viral transcripts. Tumor cells are specifically sensitive to RIG-I-induced cell death. Here we developed a potent oligonucleotide ligand for spatiotemporally controlled activation of RIG-I by light exposure. Through structural considerations and functional studies we identified a combination of two nucleoside positions in a RIG-I oligonucleotide ligand for which the substitution of both respective 2'-hydroxy groups of the ribose by photolabile protecting groups (2'-photocages) resulted in a complete loss of RIG-I ligand activity, whereas photocaging the individual positions was not sufficient to turn off RIG-I. Light exposure fully restored RIG-I activation by the photocaged RIG-I ligand, enabling light-controlled RIG-I-mediated cell death of human cancer cells which had internalized the photocaged RIG-I ligand prior to light exposure. This novel photoactivatable RIG-I oligonucleotide ligand may be applicable for precise light-controlled induction of tumor cell death in superficial cancer such as melanoma.
Collapse
Affiliation(s)
- Sandra Anika Lewash
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Vivien Rose McKenney
- Institute for Organic Chemistry and Chemical BiologyGoethe University FrankfurtMax‐von‐Laue‐Straße 960438Frankfurt am MainGermany
| | - Christine Wuebben
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Janos Ludwig
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Racha Hosni
- Institute of PathologyUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Dirk Radzey
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Marieta I. Toma
- Institute of PathologyUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Eva Bartok
- Institute of Experimental Haematology and Transfusion MedicineUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| | - Thomas Zillinger
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
- Department of BiomedicineAarhus UniversityHøegh‐Guldbergs Gade 10Aarhus8000Denmark
| | - Alexander Heckel
- Institute for Organic Chemistry and Chemical BiologyGoethe University FrankfurtMax‐von‐Laue‐Straße 960438Frankfurt am MainGermany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology – Immunology in TranslationUniversity Hospital BonnVenusberg‐Campus 153127BonnGermany
| |
Collapse
|
3
|
Shen D, Zhao Q, Zhang H, Wu C, Jin H, Guo K, Sun R, Guo H, Zhao Q, Feng H, Dong X, Gao Z, Zhang L, Liu Y. A hydrophobic photouncaging reaction to profile the lipid droplet interactome in tissues. Proc Natl Acad Sci U S A 2025; 122:e2420861122. [PMID: 40238459 PMCID: PMC12037041 DOI: 10.1073/pnas.2420861122] [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: 10/10/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Most bioorthogonal photouncaging reactions preferentially occur in polar environments to accommodate biological applications in the aqueous cellular milieu. However, they are not precisely designed to chemically adapt to the diverse microenvironments of the cell. Herein, we report a hydrophobic photouncaging reaction with tailored photolytic kinetics toward solvent polarity. Structural modulations of the aminobenzoquinone-based photocage reveal the impact of cyclic ring size, steric substituent, and electronic substituent on the individual uncaging kinetics (kH2O and kdioxane) and polarity preference (kdioxane/kH2O). Rational incorporation of optimized moieties leads to up to 20.2-fold nonpolar kinetic selectivity (kdioxane/kH2O). Further photochemical spectroscopic characterizations and theoretical calculations together uncover the mechanism underlying the polarity-dependent uncaging kinetics. The uncaged ortho-quinone methide product bears covalent reactivity toward diverse nucleophiles of a protein revealed by tandem mass spectrometry. Finally, we demonstrate the application of such lipophilic photouncaging chemistry toward selective labeling and profiling of proteins in proximity to lipid droplets inside human fatty liver tissues. Together, this work studies the solvent polarity effects of a photouncaging reaction and chemically adapts it toward suborganelle-targeted protein proximity labeling and profiling.
Collapse
Affiliation(s)
- Di Shen
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Qun Zhao
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Huaiyue Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Ci Wu
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Hao Jin
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Kun Guo
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Rui Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Hengke Guo
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Qi Zhao
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Huan Feng
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Zhenming Gao
- The Second Hospital of Dalian Medical University, Dalian116023, China
| | - Lihua Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic Research & Analysis Center, Chinese Academy of Sciences Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian116023, China
| |
Collapse
|
4
|
Caldwell S, Demyan IR, Falcone GN, Parikh A, Lohmueller J, Deiters A. Conditional Control of Benzylguanine Reaction with the Self-Labeling SNAP-tag Protein. Bioconjug Chem 2025; 36:540-548. [PMID: 39977950 PMCID: PMC11926790 DOI: 10.1021/acs.bioconjchem.5c00002] [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: 01/04/2025] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
SNAP-tag, a mutant of the O6-alkylguanine-DNA-alkyltransferase, self-labels by reacting with benzylguanine (BG) substrates, thereby forming a thioether bond. SNAP-tag has been genetically fused to a wide range of proteins of interest in order to covalently modify them. In the context of both diagnostic and therapeutic applications, as well as use as a biological recording device, precise control in a spatial and temporal fashion over the covalent bond-forming reaction is desired to direct inputs, readouts, or therapeutic actions to specific locations, at specific time points, in cells and organisms. Here, we introduce a comprehensive suite of six caged BG molecules: one light-triggered and five others that can be activated through various chemical and biochemical stimuli, such as small molecules, transition metal catalysts, reactive oxygen species, and enzymes. These molecules are unable to react with SNAP-tag until the trigger is present, which leads to near complete SNAP-tag conjugation, as illustrated both in biochemical assays and on human cell surfaces. This approach holds promise for targeted therapeutic assembly at disease sites, offering the potential to reduce off-target effects and toxicity through precise trigger titration.
Collapse
Affiliation(s)
- Steven
E. Caldwell
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Isabella R. Demyan
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Gianna N. Falcone
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Avani Parikh
- Department
of Surgery, Division of Surgical Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Jason Lohmueller
- Department
of Surgery, Division of Surgical Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
- Center
for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Alexander Deiters
- Department
of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
- Center
for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
5
|
Wurnig SL, Hanl M, Geiger TM, Zhai S, Dressel I, Pieńkowska DE, Nowak RP, Hansen FK. Light-activatable photochemically targeting chimeras (PHOTACs) enable the optical control of targeted protein degradation of HDAC6. RSC Med Chem 2025:d4md00972j. [PMID: 40135143 PMCID: PMC11931564 DOI: 10.1039/d4md00972j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/23/2025] [Indexed: 03/27/2025] Open
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional modalities that induce protein degradation via a catalytic mode of action. Photochemically targeting chimeras (PHOTACs) are a subset of PROTACs designed for light-activated protein degradation, thereby offering precise spatiotemporal control. In this study, we report the design, solid-phase synthesis, and characterization of the first PHOTACs targeting histone deacetylase 6 (HDAC6). We achieved this by incorporating an azobenzene photoswitch into our previously developed HDAC6-selective PROTAC A6. Among the synthesized compounds, PHOTAC 12 demonstrated no HDAC6 degradation in the absence of light but showed significant degradation upon activation to its cis-state with 390 nm light irradiation. Notably, we find that PHOTAC 12 in the cis-state shows significantly improved ternary complex formation compared to the trans-state correlating with its degradation efficacy. Overall, PHOTAC 12 is a promising lead compound for the development of light-activatable HDAC6 degraders.
Collapse
Affiliation(s)
- Silas L Wurnig
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Maria Hanl
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Thomas M Geiger
- Institute of Structural Biology, Medical Faculty, University of Bonn Venusberg Campus 1 53127 Bonn Germany
| | - Shiyang Zhai
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Ina Dressel
- Institute of Structural Biology, Medical Faculty, University of Bonn Venusberg Campus 1 53127 Bonn Germany
| | - Dominika E Pieńkowska
- Institute of Structural Biology, Medical Faculty, University of Bonn Venusberg Campus 1 53127 Bonn Germany
| | - Radosław P Nowak
- Institute of Structural Biology, Medical Faculty, University of Bonn Venusberg Campus 1 53127 Bonn Germany
| | - Finn K Hansen
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| |
Collapse
|
6
|
Lu H, Ye H, Xin J, You L. Photoswitchable Topological Regulation of Covalent Macrocycles, Molecular Recognition, and Interlocked Structures. Angew Chem Int Ed Engl 2025; 64:e202421175. [PMID: 39719400 DOI: 10.1002/anie.202421175] [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: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 12/26/2024]
Abstract
Macrocycles represent one important class of functional molecules, and dynamic macrocycles with the potential of cleavability, adaptability, and topological conversion are challenging. Herein we report photoswitchable allosteric and topological control of dynamic covalent macrocycles and further the use in guest binding and mechanically interlocked molecules. The manipulation of competing ring-chain equilibria and bond formation/scission within reaction systems enabled light-induced structural regulation over dithioacetal and thioacetal dynamic bonds, accordingly realizing bidirectional switching between crown ether-like covalent macrocycles and their linear counterparts. The on-demand photoswitchable topological transformation of macrocycles further allowed guest recognition/release exhibiting controllable binding affinity and selectivity. To showcase the capability light-triggered assembly/disassembly of diverse mechanically interlocked structures, such as rotaxanes and catenanes, was achieved. The realization of photoswitchable topological conversion of covalent macrocycles, which has been rarely reported before, demonstrates the potential of light-triggered reactivity control and structural reconfiguration for enhanced complexity and sophisticated function. The strategies and results should be appealing to endeavors in molecular recognition, dynamic assemblies, molecular machines, and intelligent materials.
Collapse
Affiliation(s)
- Hanwei Lu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - Hebo Ye
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - Jiafan Xin
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - Lei You
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian, 350108, China
| |
Collapse
|
7
|
Wang X, Kerckhoffs A, Riexinger J, Cornall M, Langton MJ, Bayley H, Qing Y. ON-OFF nanopores for optical control of transmembrane ionic communication. NATURE NANOTECHNOLOGY 2025; 20:432-440. [PMID: 39838209 PMCID: PMC11919769 DOI: 10.1038/s41565-024-01823-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/09/2024] [Indexed: 01/23/2025]
Abstract
Nanoscale photoswitchable proteins could facilitate precise spatiotemporal control of transmembrane communication and support studies in synthetic biology, neuroscience and bioelectronics. Here, through covalent modification of the α-haemolysin protein pore with arylazopyrazole photoswitches, we produced 'photopores' that transition between iontronic resistor and diode modes in response to irradiation at orthogonal wavelengths. In the diode mode, a low-leak OFF-state nanopore exhibits a reversible increase in unitary conductance of more than 20-fold upon irradiation at 365 nm. A rectification ratio of >5 was achieved with photopores in the diode state by either direct or alternating voltage input. Unlike conventional electronic phototransistors with intensity-dependent photoelectric responses, the photopores regulated current output solely based on the wavelength(s) of monochromatic or dual-wavelength irradiation. Dual-wavelength irradiation at various relative intensities allowed graded adjustment of the photopore conductance. By using these properties, photonic signals encoding text or graphic messages were converted into ionic signals, highlighting the potential applications of photopores as components of smart devices in synthetic biology.
