1
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2025; 298:123-184. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [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: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Reed PMM, Jang J, Woloschuk RM, Reis J, Hille JIC, Uppalapati M, Woolley GA. Effects of binding partners on thermal reversion rates of photoswitchable molecules. Proc Natl Acad Sci U S A 2025; 122:e2414748122. [PMID: 40035753 PMCID: PMC11912449 DOI: 10.1073/pnas.2414748122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/15/2025] [Indexed: 03/06/2025] Open
Abstract
The binding of photoswitchable molecules to partners forms the basis of many naturally occurring light-dependent signaling pathways and various photopharmacological and optogenetic tools. A critical parameter affecting the function of these molecules is the thermal half-life of the light state. Reports in the literature indicate that, in some cases, a binding partner can significantly influence the thermal half-life, while in other cases it has no effect. Here, we present a unifying framework for quantitatively analyzing the effects of binding partners on thermal reversion rates. We focus on photoswitchable protein/binder interactions involving LOV domains, photoactive yellow protein, and CBCR GAF domains with partners that bind either the light or the dark state of the photoswitchable domain. We show that the effect of a binding partner depends on the extent to which the transition state for reversion resembles the dark state or the light state. We quantify this resemblance with a ϕswitching value, where ϕswitching = 1 if the conformation of the part of the photoswitchable molecule that interacts with the binding partner closely resembles its dark state conformation and ϕswitching = 0 if it resembles its light state. In addition to providing information on the transition state for switching, this analysis can guide the design of photoswitchable systems that retain useful thermal half-lives in practice. The analysis also provides a basis for the use of simple kinetic measurements to determine effective changes in affinity even in complex milieu.
Collapse
Affiliation(s)
| | - Jaewan Jang
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
| | - Ryan M. Woloschuk
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
| | - Jakeb Reis
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
| | | | - Maruti Uppalapati
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SKS7N 5E5, Canada
| | - G. Andrew Woolley
- Department of Chemistry, University of Toronto, Toronto, ONM5S 3H6, Canada
| |
Collapse
|
3
|
Mei G, Pan H, Xu H, Chen K, Zheng W, Xu H, Chen Y, Lin W, Yang J, Lin Z, Liu Z, Zhang M. Optogenetics and Its Application in Nervous System Diseases. Adv Biol (Weinh) 2025; 9:e2400416. [PMID: 39927470 DOI: 10.1002/adbi.202400416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/03/2025] [Indexed: 02/11/2025]
Abstract
Optogenetics is an emerging technology that uses the light-responsive effects of photosensitive proteins to regulate the function of specific cells. This technique combines genetics with optics, allowing for the precise inhibition or activation of cell functions through the introduction of photosensitive proteins into target cells and subsequent light stimulation to activate these proteins. In recent years, numerous basic and clinical studies have demonstrated the unique advantages of this approach in the research and treatment of neurological disorders. This review aims to introduce the fundamental principles and techniques of optogenetics, as well as its applications in the research and treatment of neurological diseases.
Collapse
Affiliation(s)
- Guocheng Mei
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huiqiong Pan
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Hang Xu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kepei Chen
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Weihong Zheng
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Hualin Xu
- Department of Plastic Surgery, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Yuetong Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Lin
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Jie Yang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zhenlang Lin
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| | - Zhiming Liu
- Department of Spinal Surgery, the Affiliated Hospital of Qingdao University, No.59 Haier Road, Qingdao, Shandong, 266000, China
| | - Min Zhang
- Department of Pediatrics, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Perinatal Medicine of Wenzhou, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
- Zhejiang Provincial Clinical Research Center for Pediatric Disease, 109 Xueyuan West Road, Wenzhou, Zhejiang, 325027, China
| |
Collapse
|
4
|
Zhu L, Wang Y, Wu X, Wu G, Zhang G, Liu C, Zhang S. Protein design accelerates the development and application of optogenetic tools. Comput Struct Biotechnol J 2025; 27:717-732. [PMID: 40092664 PMCID: PMC11908464 DOI: 10.1016/j.csbj.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Optogenetics has substantially enhanced our understanding of biological processes by enabling high-precision tracking and manipulation of individual cells. It relies on photosensitive proteins to monitor and control cellular activities, thereby paving the way for significant advancements in complex system research. Photosensitive proteins play a vital role in the development of optogenetics, facilitating the establishment of cutting-edge methods