Collapse
Affiliation(s)
- Xingzao Wang
- Department of Chemistry, University of Oxford, Oxford, UK
| | | | | | | | | | - Hagan Bayley
- Department of Chemistry, University of Oxford, Oxford, UK.
| | - Yujia Qing
- Department of Chemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Charette M, Rosenblum C, Shade O, Deiters A. Optogenetics with Atomic Precision─A Comprehensive Review of Optical Control of Protein Function through Genetic Code Expansion. Chem Rev 2025; 125:1663-1717. [PMID: 39928721 PMCID: PMC11869211 DOI: 10.1021/acs.chemrev.4c00224] [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: 03/22/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 02/12/2025]
Abstract
Conditional control of protein activity is important in order to elucidate the particular functions and interactions of proteins, their regulators, and their substrates, as well as their impact on the behavior of a cell or organism. Optical control provides a perhaps optimal means of introducing spatiotemporal control over protein function as it allows for tunable, rapid, and noninvasive activation of protein activity in its native environment. One method of introducing optical control over protein activity is through the introduction of photocaged and photoswitchable noncanonical amino acids (ncAAs) through genetic code expansion in cells and animals. Genetic incorporation of photoactive ncAAs at key residues in a protein provides a tool for optical activation, or sometimes deactivation, of protein activity. Importantly, the incorporation site can typically be rationally selected based on structural, mechanistic, or computational information. In this review, we comprehensively summarize the applications of photocaged lysine, tyrosine, cysteine, serine, histidine, glutamate, and aspartate derivatives, as well as photoswitchable phenylalanine analogues. The extensive and diverse list of proteins that have been placed under optical control demonstrates the broad applicability of this methodology.
Collapse
Affiliation(s)
- Maura Charette
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Carolyn Rosenblum
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Olivia Shade
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
9
|
Osgood AO, Huang Z, Szalay KH, Chatterjee A. Strategies to Expand the Genetic Code of Mammalian Cells. Chem Rev 2025; 125:2474-2501. [PMID: 39937611 DOI: 10.1021/acs.chemrev.4c00730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Genetic code expansion (GCE) in mammalian cells has emerged as a powerful technology for investigating and engineering protein function. This method allows for the precise incorporation of a rapidly growing toolbox of noncanonical amino acids (ncAAs) into predefined sites of target proteins expressed in living cells. Due to the minimal size of these genetically encoded ncAAs, the wide range of functionalities they provide, and the ability to introduce them freely at virtually any site of any protein by simple mutagenesis, this technology holds immense potential for probing the complex biology of mammalian cells and engineering next-generation biotherapeutics. In this review, we provide an overview of the underlying machinery that enables ncAA mutagenesis in mammalian cells and how these are developed. We have also compiled an updated list of ncAAs that have been successfully incorporated into proteins in mammalian cells. Finally, we provide our perspectives on the current challenges that need to be addressed to fully harness the potential of this technology.
Collapse
Affiliation(s)
- Arianna O Osgood
- Department of Chemistry, Boston College, 201 Merkert Chemistry Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, United States
| | - Zeyi Huang
- Department of Chemistry, Boston College, 201 Merkert Chemistry Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, United States
| | - Kaitlyn H Szalay
- Department of Chemistry, Boston College, 201 Merkert Chemistry Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, United States
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 201 Merkert Chemistry Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02467, United States
| |
Collapse
|
10
|
Pöschko P, Berrou CM, Pakari K, Ziegler MJ, Kern C, Koch B, Wittbrodt J, Wombacher R. Photoactivatable Plant Hormone-Based Chemical Inducers of Proximity for In Vivo Applications. ACS Chem Biol 2025; 20:332-339. [PMID: 39868662 PMCID: PMC11851429 DOI: 10.1021/acschembio.4c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025]
Abstract
Protein interactions play a crucial role in regulating cellular mechanisms, highlighting the need for effective methods to control these processes. In this regard, chemical inducers of proximity (CIPs) offer a promising approach to precisely manipulate protein-protein interactions in live cells and in vivo. In this study, we introduce pMandi, a photocaged version of the plant hormone-based CIP mandipropamid (Mandi), which allows the use of light as an external trigger to induce protein proximity in live mammalian cells. Furthermore, we present opabactin (OP) as a new plant hormone-based CIP that is effective in live mammalian cells at low nanomolar concentration and in live medaka embryos at submicromolar concentration. Its photocaged derivative, pOP, enables the induction of protein proximity upon light exposure in individual cells, enhancing spatiotemporal control to the level of single-cell resolution. Additionally, we explored the use of both photocaged CIPs to promote protein proximity in live medaka embryos.
Collapse
Affiliation(s)
- Philipp Pöschko
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
- Faculty
of Biosciences, Heidelberg University, Im Neuenheimer Feld 234, 69120 Heidelberg, Germany
| | - Caroline M. Berrou
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Kaisa Pakari
- Centre
for Organismal Studies Heidelberg (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
- Heidelberg
Biosciences International Graduate School (HBIGS), Heidelberg University, Im Neuenheimer Feld 501, 69120 Heidelberg, Germany
| | - Michael J. Ziegler
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Christoph Kern
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Birgit Koch
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre
for Organismal Studies Heidelberg (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Richard Wombacher
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstraße 29, 69120 Heidelberg, Germany
| |
Collapse
|
11
|
von Glasenapp V, C Almeida A, Chang D, Gasic I, Winssinger N, Gotta M. Spatio-temporal control of mitosis using light via a Plk1 inhibitor caged for activity and cellular permeability. Nat Commun 2025; 16:1599. [PMID: 39971898 PMCID: PMC11840123 DOI: 10.1038/s41467-025-56746-5] [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: 03/26/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
The ability to control the activity of kinases spatially and temporally is essential to elucidate the role of signalling pathways in development and physiology. Progress in this direction has been hampered by the lack of tools to manipulate kinase activity in a highly controlled manner in vivo. Here we report a strategy to modify BI2536, the well characterized inhibitor of the conserved and essential mitotic kinase Polo-like kinase 1 (Plk1). We introduce the same coumarin photolabile protecting group (PPG) at two positions of the inhibitor. At one position, the coumarin prevents the interaction with Plk1, at the second it masks an added carboxylic acid, important for cellular retention. Exposure to light results in removal of both PPGs, leading to the activation of the inhibitor and its trapping inside cells. We demonstrate the efficacy of the caged inhibitor in three-dimensional spheroid cultures: by uncaging it with a single light pulse, we can inhibit Plk1 and arrest cell division, a highly dynamic process, with spatio-temporal control. Our design can be applied to other small molecules, providing a solution to control their activity in living cells with unprecedented precision.
Collapse
Affiliation(s)
- Victoria von Glasenapp
- Department of Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Ana C Almeida
- Department of Molecular and Cellular Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Dalu Chang
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Ivana Gasic
- Department of Molecular and Cellular Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Nicolas Winssinger
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland.
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva, Switzerland.
| | - Monica Gotta
- Department of Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- NCCR Chemical Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
12
|
Banerjee S, Cakil ZV, Gallant K, van den Boom J, Palei S, Meyer H, Gersch M, Summerer D. Light-Activatable Ubiquitin for Studying Linkage-Specific Ubiquitin Chain Formation Kinetics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406570. [PMID: 39716962 PMCID: PMC11809417 DOI: 10.1002/advs.202406570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/08/2024] [Indexed: 12/25/2024]
Abstract
Ubiquitination is a dynamic post-translational modification governing protein abundance, function, and localization in eukaryotes. The Ubiquitin protein is conjugated to lysine residues of target proteins, but can also repeatedly be ubiquitinated itself, giving rise to a complex code of ubiquitin chains with different linkage types. To enable studying the cellular dynamics of linkage-specific ubiquitination, light-activatable polyubiquitin chain formation is reported here. By incorporating a photocaged lysine at specific sites within ubiquitin through amber codon suppression, light-dependent activation of ubiquitin chain extension is enabled for the monitoring of linkage-specific polyubiquitination. The studies reveal rapid, minute-scale ubiquitination kinetics for K11, K48, and K63 linkages. The role of individual components of the ubiquitin-proteasome system in K48-initiated chain synthesis is further studied by small molecule inhibition. The approach expands current perturbation strategies with the ability to control linkage-specific ubiquitination with high temporal resolution and should find broad application for studying ubiquitinome dynamics.
Collapse
Affiliation(s)
- Sudakshina Banerjee
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Zeyneb Vildan Cakil
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Kai Gallant
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
- Max Planck Institute of Molecular PhysiologyChemical Genomics CenterOtto‐Hahn Str. 1544227DortmundGermany
| | - Johannes van den Boom
- Center of Medical BiotechnologyFaculty of BiologyUniversity of Duisburg‐EssenUniversitätsstr. 245141EssenGermany
| | - Shubhendu Palei
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Hemmo Meyer
- Center of Medical BiotechnologyFaculty of BiologyUniversity of Duisburg‐EssenUniversitätsstr. 245141EssenGermany
| | - Malte Gersch
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
- Max Planck Institute of Molecular PhysiologyChemical Genomics CenterOtto‐Hahn Str. 1544227DortmundGermany
| | - Daniel Summerer
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| |
Collapse
|
13
|
Jia J, Zhang X, Li Y, Wang T, An Y, Yan X, Liu B, Yang C, Ju H. Remotely Sequential Activation of Biofunctional MXenes for Spatiotemporally Controlled Photothermal Cancer Therapy Integrated with Multimodal Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410535. [PMID: 39665387 DOI: 10.1002/smll.202410535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Spatiotemporally controlled cancer therapy may offer great advantages in precision medicine, but still remains some challenges in programmed sequential release and co-localization of components at target sites. Herein, a MXene-based nanoprobe (TCC@M) is meticulously designed by engineering of photodynamically activated CRISPR-Cas9 and cancer cell membrane-camouflaged Ti3C2 MXenes for targeting delivery and spatiotemporally controlled gene regulation followed by enhanced photothermal therapy (PTT) via two near-infrared irradiations. The first irradiation can activate the photosensitizer bound in cancer cells internalized TCC@M to release Cas9 ribonucleoprotein (RNP) by photodynamic effect. The released Cas9 RNP then enters the nuclei directed by the fused nuclear localization sequence in Cas9 to cleave the heat shock protein (HSP) 90α gene, which greatly reduces the expression of HSP90α protein and thus effectively sensitizes cancer cells to heat, leading to enhanced PTT at a mild temperature (<45 °C) risen by Ti₃C₂ MXenes under the second irradiation. Simultaneously, TCC@M can produce fluorescence, photoacoustic, and thermal imaging signals to guide the optimal irradiation timing. The in vivo studies have demonstrated the spatiotemporally selective therapeutic efficacy of the designed TCC@M. This innovative approach presents an effective integration of gene regulation and enhanced PTT, exemplifying a precise cancer treatment strategy.
Collapse
Affiliation(s)
- Jing Jia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xiaobo Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Tian Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ying An
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xinrong Yan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Bin Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Chaoyi Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
14
|
Wu D, Sun X, Chen X. Chemo-optogenetic Dimerization Dissects Complex Biological Processes. SMALL METHODS 2025:e2401271. [PMID: 39815164 DOI: 10.1002/smtd.202401271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Light offers superior control in terms of high temporal precision, high spatial precision, and non-invasiveness for the regulation of cellular functions. In recent years, chemical biologists have adopted chemo-optogenetic dimerization approaches, such as photo-triggered chemical inducers of dimerization (pCIDs), as a general tool for spatiotemporal regulation of cellular functions. Traditional chemo-optogenetic dimerization triggers either a single ON or a single OFF of cellular activity. However, more sophisticated approaches are introduced in recent years. These include the ability to turn ON and OFF using different wavelengths of light, tools enabling multi-layer control of cellular activities, and nanobody-tethered photodimerizers. These advancements not only shed light on the study of ubiquitously existing multi-functional proteins but also create new opportunities for investigating complex cellular activity networks.
Collapse
Affiliation(s)
- Donglian Wu
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Xiaofeng Sun
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| | - Xi Chen
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences, Harbin Institute of Technology (HIT), Harbin, 150001, P. R. China
- Faculty of Life Science and Medicine, Harbin Institute of Technology, Harbin, 150001, P. R. China
| |
Collapse
|
15
|
Gong Y, Yang H, Ding C. NIR-photoactivatable DNA nanomachines for spatiotemporally controllable monitoring of microRNA-21 in living cells based on signal amplification strategy. Biosens Bioelectron 2025; 267:116755. [PMID: 39244838 DOI: 10.1016/j.bios.2024.116755] [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: 07/01/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Precise and spatiotemporally controllable analysis of microRNA-21 in living cells is crucial for accurate diagnosis and effective treatment of related diseases. Herein, a near-infrared (NIR)-photoactivatable DNA nanomachine (PUCNPs-NH2/PEG-ZL-DNA) was constructed for the precise analysis and diagnosis of microRNA-21 in tumor cells. Peanut-shaped upconversion nanoparticles (PUCNPs) were employed as the carriers and activators for the intelligent DNA probe, specifically enabling the cleavage of the photocleavable linker (PC-linker) from the hairpin DNA probe (Hp-Dzy) upon exposure to 808 nm irradiation. In the presence of the target microRNA-21, the locker DNA hybridized with microRNA-21 and the DNAzymes was freed to hybridize with the looped portion of the hairpin DNA (Hp-1). Mg2+ was employed as the cofactor, facilitating the precise cleavage of Hp-1, which triggered the restoration of fluorescence signals. Subsequently, DNAzymes exhibited the competency to selectively recognize and engage with additional Hp-1, and the fluorescence signals were effectively amplified by the recycling process. Consequently, the DNA nanomachine exhibited a linear response to microRNA-21 concentrations ranging from 0.5 nM to 1.0 μM, achieving a remarkable detection limit (LOD) of 1.19 nM under the optimal conditions. This strategy is realized through the integration of photocontrollable upconversion nanotechnology with the signal amplification approach, showing feasible prospects for spatiotemporally precise and highly sensitive monitoring of microRNA in tumor cells.
Collapse
Affiliation(s)
- Yan Gong
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China; College of Chemistry and Chemical Engineering, Huangshan University, Huangshan, 245041, PR China
| | - Huiwen Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Caifeng Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
16
|
Knapp F, Hogenkamp F, Paik SH, Jaeger KE, Pietruszka J, Drepper T. Synthesis and Application of Photocaged Isopropyl β-D-1-Thiogalactopyranoside for Light-Mediated Control of Bacterial Gene Expression. Methods Mol Biol 2025; 2840:133-148. [PMID: 39724349 DOI: 10.1007/978-1-0716-4047-0_10] [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: 12/28/2024]
Abstract
Photocaged compounds are chemical conjugates that are designed to release an active molecule upon exposure to light of a specific wavelength. In recent years, photocaged inducer molecules such as caged isopropyl β-D-1-thiogalactopyranoside (cIPTG) have been increasingly used as a powerful tool for light-driven gene expression in bacteria, allowing researchers to precisely and noninvasively tune the expression of specific target genes. In this chapter, we present a guideline for the synthesis of 6-nitropiperonyl photocaged IPTG (NP-cIPTG) as well as its in vivo application as an optochemical on-switch of gene transcription in Escherichia coli and other bacteria.
Collapse
Affiliation(s)
- Fabienne Knapp
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Fabian Hogenkamp
- Institute of Bioorganic Chemistry, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Seung-Hyun Paik
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Karl-Erich Jaeger
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany
- Institute of Bio- and Geosciences IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Jörg Pietruszka
- Institute of Bioorganic Chemistry, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany.
- Institute of Bio- and Geosciences IBG-1: Biotechnology, Forschungszentrum Jülich GmbH, Jülich, Germany.
| | - Thomas Drepper
- Institute of Molecular Enzyme Technology, Heinrich Heine University Düsseldorf, Forschungszentrum Jülich GmbH, Jülich, Germany.
| |
Collapse
|
17
|
Das S, Pal P, Biswas R, Baitalik S. Design of Near-Infrared Emissive Molecular Switches Based on Stilbene-Appended Cyclometalated Bimetallic Ru(II)-Terpyridine Complexes. Inorg Chem 2024; 63:23725-23741. [PMID: 39614808 DOI: 10.1021/acs.inorgchem.4c03854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
In the present study, we have synthesized and thoroughly characterized two Ru(II) dimers with compositions [(ttpy)Ru(tpvpt')Ru(ttpy)](ClO4)3 and [(ttpy)Ru(t'pvpvpt')Ru(ttpy)](ClO4)2 incorporating phenylene-vinylene-substituted terpyridine bridging ligands capable of coordinating in both an NNN- and cyclometalated NNC-fashion. The complexes display strong absorption across the entire UV-vis spectral domain and exhibit luminescence in the NIR region (820-850 nm). The N atoms in the outer coordination sphere were employed for alteration of the photoredox behaviors of the complexes via acid-base equilibria. Decoordination of the Ru-C bonds in the presence of acid, followed by recoordination in the presence of base and heat, is also possible. Additionally, the phenylene-vinylene units in the bridging ligands facilitate trans → cis and trans-trans → trans-cis isomerization under visible light irradiation. The reverse isomerization (cis → trans and trans-cis → trans-trans) is also achieved upon UV light irradiation. Thus, "on-off" and "off-on" emission switching is facilitated through a judicious choice of external stimuli like acid, base, temperature, or light of particular wavelengths. Interestingly, the rate of photoswitching is significantly faster in the presence of acid compared to that in the absence of acid. DFT and TD-DFT calculations were also conducted to clearly visualize the electronic environment around the complex backbone and also for accurate assignment of the absorption and emission bands.
Collapse
Affiliation(s)
- Soumi Das
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| | - Poulami Pal
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & B Raja S C Mullick Road, Kolkata 700032, India
| | - Raju Biswas
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| | - Sujoy Baitalik
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
18
|
Sakovina L, Vokhtantsev I, Akhmetova E, Vorobyeva M, Vorobjev P, Zharkov DO, Novopashina D. Photocleavable Guide crRNAs for a Light-Controllable CRISPR/Cas9 System. Int J Mol Sci 2024; 25:12392. [PMID: 39596457 PMCID: PMC11594570 DOI: 10.3390/ijms252212392] [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: 10/15/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
The design of controllable and precise RNA-targeted CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats) systems is an important problem of modern molecular biology and genetic technology. Herein, we have designed a series of photocleavable guide CRISPR RNAs (crRNA) and their 2'-modified (2'-fluoro and locked nucleic acid) analogs containing one or two 1-(2-nitrophenyl)-1,2-ethanediol photolabile linkers (PL). We have demonstrated that these crRNAs can be destroyed by relatively mild UVA irradiation with the rate constants 0.24-0.77 min-1 and that the photocleavage markedly slows down the action of Cas9 nuclease in the model in vitro system. Two PLs provide more rapid crRNA destruction than a single linker. PLs in the crRNA structure improve the specificity of DNA cleavage by Cas9 nuclease for the fully complementary target. The application of photocleavable crRNA in CRISPR/Cas9 genome editing permits the system to be switched off in a spatiotemporally controlled manner, thus alleviating its off-target effects.
Collapse
Affiliation(s)
- Lubov Sakovina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ivan Vokhtantsev
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Elizaveta Akhmetova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Mariya Vorobyeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
| | - Pavel Vorobjev
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
| | - Dmitry O. Zharkov
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Darya Novopashina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (L.S.); (I.V.); (E.A.); (M.V.); (P.V.); (D.O.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
19
|
Maller C, Marouda E, Köhn M. Photo-Claisen Rearrangement in a Coumarin-Caged Peptide Leads to a Surprising Enzyme Hyperactivation. Chembiochem 2024; 25:e202400561. [PMID: 39172538 DOI: 10.1002/cbic.202400561] [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: 06/28/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Protein phosphatase-1 (PP1) is a ubiquitous enzyme that counteracts hundreds of kinases in cells. PP1 interacts with regulatory proteins via an RVxF peptide motif that binds to a hydrophobic groove on the enzyme. PP1-disrupting peptides (PDPs) compete with these regulatory proteins, leading to the release of the active PP1 subunit and promoting substrate dephosphorylation. Building on previous strategies employing the ortho-nitrobenzyl (o-Nb) group as a photocage to control PDP activity, we introduced coumarin derivatives into a PDP via an ether bond to explore their effects on PP1 activity. Surprisingly, our study revealed that the coumarin-caged peptides (PDP-DEACM and PDP-CM) underwent a photo-Claisen rearrangement, resulting in an unexpected hyperactivation of PP1. Our findings underscore the importance of linker design in controlling uncaging efficiency of photocages and highlight the need for comprehensive in vitro analysis before cellular experiments.
Collapse
Affiliation(s)
- Corina Maller
- Signalling Research Centres BIOSS and CIBSS, Faculty of Biology, University of Freiburg, Schänzlestrasse 18, Freiburg, 79104, Germany
- Faculty of Chemistry and Pharmacy, Hermann-Staudinger Graduate School, University of Freiburg, Hebelstrasse 27, Freiburg, 79104, Germany
| | - Eirini Marouda
- Signalling Research Centres BIOSS and CIBSS, Faculty of Biology, University of Freiburg, Schänzlestrasse 18, Freiburg, 79104, Germany
| | - Maja Köhn
- Signalling Research Centres BIOSS and CIBSS, Faculty of Biology, University of Freiburg, Schänzlestrasse 18, Freiburg, 79104, Germany
| |
Collapse
|
20
|
Weng W, Zhang P, Pan Z. Potent Inhibition and Rapid Photoactivation of Endogenous Bruton's Tyrosine Kinase Activity in Native Cells via Opto-Covalent Modulators. J Am Chem Soc 2024; 146:28717-28727. [PMID: 39388725 DOI: 10.1021/jacs.4c06459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Naturally, kinases exert their activities in a highly regulated fashion. A number of ingenious approaches have been developed to artificially control kinase activity by external stimuli, such as the incorporation of unnatural amino acids or the fusion of additional protein domains; however, methods that directly modulate endogenous kinases in native cells are lacking. Herein, we present a facile and potent method that takes advantage of recent developments in targeted covalent inhibitors and rapid light-mediated uncaging chemistry. Using an important drug target, Bruton's tyrosine kinase (BTK), as an example, these opto-covalent modulators successfully blocked the activity of endogenous BTK in native cells after simple incubation and washout steps. However, upon a few minutes of light irradiation, BTK activity was cleanly restored, and could be blocked again by conventional inhibitors. Promisingly, this photoactivation strategy easily worked in human peripheral blood mononuclear cells (hPBMCs).
Collapse
Affiliation(s)
- Weizhi Weng
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518055, China
| | - Ping Zhang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518055, China
| | - Zhengying Pan
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen 518055, China
| |
Collapse
|
21
|
Hildebrandt M, Koshimizu M, Asada Y, Fukumitsu K, Ohkuma M, Sang N, Nakano T, Kunikata T, Okazaki K, Kawaguchi N, Yanagida T, Lian L, Zhang J, Yamashita T. Comparative Validation of Scintillator Materials for X-Ray-Mediated Neuronal Control in the Deep Brain. Int J Mol Sci 2024; 25:11365. [PMID: 39518918 PMCID: PMC11547033 DOI: 10.3390/ijms252111365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
When exposed to X-rays, scintillators emit visible luminescence. X-ray-mediated optogenetics employs scintillators for remotely activating light-sensitive proteins in biological tissue through X-ray irradiation. This approach offers advantages over traditional optogenetics, allowing for deeper tissue penetration and wireless control. Here, we assessed the short-term safety and efficacy of candidate scintillator materials for neuronal control. Our analyses revealed that lead-free halide scintillators, such as Cs3Cu2I5, exhibited significant cytotoxicity within 24 h and induced neuroinflammatory effects when injected into the mouse brain. In contrast, cerium-doped gadolinium aluminum gallium garnet (Ce:GAGG) nanoparticles showed no detectable cytotoxicity within the same period, and injection into the mouse brain did not lead to observable neuroinflammation over four weeks. Electrophysiological recordings in the cerebral cortex of awake mice showed that X-ray-induced radioluminescence from Ce:GAGG nanoparticles reliably activated 45% of the neuronal population surrounding the implanted particles, a significantly higher activation rate than europium-doped GAGG (Eu:GAGG) microparticles, which activated only 10% of neurons. Furthermore, we established the cell-type specificity of this technique by using Ce:GAGG nanoparticles to selectively stimulate midbrain dopamine neurons. This technique was applied to freely behaving mice, allowing for wireless modulation of place preference behavior mediated by midbrain dopamine neurons. These findings highlight the unique suitability of Ce:GAGG nanoparticles for X-ray-mediated optogenetics. The deep tissue penetration, short-term safety, wireless neuronal control, and cell-type specificity of this system offer exciting possibilities for diverse neuroscience applications and therapeutic interventions.
Collapse
Affiliation(s)
- Mercedes Hildebrandt
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (M.H.); (K.F.); (M.O.); (N.S.)
| | - Masanori Koshimizu
- Research Institute of Electronics, Shizuoka University, Hamamatsu 432-8011, Shizuoka, Japan;
| | - Yasuki Asada
- Faculty of Radiological Technology, School of Medical Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| | - Kansai Fukumitsu
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (M.H.); (K.F.); (M.O.); (N.S.)
- International Center for Brain Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| | - Mahito Ohkuma
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (M.H.); (K.F.); (M.O.); (N.S.)
| | - Na Sang
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (M.H.); (K.F.); (M.O.); (N.S.)
| | - Takashi Nakano
- International Center for Brain Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
- Department of Computational Biology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Toshiaki Kunikata
- Division of Materials Science, Nara Institute of Science and Technology (NAIST), Ikoma 630-0192, Nara, Japan; (T.K.); (K.O.); (N.K.); (T.Y.)
| | - Kai Okazaki
- Division of Materials Science, Nara Institute of Science and Technology (NAIST), Ikoma 630-0192, Nara, Japan; (T.K.); (K.O.); (N.K.); (T.Y.)
| | - Noriaki Kawaguchi
- Division of Materials Science, Nara Institute of Science and Technology (NAIST), Ikoma 630-0192, Nara, Japan; (T.K.); (K.O.); (N.K.); (T.Y.)
| | - Takayuki Yanagida
- Division of Materials Science, Nara Institute of Science and Technology (NAIST), Ikoma 630-0192, Nara, Japan; (T.K.); (K.O.); (N.K.); (T.Y.)
| | - Linyuan Lian
- Key Laboratory of Materials Physics of Ministry of Education, School of Physics, Zhengzhou University, Zhengzhou 450052, China;
| | - Jianbing Zhang
- School of Integrated Circuits, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Takayuki Yamashita
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (M.H.); (K.F.); (M.O.); (N.S.)
- International Center for Brain Science, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| |
Collapse
|
22
|
Gelabert R, Moreno M, Lluch JM. Effect of Leaving Centrosymmetric Character on Spectral Properties in Mono-, Bi-, and Triphotonic Absorption Spectroscopies. ACS OMEGA 2024; 9:41968-41977. [PMID: 39398148 PMCID: PMC11465556 DOI: 10.1021/acsomega.4c06922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 08/23/2024] [Indexed: 10/15/2024]
Abstract
Numerical simulations of the absorption bands of photoswitch E-o-tetrafluoroazobenzene in DMSO solution under one-, two-, and three-photon absorption conditions combined with the analysis of the behavior of transition probability under distortion of planarity reveal many similarities between the mono- and triphoton spectroscopic behaviors with a two-photon spectrum being set apart. The position of the absorption peak for the studied nπ* and ππ* transitions appears shifted to lower energies (longer wavelengths) than the conventional estimate based on vertical excitation from the ground-state potential energy minimum.
Collapse
Affiliation(s)
- Ricard Gelabert
- Departament
de Química, Universitat Autònoma
de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Miquel Moreno
- Departament
de Química, Universitat Autònoma
de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - José M. Lluch
- Departament
de Química, Universitat Autònoma
de Barcelona, 08193 Bellaterra, Barcelona, Spain
- Institut
de Biotecnologia i de Biomedicina, Universitat
Autònoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
23
|
Löffler M, Frühschulz S, Rockel Z, Pečak M, Tampé R, Wieneke R. Antigen Delivery Controlled by an On-Demand Photorelease. Angew Chem Int Ed Engl 2024; 63:e202405035. [PMID: 38818622 DOI: 10.1002/anie.202405035] [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: 03/13/2024] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
To eliminate infected and cancerous cells, antigen processing and presentation play a pivotal role through the recognition of antigenic peptides displayed on Major Histocompatibility Complex class I (MHC I) molecules. Here, we developed a photostimulated antigen release system that enables the temporal inception of antigen flux. Simple and effective photocaging of the human immunodeficiency virus (HIV)-Nef73-derived epitope, a representative high-affinity MHC I ligand, was provided by steric hindrance to block the recognition by the transporter associated with antigen processing (TAP) in the peptide loading complex (PLC). In response to light, a heteronomous release of antigens and subsequent translocation in various scenarios is demonstrated, including a TAP-related ATP-binding cassette (ABC) transporter reconstituted in liposomes and the native PLC in the endoplasmic reticulum (ER) membrane of human cells. The photochemically induced 'burst' of antigens opens new opportunities for a mechanistic analysis of the antigen translocation machinery and will help to provide insights into antigen processing pathways via an on-demand, subcellular pulse-chase release of antigens.
Collapse
Affiliation(s)
- Max Löffler
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Stefan Frühschulz
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Zoe Rockel
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Matija Pečak
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| |
Collapse
|
24
|
Wang F, Liu Z, Liu Y, Zhang J, Xu W, Liu B, Sun Z, Chu H. A Spatiotemporally Controlled Gene-Regulation Strategy for Combined Tumor Therapy Based on Upconversion Hybrid Nanosystem. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405640. [PMID: 39207039 PMCID: PMC11515897 DOI: 10.1002/advs.202405640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/09/2024] [Indexed: 09/04/2024]
Abstract
The lack of precise spatiotemporal gene modulation and therapy impedes progress in medical applications. Herein, a 980 nm near-infrared (NIR) light-controlled nanoplatform, namely URMT, is developed, which can allow spatiotemporally controlled photodynamic therapy and trigger the enzyme-activated gene expression regulation in tumors. URMT is constructed by engineering an enzyme-activatable antisense oligonucleotide, which combined with an upconversion nanoparticle (UCNP)-based photodynamic nanosystem, followed by the surface functionalization of triphenylphosphine (TPP), a mitochondria-targeting ligand. URMT allows for the 980 nm NIR light-activated generation of reactive oxygen species, which can induce the translocation of a DNA repair enzyme (namely apurinic/apyrimidinic endonuclease 1, APE1) from the nucleus to mitochondria. APE1 can recognize the basic apurinic/apyrimidinic (AP) sites in DNA double-strands and perform cleavage, thereby releasing the functional single-strands for gene regulation. Overall, an augmented antitumor effect is observed due to NIR light-controlled mitochondrial damage and enzyme-activated gene regulation. Altogether, the approach reported in this study offers high spatiotemporal precision and shows the potential to achieve precise and specific gene regulation for targeted tumor treatment.
Collapse
Affiliation(s)
- Fang Wang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Zechao Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Yuechen Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Jiayi Zhang
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Weizhe Xu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Bei Liu
- College of ScienceMinzu University of China27 Zhongguancun South AvenueBeijing100081China
| | - Zhaogang Sun
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| | - Hongqian Chu
- Translational Medicine CenterBeijing Chest HospitalCapital Medical University9 Beiguan StreetBeijing101149China
| |
Collapse
|
25
|
Choi JH, Kim S, Kang OY, Choi SY, Hyun JY, Lee HS, Shin I. Selective fluorescent labeling of cellular proteins and its biological applications. Chem Soc Rev 2024; 53:9446-9489. [PMID: 39109465 DOI: 10.1039/d4cs00094c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Proteins, which are ubiquitous in cells and critical to almost all cellular functions, are indispensable for life. Fluorescence imaging of proteins is key to understanding their functions within their native milieu, as it provides insights into protein localization, dynamics, and trafficking in living systems. Consequently, the selective labeling of target proteins with fluorophores has emerged as a highly active research area, encompassing bioorganic chemistry, chemical biology, and cell biology. Various methods for selectively labeling proteins with fluorophores in cells and tissues have been established and are continually being developed to visualize and characterize proteins. This review highlights research findings reported since 2018, with a focus on the selective labeling of cellular proteins with small organic fluorophores and their biological applications in studying protein-associated biological events. We also discuss the strengths and weaknesses of each labeling approach for their utility in living systems.
Collapse
Affiliation(s)
- Joo Hee Choi
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Sooin Kim
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - On-Yu Kang
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Seong Yun Choi
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Hyun Soo Lee
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| |
Collapse
|
26
|
Porro A, Armano E, Brandalise F, Appiani R, Beltrame M, Saponaro A, Dallanoce C, Nakajo K, Ryu K, Leone R, Thiel G, Pallavicini M, Moroni A, Bolchi C. A Photoactivatable Version of Ivabradine Enables Light-Induced Block of HCN Current In Vivo. J Med Chem 2024; 67:16209-16221. [PMID: 39238314 DOI: 10.1021/acs.jmedchem.4c01047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Therapeutic drugs, whose bioactivity is hindered by a photoremovable cage, offer the advantage of spatiotemporal confinement of their action to the target diseased tissue with improved bioavailability and efficacy. Here, we have applied such an approach to ivabradine (IVA), a bradycardic agent indicated for angina pectoris and heart failure, acting as a specific HCN channel blocker. To overcome the side effects due to its poor discrimination among HCN channel subtypes (HCN1-4), we prepared a caged version of IVA linked to a photocleavable bromoquinolinylmethyl group (BHQ-IVA). We show that upon illumination with blue light (440 nm), BHQ-IVA releases active IVA that blocks HCN channel currents in vitro and exerts a bradycardic effect in vivo. Both BHQ-IVA and the cage are inactive. Caging is stable in aqueous medium and in the dark, and it does not impair aqueous solubility and cell permeation, indispensable for IVA activity. This approach allows for bypassing the poor subtype-specificity of IVA, expanding its prescription to HCN-related diseases besides cardiac.
Collapse
Affiliation(s)
- Alessandro Porro
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Edoardo Armano
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | | | - Rebecca Appiani
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Monica Beltrame
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Andrea Saponaro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano 20133, Italy
| | - Clelia Dallanoce
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Koichi Nakajo
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Kawachi District, Shimotsuke 329-0498, Japan
| | - Kaei Ryu
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Kawachi District, Shimotsuke 329-0498, Japan
| | - Roberta Leone
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Gerhard Thiel
- Department of Biology, TU-Darmstadt, Darmstadt 64287, Germany
| | - Marco Pallavicini
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milano 20133, Italy
| | - Cristiano Bolchi
- Department of Pharmaceutical Sciences, University of Milan, Milano 20133, Italy
| |
Collapse
|
27
|
Mateus M, Hammill ML, Simmons DBD, Desaulniers JP. In Vivo Injection of Reversible Optically Controlled Short Interfering RNA into Japanese Medaka Embryos ( Oryzias latipes) to Regulate Gene Silencing. ACS Chem Biol 2024; 19:1904-1909. [PMID: 39162696 PMCID: PMC11421425 DOI: 10.1021/acschembio.4c00290] [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: 04/25/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Photoswitchable ortho-functionalized tetrafluorinated azobenzene-modified siRNAs (F-azo-siRNAs) were synthesized using solid-phase phosphoramidite chemistry. The activity of an F-azo-siRNA targeting enhanced green fluorescence protein (eGFP) in transgenic (Tg) Japanese Medaka (Oryzias latipes) was reversibly photocontrolled with blue (470 nm) and green (530 nm) light, to activate and inactivate the siRNA, respectively. This study highlights the first reversible in vivo study with photoswitchable siRNA. Controlling siRNA function reversibly in vivo could open new opportunities for biotech research to better understand gene function and cellular mechanisms.
Collapse
Affiliation(s)
- Makenzie Mateus
- Faculty of Science, Ontario
Tech University, 2000
Simcoe Street North, Oshawa ON L1G 0C5, Canada
| | - Matthew L. Hammill
- Faculty of Science, Ontario
Tech University, 2000
Simcoe Street North, Oshawa ON L1G 0C5, Canada
| | - Denina B. D. Simmons
- Faculty of Science, Ontario
Tech University, 2000
Simcoe Street North, Oshawa ON L1G 0C5, Canada
| | | |
Collapse
|
28
|
Rigault D, Nizard P, Daniel J, Blanćhard-Desce M, Deprez E, Tauc P, Dhimane H, Dalko PI. Triphenylamine Sensitized 8-Dimethylaminoquinoline: An Efficient Two-Photon Caging Group for Intracellular Delivery. Chemistry 2024; 30:e202401289. [PMID: 38959014 DOI: 10.1002/chem.202401289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Triphenylamine-sensitized 8-dimethylaminoquinoline (TAQ) probes showed fair two-photon absorption and fragmentation cross sections in releasing kainate and GABA ligands. The water-soluble PEG and TEG-analogs allowed cell internalization and efficient light-gated liberation of the rhodamine reporter under UV and two-photon (NIR) irradiation conditions.
Collapse
Affiliation(s)
- Delphine Rigault
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Philippe Nizard
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Jonathan Daniel
- Institut des Sciences Moleéculaires, Universite de Bordeaux, Bâtiment A12 351 Cours de la Libération, 33405, TALENCE cedex, France
| | - Mireille Blanćhard-Desce
- Institut des Sciences Moleéculaires, Universite de Bordeaux, Bâtiment A12 351 Cours de la Libération, 33405, TALENCE cedex, France
| | - Eric Deprez
- LBPA, ENS Paris-Saclay, CNRS, Université Paris-Saclay, Gif-sur-Yvette, 91190, France
| | - Patrick Tauc
- LBPA, ENS Paris-Saclay, CNRS, Université Paris-Saclay, Gif-sur-Yvette, 91190, France
| | - Hamid Dhimane
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Peter I Dalko
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| |
Collapse
|
29
|
Klimezak M, Chaud J, Brion A, Bolze F, Frisch B, Heurtault B, Kichler A, Specht A. Triplet-Triplet Annihilation Upconversion-Based Photolysis: Applications in Photopharmacology. Adv Healthc Mater 2024; 13:e2400354. [PMID: 38613491 DOI: 10.1002/adhm.202400354] [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: 01/29/2024] [Revised: 04/04/2024] [Indexed: 04/15/2024]
Abstract
The emerging field of photopharmacology is a promising chemobiological methodology for optical control of drug activities that could ultimately solve the off-target toxicity outside the disease location of many drugs for the treatment of a given pathology. The use of photolytic reactions looks very attractive for a light-activated drug release but requires to develop photolytic reactions sensitive to red or near-infrared light excitation for better tissue penetration. This review will present the concepts of triplet-triplet annihilation upconversion-based photolysis and their recent in vivo applications for light-induced drug delivery using photoactivatable nanoparticles.
Collapse
Affiliation(s)
- Maxime Klimezak
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST), Équipe Nanoparticules Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, Illkirch Cedex, F-67401, France
| | - Juliane Chaud
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST), Équipe Nanoparticules Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, Illkirch Cedex, F-67401, France
- Inserm UMR_S 1121, EMR 7003 CNRS, Université de Strasbourg, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, 1 rue Eugène Boeckel, Strasbourg, F-67000, France
| | - Anaïs Brion
- Inserm UMR_S 1121, EMR 7003 CNRS, Université de Strasbourg, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, 1 rue Eugène Boeckel, Strasbourg, F-67000, France
| | - Frédéric Bolze
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST), Équipe Nanoparticules Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, Illkirch Cedex, F-67401, France
| | - Benoit Frisch
- Inserm UMR_S 1121, EMR 7003 CNRS, Université de Strasbourg, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, 1 rue Eugène Boeckel, Strasbourg, F-67000, France
| | - Béatrice Heurtault
- Inserm UMR_S 1121, EMR 7003 CNRS, Université de Strasbourg, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, 1 rue Eugène Boeckel, Strasbourg, F-67000, France
| | - Antoine Kichler
- Inserm UMR_S 1121, EMR 7003 CNRS, Université de Strasbourg, Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, 1 rue Eugène Boeckel, Strasbourg, F-67000, France
| | - Alexandre Specht
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique (CBST), Équipe Nanoparticules Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, Illkirch Cedex, F-67401, France
| |
Collapse
|
30
|
Lu Q, Sun Y, Liang Z, Zhang Y, Wang Z, Mei Q. Nano-optogenetics for Disease Therapies. ACS NANO 2024; 18:14123-14144. [PMID: 38768091 DOI: 10.1021/acsnano.4c00698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Optogenetic, known as the method of 21 centuries, combines optic and genetic engineering to precisely control photosensitive proteins for manipulation of a broad range of cellular functions, such as flux of ions, protein oligomerization and dissociation, cellular intercommunication, and so on. In this technique, light is conventionally delivered to targeted cells through optical fibers or micro light-emitting diodes, always suffering from high invasiveness, wide-field illumination facula, strong absorption, and scattering by nontargeted endogenous substance. Light-transducing nanomaterials with advantages of high spatiotemporal resolution, abundant wireless-excitation manners, and easy functionalization for recognition of specific cells, recently have been widely explored in the field of optogenetics; however, there remain a few challenges to restrain its clinical applications. This review summarized recent progress on light-responsive genetically encoded proteins and the myriad of activation strategies by use of light-transducing nanomaterials and their disease-treatment applications, which is expected for sparking helpful thought to push forward its preclinical and translational uses.
Collapse
Affiliation(s)
- Qi Lu
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yaru Sun
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Zhengbing Liang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yi Zhang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Zhigang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Qingsong Mei
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| |
Collapse
|
31
|
Ma J, Wehrle J, Frank D, Lorenzen L, Popp C, Driever W, Grosse R, Jessen HJ. Intracellular delivery and deep tissue penetration of nucleoside triphosphates using photocleavable covalently bound dendritic polycations. Chem Sci 2024; 15:6478-6487. [PMID: 38699261 PMCID: PMC11062083 DOI: 10.1039/d3sc05669d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/15/2024] [Indexed: 05/05/2024] Open
Abstract
Nucleoside triphosphates (NTPs) are essential in various biological processes. Cellular or even organismal controlled delivery of NTPs would be highly desirable, yet in cellulo and in vivo applications are hampered owing to their negative charge leading to cell impermeability. NTP transporters or NTP prodrugs have been developed, but a spatial and temporal control of the release of the investigated molecules remains challenging with these strategies. Herein, we describe a general approach to enable intracellular delivery of NTPs using covalently bound dendritic polycations, which are derived from PAMAM dendrons and their guanidinium derivatives. By design, these modifications are fully removable through attachment on a photocage, ready to deliver the native NTP upon irradiation enabling spatiotemporal control over nucleotide release. We study the intracellular distribution of the compounds depending on the linker and dendron generation as well as side chain modifications. Importantly, as the polycation is bound covalently, these molecules can also penetrate deeply into the tissue of living organisms, such as zebrafish.
Collapse
Affiliation(s)
- Jiahui Ma
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Freiburg Albertstr. 21 79104 Freiburg Germany
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
| | - Johanna Wehrle
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
- Faculty of Biology, University of Freiburg Hauptstr. 1 79104 Freiburg Germany
| | - Dennis Frank
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg Albertstr. 25 79104 Freiburg Germany
| | - Lina Lorenzen
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg Albertstr. 25 79104 Freiburg Germany
| | - Christoph Popp
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Freiburg Albertstr. 21 79104 Freiburg Germany
| | - Wolfgang Driever
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
- Faculty of Biology, University of Freiburg Hauptstr. 1 79104 Freiburg Germany
| | - Robert Grosse
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg Albertstr. 25 79104 Freiburg Germany
| | - Henning J Jessen
- Institute of Organic Chemistry, Faculty of Chemistry and Pharmacy, University of Freiburg Albertstr. 21 79104 Freiburg Germany
- CIBSS-Centre for Integrative Biological Signaling Studies, University of Freiburg 79104 Freiburg Germany
| |
Collapse
|
32
|
Li L, Li M. Modular Engineering of Aptamer-Based Nanobiotechnology for Conditional Control of ATP Sensing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2302972. [PMID: 38009471 DOI: 10.1002/adma.202302972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/24/2023] [Indexed: 11/29/2023]
Abstract
Dynamic changes of intracellular, extracellular, and subcellular adenosine triphosphates (ATPs) have fundamental interdependence with the physio-pathological states of cells. Spatially selective in situ imaging of such ATP dynamics offers valuable mechanistic insights into the related biological activities. Despite significant advances in the design of aptamer sensors for ATP detection, the dearth of methods that enable precise ATP imaging in specific cellular locations remains a challenge in this field. This review focuses on the modular engineering of regulatable sensing technology via the integration of aptamer probe designs with advanced functional nanomaterials, allowing conditional control of ATP sensing and imaging with high spatial precision from subcellular organelles to living animals. Highlighting the recent advances in the design of photo-triggered nanosensors for spatiotemporally controlled ATP imaging, endogenously-triggered ATP sensing in a cell-selective manner, and spatially-controlled nanodevices for ATP imaging in specific organelles and extracellular microenvironments. Emphasis will be put on elucidating the principles of how nanotechnology can be applied to regulate the spatial precision of aptamer-based ATP sensing activities. The authors envision that this perspective provides insights into the engineering of aptamer-based nanobiotechnology for opening new frontiers in precise molecular sensing and other bio-applications.
Collapse
Affiliation(s)
- Lele Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyuan Li
- School of Chemistry and Biological Engineering, Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing, 100083, China
| |
Collapse
|
33
|
Bardhan A, Brown W, Albright S, Tsang M, Davidson LA, Deiters A. Direct Activation of Nucleobases with Small Molecules for the Conditional Control of Antisense Function. Angew Chem Int Ed Engl 2024; 63:e202318773. [PMID: 38411401 DOI: 10.1002/anie.202318773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 02/28/2024]
Abstract
Conditionally controlled antisense oligonucleotides provide precise interrogation of gene function at different developmental stages in animal models. Only one example of small molecule-induced activation of antisense function exist. This has been restricted to cyclic caged morpholinos that, based on sequence, can have significant background activity in the absence of the trigger. Here, we provide a new approach using azido-caged nucleobases that are site-specifically introduced into antisense morpholinos. The caging group design is a simple azidomethylene (Azm) group that, despite its very small size, efficiently blocks Watson-Crick base pairing in a programmable fashion. Furthermore, it undergoes facile decaging via Staudinger reduction when exposed to a small molecule phosphine, generating the native antisense oligonucleotide under conditions compatible with biological environments. We demonstrated small molecule-induced gene knockdown in mammalian cells, zebrafish embryos, and frog embryos. We validated the general applicability of this approach by targeting three different genes.
Collapse
Affiliation(s)
- Anirban Bardhan
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Wes Brown
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Savannah Albright
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Michael Tsang
- Department of Cell Biology, Center for Integrative Organ Systems., University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Lance A Davidson
- Department of Bioengineering, Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| |
Collapse
|
34
|
Mandal M, Scerbo P, Coghill I, Riou JF, Bochet CG, Ducos B, Bensimon D, Le Saux T, Aujard I, Jullien L. Caged Dexamethasone to Photo-control the Development of Embryos through Activation of the Glucocorticoid Receptor. Chemistry 2024; 30:e202400579. [PMID: 38350020 DOI: 10.1002/chem.202400579] [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: 02/12/2024] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/15/2024]
Abstract
Efficient tools for controlling molecular functions with exquisite spatiotemporal resolution are much in demand to investigate biological processes in living systems. Here we report an easily synthesized caged dexamethasone for photo-activating cytoplasmic proteins fused to the glucocorticoid receptor. In the dark, it is stable in vitro as well as in vivo in both zebrafish (Danio rerio) and Xenopus sp, two significant models of vertebrates. In contrast, it liberates dexamethasone upon UV illumination, which has been harnessed to interfere with developmental steps in embryos of these animals. Interestingly, this new system is biologically orthogonal to the one for photo-activating proteins fused to the estrogen ERT receptor, which brings great prospect for activating two distinct proteins down to the single cell level.
Collapse
Affiliation(s)
- Mrinal Mandal
- PASTEUR, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Pierluigi Scerbo
- Laboratoire de Physique de l'Ecole Normale Supérieure, Paris Sciences Lettres University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005, Paris, France
| | - Ian Coghill
- PASTEUR, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Jean-François Riou
- Laboratory of Developmental Biology, Institute of Biology Paris-Seine, Sorbonne University, CNRS, 9, Quai Saint-Bernard, 75252, Paris, Cedex 05, France
| | - Christian G Bochet
- Department of Chemistry, University of Fribourg, 9 Ch. du Musée, CH-1700, Fribourg, Switzerland
| | - Bertrand Ducos
- Laboratoire de Physique de l'Ecole Normale Supérieure, Paris Sciences Lettres University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005, Paris, France
- High Throughput qPCR Core Facility, Ecole Normale Supérieure, Paris Sciences Lettres University, 46 Rue d'Ulm, 75005, Paris, France
| | - David Bensimon
- Laboratoire de Physique de l'Ecole Normale Supérieure, Paris Sciences Lettres University, Sorbonne Université, Université de Paris, Centre National de la Recherche Scientifique, 24 Rue Lhomond, 75005, Paris, France
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - Thomas Le Saux
- PASTEUR, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Isabelle Aujard
- PASTEUR, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Ludovic Jullien
- PASTEUR, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| |
Collapse
|
35
|
Breton-Patient C, Billotte S, Duchambon P, Fontaine G, Bombard S, Piguel S. Light-Activatable Photocaged UNC2025 for Triggering TAM Kinase Inhibition in Bladder Cancer. Chembiochem 2024; 25:e202300855. [PMID: 38363151 DOI: 10.1002/cbic.202300855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/17/2024]
Abstract
Photopharmacology is an emerging field that utilizes photo-responsive molecules to enable control over the activity of a drug using light. The aim is to limit the therapeutic action of a drug at the level of diseased tissues and organs. Considering the well-known implications of protein kinases in cancer and the therapeutic issues associated with protein kinase inhibitors, the photopharmacology is seen as an innovative and alternative solution with great potential in oncology. In this context, we developed the first photocaged TAM kinase inhibitors based on UNC2025, a first-in-class small molecule kinase inhibitor. These prodrugs showed good stability in biologically relevant buffer and rapid photorelease of the photoremovable protecting group upon UV-light irradiation (<10 min.). These light-activatable prodrugs led to a 16-fold decrease to a complete loss of kinase inhibition, depending on the protein and the position at which the coumarin-type phototrigger was introduced. The most promising candidate was the N,O-dicaged compound, showing the superiority of having two photolabile protecting groups on UNC2025 for being entirely inactive on TAM kinases. Under UV-light irradiation, the N,O-dicaged compound recovered its inhibitory potency in enzymatic assays and displayed excellent antiproliferative activity in RT112 cell lines.
Collapse
Affiliation(s)
- Chloé Breton-Patient
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sébastien Billotte
- Université Paris-Saclay, Faculté de Pharmacie CNRS UMR 8076, 91400, Orsay, France
| | - Patricia Duchambon
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Gaëlle Fontaine
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sophie Bombard
- Institut Curie, Université PSL CNRS UMR9187, Inserm U119, 91400, Orsay, France
- Université Paris-Saclay CNRS UMR9187, Inserm U119, 91400, Orsay, France
| | - Sandrine Piguel
- Université Paris-Saclay, Faculté de Pharmacie CNRS UMR 8076, 91400, Orsay, France
| |
Collapse
|
36
|
Das S, Bar M, Ganguly T, Baitalik S. Control of Photoisomerization Kinetics via Multistage Switching in Bimetallic Ru(II)-Terpyridine Complexes. Inorg Chem 2024; 63:6600-6615. [PMID: 38557011 DOI: 10.1021/acs.inorgchem.3c04255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
In this study, we carried out detailed experimental and theoretical investigation on photophysical, electrochemical, and photoisomerization behaviors of a new array of luminescent binuclear Ru(II) complexes derived from a phenylene-vinylene-substituted terpyridyl ligand possessing RT lifetimes within 60.3-410.5 ns. The complexes experienced trans-to-cis isomerization in MeCN on irradiation with visible light, accompanied by significant changes in their absorption and emission spectral profiles. The reverse cis-to-trans process is also possible with the use of ultraviolet (UV) light. On conversion from trans to cis isomers, the emission intensity increases substantially, while for the reverse process, luminescence quenching occurs. Thus, "off-on" and "on-off" emission switching is facilitated upon treatment with visible and UV light alternatively. By the use of chemical oxidants (ceric ammonium nitrate and potassium permanganate) and reductants (metallic sodium) as well as light of appropriate wavelengths, multistate switching phenomena involving reversible oxidation-reduction and trans-cis isomerization have been achieved. Interestingly, the rate of this multistate photoswitching process becomes much faster compared to only two-state trans-cis isomerization of these complexes. Density functional theory (DFT) and time-dependent-DFT (TD-DFT) calculations are also performed to obtain a clear picture of the electronic environment of the complexes and also for the appropriate assignment of absorption and emission spectral bands.
Collapse
Affiliation(s)
- Soumi Das
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| | - Manoranjan Bar
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| | - Tanusree Ganguly
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| | - Sujoy Baitalik
- Department of Chemistry, Inorganic Chemistry Section, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
37
|
Lu H, Ye H, You L. Photoswitchable Cascades for Allosteric and Bidirectional Control over Covalent Bonds and Assemblies. J Am Chem Soc 2024. [PMID: 38620077 DOI: 10.1021/jacs.4c01240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Studies of complex systems and emerging properties to mimic biosystems are at the forefront of chemical research. Dynamic multistep cascades, especially those exhibiting allosteric regulation, are challenging. Herein, we demonstrate a versatile platform of photoswitchable covalent cascades toward remote and bidirectional control of reversible covalent bonds and ensuing assemblies. The relay of a photochromic switch, keto-enol equilibrium, and ring-chain equilibrium allows light-mediated reversible allosteric structural changes. The accompanying distinct reactivity further enables photoswitchable dynamic covalent bonding and release of substrates bidirectionally through alternating two wavelengths of light, essentially realizing light-mediated signaling cycles. The downfall of energy by covalent bond formation/scission upon photochemical reactions offers the driving force for the controlled direction of the cascade. To show the molecular diversity, photoswitchable on-demand assembly/disassembly of covalent polymers, including structurally reconfigurable polymers, was realized. This work achieves photoswitchable allosteric regulation of covalent architectures within dynamic multistep cascades, which has rarely been reported before. The results resemble allosteric control within biological signaling networks and should set the stage for many endeavors, such as dynamic assemblies, molecular motors, responsive polymers, and intelligent materials.
Collapse
Affiliation(s)
- Hanwei Lu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hebo Ye
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
| | - Lei You
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, China
| |
Collapse
|
38
|
Kashida H, Azuma H, Sotome H, Miyasaka H, Asanuma H. Site-Selective Photo-Crosslinking of Stilbene Pairs in a DNA Duplex Mediated by Ruthenium Photocatalyst. Angew Chem Int Ed Engl 2024; 63:e202319516. [PMID: 38282170 DOI: 10.1002/anie.202319516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 01/30/2024]
Abstract
We herein report a method for site-selective photo-crosslinking of a DNA duplex. A stilbene pair was introduced into a DNA duplex and a ruthenium complex was conjugated with a triplex-forming oligonucleotide. We demonstrated that [2+2] photocycloaddition of the stilbene pair occurred upon irradiation with visible light when the ruthenium complex was in close proximity due to triplex formation. No reaction occurred when the ruthenium complex was not in proximity to the stilbene pair. The wavelength of visible light used was of lower energy than the wavelength of UV light necessary for direct excitation of stilbene. Quantum chemical calculation indicated that ruthenium complex catalyzed the photocycloaddition via triplet-triplet energy transfer. Site selectivity of this photo-crosslinking system was evaluated using a DNA duplex bearing two stilbene pairs as a substrate; we showed that the site of crosslinking was precisely regulated by the sequence of the oligonucleotide linked to the ruthenium complex. Since this method does not require orthogonal photoresponsive molecules, it will be useful in construction of complex photoresponsive DNA circuits, nanodevices and biological tools.
Collapse
Affiliation(s)
- Hiromu Kashida
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Hidenori Azuma
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Hikaru Sotome
- Division of Frontier Materials Science and, Center for Advanced Interdisciplinary Research, Graduate School of Engineering Science, Osaka University 1-3 Machikaneyama, Toyonaka, Osaka, 560-8531, Japan
| | - Hiroshi Miyasaka
- Division of Frontier Materials Science and, Center for Advanced Interdisciplinary Research, Graduate School of Engineering Science, Osaka University 1-3 Machikaneyama, Toyonaka, Osaka, 560-8531, Japan
| | - Hiroyuki Asanuma
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| |
Collapse
|
39
|
Su K, Vázquez O. Enlightening epigenetics: optochemical tools illuminate the path. Trends Biochem Sci 2024; 49:290-304. [PMID: 38350805 DOI: 10.1016/j.tibs.2024.01.003] [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: 10/04/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
Optochemical tools have become potent instruments for understanding biological processes at the molecular level, and the past decade has witnessed their use in epigenetics and epitranscriptomics (also known as RNA epigenetics) for deciphering gene expression regulation. By using photoresponsive molecules such as photoswitches and photocages, researchers can achieve precise control over when and where specific events occur. Therefore, these are invaluable for studying both histone and nucleotide modifications and exploring disease-related mechanisms. We systematically report and assess current examples in the field, and identify open challenges and future directions. These outstanding proof-of-concept investigations will inspire other chemical biologists to participate in these emerging fields given the potential of photochromic molecules in research and biomedicine.
Collapse
Affiliation(s)
- Kaijun Su
- Department of Chemistry, University of Marburg, Marburg D-35043, Germany
| | - Olalla Vázquez
- Department of Chemistry, University of Marburg, Marburg D-35043, Germany; Center for Synthetic Microbiology (SYNMIKRO), University of Marburg, Marburg D-35043, Germany.
| |
Collapse
|
40
|
Lin T, Engelhard L, Söldner B, Linser R, Summerer D. Light-Activatable MBD-Readers of 5-Methylcytosine Reveal Domain-Dependent Chromatin Association Kinetics In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307930. [PMID: 38164822 PMCID: PMC10953577 DOI: 10.1002/advs.202307930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/11/2023] [Indexed: 01/03/2024]
Abstract
5-Methylcytosine (5mC) is the central epigenetic mark of mammalian DNA, and plays fundamental roles in chromatin regulation. 5mC is dynamically read and translated into regulatory outputs by methyl-CpG-binding domain (MBD) proteins. These multidomain readers recognize 5mC via an MBD domain, and undergo additional domain-dependent interactions with multiple additional chromatin components. However, studying this dynamic process is limited by a lack of methods to conditionally control the 5mC affinity of MBD readers in cells. Light-control of MBD association to chromatin by genetically encoding a photocaged serine at the MBD-DNA interface is reported. The authors study the association of MBD1 to mouse pericentromeres, dependent on its CxxC3 and transcriptional repressor domains (TRD) which interact with unmethylated CpG and heterochromatin-associated complexes, respectively. Both domains significantly modulate association kinetics, arguing for a model in which the CxxC3 delays methylation responses of MBD1 by holding it at unmethylated loci, whereas the TRD promotes responses by aiding heterochromatin association is studied. Their approach offers otherwise inaccessible kinetic insights into the domain-specific regulation of a central MBD reader, and sets the basis for further unravelling how the integration of MBDs into complex heterochromatin interaction networks control the kinetics of 5mC reading and translation into altered chromatin states.
Collapse
Affiliation(s)
- Tzu‐Chen Lin
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Lena Engelhard
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Benedikt Söldner
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Rasmus Linser
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| | - Daniel Summerer
- Department of Chemistry and Chemical BiologyTU Dortmund UniversityOtto‐Hahn Str. 4a44227DortmundGermany
| |
Collapse
|
41
|
Zhou C, He H, Chen X. Photoactivatable Nanobody Conjugate Dimerizer Temporally Resolves Tiam1-Rac1 Signaling Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307549. [PMID: 38225743 PMCID: PMC10953561 DOI: 10.1002/advs.202307549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/07/2023] [Indexed: 01/17/2024]
Abstract
The precise spatiotemporal dynamics of protein activities play a crucial role in cell signaling pathways. To control cellular functions in a spatiotemporal manner, a powerful method called photoactivatable chemically induced dimerization (pCID) is used. In this study, photoactivatable nanobody conjugate inducers of dimerization (PANCIDs) is introduced, which combine pCID with nanobody technology. A PANCID consists of a nanobody module that directly binds to an antigenic target, a photocaged small molecule ligand, and a cyclic decaarginine (cR10 *) cell-penetrating peptide (CPP) for efficient nonendocytic intracellular delivery. Therefore, PANCID photodimerizers also benefit from nanobodies, such as their high affinities (in the nm or pm range), specificities, and ability to modulate endogenous proteins. Additionally it is demonstrated that the nanobody moiety can be easily replaced with alternative ones, expanding the potential applications. By using PANCIDs, the dynamics of the Tiam1-Rac1 signaling cascade is investigated and made an interesting finding. It is found that Rac1 and Tiam1 exhibit distinct behaviors in this axis, acting as time-resolved "molecular oscillators" that transit between different functions in the signaling cascade when activated either slowly or rapidly.
Collapse
Affiliation(s)
- Chengjian Zhou
- Laboratory of Chemical Biology and Frontier BiotechnologiesThe HIT Center for Life Sciences (HCLS)Harbin Institute of TechnologyHarbin150001P. R. China
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150001P. R. China
| | - Huiping He
- Laboratory of Chemical Biology and Frontier BiotechnologiesThe HIT Center for Life Sciences (HCLS)Harbin Institute of TechnologyHarbin150001P. R. China
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150001P. R. China
| | - Xi Chen
- Laboratory of Chemical Biology and Frontier BiotechnologiesThe HIT Center for Life Sciences (HCLS)Harbin Institute of TechnologyHarbin150001P. R. China
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150001P. R. China
| |
Collapse
|
42
|
Xuan W, Ma JA. Pinpointing Acidic Residues in Proteins. ChemMedChem 2024; 19:e202300623. [PMID: 38303683 DOI: 10.1002/cmdc.202300623] [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: 11/08/2023] [Revised: 12/18/2023] [Indexed: 02/03/2024]
Abstract
It is of great importance to pinpoint specific residues or sites of a protein in biological contexts to enable desired mechanism of action for small molecules or to precisely control protein function. In this regard, acidic residues including aspartic acid (Asp) and glutamic acid (Glu) hold great potential due to their great prevalence and unique function. To unlock the largely untapped potential, great efforts have been made recently by synthetic chemists, chemical biologists and pharmacologists. Herein, we would like to highlight the remarkable progress and particularly introduce the electrophiles that exhibit reactivity to carboxylic acids, the light-induced reactivities to carboxylic acids and the genetically encoded noncanonical amino acids that allow protein manipulations at acidic residues. We also comment on certain unresolved challenges, hoping to draw more attention to this rapidly developing area.
Collapse
Affiliation(s)
- Weimin Xuan
- Frontiers Science Center for Synthetic Biology, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Jun-An Ma
- Department of Chemistry, Frontiers Science Center for Synthetic Biology, Tianjin University, Tianjin, 300072, P. R. China
| |
Collapse
|
43
|
Kiy Z, Chaud J, Xu L, Brandhorst E, Kamali T, Vargas C, Keller S, Hong H, Specht A, Cambridge S. Towards a Light-mediated Gene Therapy for the Eye using Caged Ethinylestradiol and the Inducible Cre/lox System. Angew Chem Int Ed Engl 2024; 63:e202317675. [PMID: 38127455 DOI: 10.1002/anie.202317675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Increasingly, retinal pathologies are being treated with virus-mediated gene therapies. To be able to target viral transgene expression specifically to the pathological regions of the retina with light, we established an in vivo photoactivated gene expression paradigm for retinal tissue. Based on the inducible Cre/lox system, we discovered that ethinylestradiol is a suitable alternative to Tamoxifen as ethinylestradiol is more amenable to modification with photosensitive protecting compounds, i.e., "caging." Identification of ethinylestradiol as a ligand for the mutated human estradiol receptor was supported by in silico binding studies showing the reduced binding of caged ethinylestradiol. Caged ethinylestradiol was injected into the eyes of double transgenic GFAP-CreERT2 mice with a Cre-dependent tdTomato reporter transgene followed by irradiation with light of 450 nm. Photoactivation significantly increased retinal tdTomato expression compared to controls. We thus demonstrated a first step towards the development of a targeted, light-mediated gene therapy for the eyes.
Collapse
Affiliation(s)
- Zoe Kiy
- Heidelberg University, 69120, Heidelberg, Germany
| | - Juliane Chaud
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, CNRS, CAMB UMR 7199, 67000, Strasbourg, France
| | - Liang Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Eric Brandhorst
- Sektion Endokrinologie, Medizinische Fakultät Mannheim, 68167, Mannheim, Germany
| | - Tschackad Kamali
- Heidelberg Engineering GmbH, Max-Jarecki-Straße 8, 69115, Heidelberg, Germany
| | - Carolyn Vargas
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, 8010, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Sandro Keller
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, 8010, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Alexandre Specht
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, CNRS, CAMB UMR 7199, 67000, Strasbourg, France
| | - Sidney Cambridge
- Heidelberg University, 69120, Heidelberg, Germany
- Institute for Anatomy II, Dr. Senckenberg Anatomy, Goethe-University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| |
Collapse
|
44
|
Zeng K, Jiao ZH, Jiang Q, He R, Zhang Y, Li WG, Xu TL, Chen Y. Genetically Encoded Photocatalysis Enables Spatially Restricted Optochemical Modulation of Neurons in Live Mice. ACS CENTRAL SCIENCE 2024; 10:163-175. [PMID: 38292609 PMCID: PMC10823520 DOI: 10.1021/acscentsci.3c01351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 02/01/2024]
Abstract
Light provides high temporal precision for neuronal modulations. Small molecules are advantageous for neuronal modulation due to their structural diversity, allowing them to suit versatile targets. However, current optochemical methods release uncaged small molecules with uniform concentrations in the irradiation area, which lack spatial specificity as counterpart optogenetic methods from genetic encoding for photosensitive proteins. Photocatalysis provides spatial specificity by generating reactive species in the proximity of photocatalysts. However, current photocatalytic methods use antibody-tagged heavy-metal photocatalysts for spatial specificity, which are unsuitable for neuronal applications. Here, we report a genetically encoded metal-free photocatalysis method for the optochemical modulation of neurons via deboronative hydroxylation. The genetically encoded photocatalysts generate doxorubicin, a mitochondrial uncoupler, and baclofen by uncaging stable organoboronate precursors. The mitochondria, nucleus, membrane, cytosol, and ER-targeted drug delivery are achieved by this method. The distinct signaling pathway dissection in a single projection is enabled by the dual optogenetic and optochemical control of synaptic transmission. The itching signaling pathway is investigated by photocatalytic uncaging under live-mice skin for the first time by visible light irradiation. The cell-type-specific release of baclofen reveals the GABABR activation on NaV1.8-expressing nociceptor terminals instead of pan peripheral sensory neurons for itch alleviation in live mice.
Collapse
Affiliation(s)
- Kaixing Zeng
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, University of Chinese Academy
of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032 China
- School
of Physical Science and Technology, ShanghaiTech
University, 100 Haike Road, Shanghai 201210, China
| | - Zhi-Han Jiao
- Centre
for Brain Science and Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Qin Jiang
- Centre
for Brain Science and Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Ru He
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, University of Chinese Academy
of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032 China
- School
of Physical Science and Technology, ShanghaiTech
University, 100 Haike Road, Shanghai 201210, China
| | - Yixin Zhang
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, University of Chinese Academy
of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032 China
| | - Wei-Guang Li
- Centre
for Brain Science and Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Department
of Rehabilitation Medicine, Huashan Hospital, Institute for Translational
Brain Research, State Key Laboratory of Medical Neurobiology and Ministry
of Education Frontiers Centre for Brain Science, Fudan University, 131 Dongan Road, Shanghai 200032, China
| | - Tian-Le Xu
- Centre
for Brain Science and Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yiyun Chen
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Organic
Chemistry, University of Chinese Academy
of Sciences, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032 China
- School
of Physical Science and Technology, ShanghaiTech
University, 100 Haike Road, Shanghai 201210, China
- School
of Chemistry and Material Sciences, Hangzhou Institute for Advanced
Study, University of Chinese Academy of
Sciences, Sub-lane Xiangshan, Hangzhou 310024, China
| |
Collapse
|
45
|
Xiao L, Fang L, Kool ET. 2'-OH as a universal handle for studying intracellular RNAs. Cell Chem Biol 2024; 31:110-124. [PMID: 37992716 PMCID: PMC10841764 DOI: 10.1016/j.chembiol.2023.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/28/2023] [Accepted: 10/29/2023] [Indexed: 11/24/2023]
Abstract
RNA plays pivotal roles in most cellular processes, serving as both the traditional carrier of genetic information and as a key regulator of cellular functions. The advent of chemical technologies has contributed critically to the analysis of cellular RNA structures, functions, and interactions. Many of these methods and molecules involve the utilization of chemically reactive handles in RNAs, either introduced externally or inherent within the polymer itself. Among these handles, the 2'-hydroxyl (2'-OH) group has emerged as an exceptionally well-suited and general chemical moiety for the modification and profiling of RNAs in intracellular studies. In this review, we provide an overview of the recent advancements in intracellular applications of acylation at the 2'-OH group of RNA. We outline progress made in probing RNA structure and interactomes, controlling RNA function, RNA imaging, and analyzing RNA-small molecule interactions, all achieved in living cells through this simple chemical handle on the biopolymer.
Collapse
Affiliation(s)
- Lu Xiao
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Linglan Fang
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Eric T Kool
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
46
|
Morihiro K, Morita S, Harada N, Baba M, Yum J, Naito M, Miyata K, Nagae G, Okamoto A. RNA Oncological Therapeutics: Intracellular Hairpin RNA Assembly Enables MicroRNA-Triggered Anticancer Functionality. J Am Chem Soc 2024; 146:1346-1355. [PMID: 38170469 DOI: 10.1021/jacs.3c09524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
RNA therapeutics are of global interest because of their versatility in targeting a variety of intracellular and extracellular biomolecules. In that context, long double-stranded RNA (dsRNA) has been studied as an antitumor agent that activates the immune response. However, its performance is constrained by poor cancer selectivity and cell-penetration ability. Here, we designed and synthesized an oncolytic RNA hairpin pair (oHP) that was selectively cytotoxic toward cancer cells expressing abundant oncogenic microRNA-21 (miR-21). Although the structure of each hairpin RNA was thermodynamically metastable, catalytic miR-21 input triggered it to open to generate a long nicked dsRNA. We demonstrated that oHP functioned as a cytotoxic amplifier of information in the presence of miR-21 in various cancer cells and tumor-bearing mice. This work represents the first example of the use of short RNA molecules as build-up-type anticancer agents that are triggered by an oncogenic miRNA.
Collapse
Affiliation(s)
- Kunihiko Morihiro
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shunto Morita
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Naoki Harada
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Manami Baba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Jongmin Yum
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Mitsuru Naito
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Genta Nagae
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Akimitsu Okamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
47
|
Hu T, Yu Y, Lin Y, Chen C. Light-Triggered Signal Enhancement Strategy Integrated with a CRISPR/Cas13a-Based Assay for Ultrasensitive and Specific miRNA Detection. Anal Chem 2023; 95:18587-18594. [PMID: 38078412 DOI: 10.1021/acs.analchem.3c04487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
The development of facile, accurate, and affordable assays for microRNAs (miRNAs) in early cancer is greatly desirable but encounters an obstacle due to low cellular abundance in biofuids. In this study, we present a novel approach called a light-triggered exponential amplification strategy coupled with a CRISPR/Cas13a-based diagnostic system (LEXPA-CRISPR), which directly transduces rare miRNA targets into photocontrolled signal enhancement response. This innovative platform leverages trans-cleavage of CRISPR/Cas13a, activated by the miRNA target, to cleave specific RNA fragments within the MB@PC-NAC assembly, thus releasing free PC-single-stranded DNA (PC-ssDNA) that is modified by a photocleavable linker (PC linker). UV irradiation is further employed toward the photoresponsive PC-ssDNA, resulting in instantaneous generation of oligo with a new 5' phosphate group (Pho-ssDNA). The Pho-ssDNA serves as a trigger for rolling circle amplification (RCA) reaction, which generates thousands of long ssDNA repeats of diverse lengths with a strong fluorescence signal. Through optimization, we achieved a detection limit of 1 fM for miR21 without the need for target amplification. Moreover, the programmable versatility of LEXPA-CRISPR is also demonstrated for miR17 determination only with simple modification of CRISPR RNA (crRNA) sequences. This proposed biosensor successfully monitored the levels of miR21 and miR17 in tumor cells, showing a satisfactory consistency with the standard qRT-PCR method. Conclusively, LEXPA-CRISPR represents a promising strategy for ultrasensitive miRNA detection. It combines the advantages of light-triggered signal amplification and robust collateral cleavage activity of Cas13a, making it an attractive tool for practical CRISPR-based diagnostics.
Collapse
Affiliation(s)
- Tao Hu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Yingying Yu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Yu Lin
- International Peace Maternity & Child Health Hospital, Shanghai Municipal Key Clinical Specialty, Institute of Embryo-Fetal Original Adult Disease, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chuanxia Chen
- School of Materials Science and Engineering, University of Jinan, Jinan 250022, China
| |
Collapse
|
48
|
Long K, Lv W, Wang Z, Zhang Y, Chen K, Fan N, Li F, Zhang Y, Wang W. Near-infrared light-triggered prodrug photolysis by one-step energy transfer. Nat Commun 2023; 14:8112. [PMID: 38062051 PMCID: PMC10703928 DOI: 10.1038/s41467-023-43805-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Prodrug photolysis enables spatiotemporal control of drug release at the desired lesions. For photoactivated therapy, near-infrared (NIR) light is preferable due to its deep tissue penetration and low phototoxicity. However, most of the photocleavable groups cannot be directly activated by NIR light. Here, we report a upconversion-like process via only one step of energy transfer for NIR light-triggered prodrug photolysis. We utilize a photosensitizer (PS) that can be activated via singlet-triplet (S-T) absorption and achieve photolysis of boron-dipyrromethene (BODIPY)-based prodrugs via triplet-triplet energy transfer. Using the strategy, NIR light can achieve green light-responsive photolysis with a single-photon process. A wide range of drugs and bioactive molecules are designed and demonstrated to be released under low-irradiance NIR light (100 mW/cm2, 5 min) with high yields (up to 87%). Moreover, a micellar nanosystem encapsulating both PS and prodrug is developed to demonstrate the practicality of our strategy in normoxia aqueous environment for cancer therapy. This study may advance the development of photocleavable prodrugs and photoresponsive drug delivery systems for photo-activated therapy.
Collapse
Affiliation(s)
- Kaiqi Long
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wen Lv
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
- State Key Laboratory of Organic Electronics and Information Displays, Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Zihan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yaming Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ni Fan
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Feiyang Li
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Yichi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
49
|
Zheng X, Li Y, Cui T, Yang J, Meng X, Wang H, Chen L, He J, Chen N, Meng L, Ding L, Xie R. Traceless Protein-Selective Glycan Labeling and Chemical Modification. J Am Chem Soc 2023; 145:23670-23680. [PMID: 37857274 DOI: 10.1021/jacs.3c07889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Executing glycan editing at a molecular level not only is pivotal for the elucidation of complicated mechanisms involved in glycan-relevant biological processes but also provides a promising solution to potentiate disease therapy. However, the precision control of glycan modification or glyco-editing on a selected glycoprotein is by far a grand challenge. Of note is to preserve the intact cellular glycan landscape, which is preserved after editing events are completed. We report herein a versatile, traceless glycan modification methodology for customizing the glycoforms of targeted proteins (subtypes), by orchestrating chemical- and photoregulation in a protein-selective glycoenzymatic system. This method relies on a three-module, ligand-photocleavable linker-glycoenzyme (L-P-G) conjugate. We demonstrated that RGD- or synthetic carbohydrate ligand-containing conjugates (RPG and SPG) would not activate until after the ligand-receptor interaction is accomplished (chemical regulation). RPG and SPG can both release the glycoenzyme upon photoillumination (photoregulation). The adjustable glycoenzyme activity, combined with ligand recognition selectivity, minimizes unnecessary glycan editing perturbation, and photolytic cleavage enables precise temporal control of editing events. An altered target protein turnover and dimerization were observed in our system, emphasizing the significance of preserving the native physiological niche of a particular protein when precise modification on the carbohydrate epitope occurs.
Collapse
Affiliation(s)
- Xiaocui Zheng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Tongxiao Cui
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jing Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiangfeng Meng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Nan Chen
- ChinaChomiX Biotech (Nanjing) Co., Ltd., Nanjing 210061, China
| | - Liying Meng
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Ran Xie
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| |
Collapse
|
50
|
Gagarin AA, Minin AS, Shevyrin VA, Kostova IP, Benassi E, Belskaya NP. Photocaging of Carboxylic Function Bearing Biomolecules by New Thiazole Derived Fluorophore. Chemistry 2023; 29:e202302079. [PMID: 37530503 DOI: 10.1002/chem.202302079] [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: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/03/2023]
Abstract
The design and synthesis of a new fluorophore containing an arylidene thiazole scaffold resulted in a compound with good photophysical characteristics. Furthermore, the thiazole C5-methyl group was easily modified into specific functional groups (CH2 Br and CH2 OH) for the formation of a series of photocourier molecules containing model compounds (benzoic acids), as well as prodrugs, including salicylic acid, caffeic acid, and chlorambucil via a "benzyl" linker. Spectral characteristics (1 H, 13 C NMR, and high-resolution mass spectra) corresponded to the proposed structures. The photocourier molecules demonstrated absorption with high values of coefficient of molar extinction, exhibited contrasting green emission, and showed good dark stability. The mechanism of the photorelease was investigated through spectral analysis, HPLC-HRMS, and supported by TD-DFT calculations. The photoheterolysis and elimination of carboxylic acids were proved to occur in the excited state, yielding a carbocation as an intermediate moiety. The fluorophore structure provided stability to the carbocation through the delocalization of the positive charge via resonance structures. Viability assessment of Vero cells using the MTT-test confirmed the weak cytotoxicity of prodrugs without irradiation and it increase upon UV-light.
Collapse
Affiliation(s)
- Aleksey A Gagarin
- Department of Technology for Organic Synthesis, Ural Federal University, 19 Mira Str., Yekaterinburg, 620002, Russia
| | - Artem S Minin
- Department of Technology for Organic Synthesis, Ural Federal University, 19 Mira Str., Yekaterinburg, 620002, Russia
- M. N. Mikheev Institute of Metal Physics, Ural Branch of Russian Academy of Science, 18S. Kovalevskaya Str., Yekaterinburg, 620108, Russia
| | - Vadim A Shevyrin
- Department of Technology for Organic Synthesis, Ural Federal University, 19 Mira Str., Yekaterinburg, 620002, Russia
| | - Irena P Kostova
- Department of Chemistry, Faculty of Pharmacy, Medical University-Sofia, 2 Dunav Str., Sofia, Bulgaria
| | - Enrico Benassi
- Novosibirsk State University, Pirogova Str. 2, 630090, Novosibirsk, Russia
| | - Nataliya P Belskaya
- Department of Technology for Organic Synthesis, Ural Federal University, 19 Mira Str., Yekaterinburg, 620002, Russia
| |
Collapse
|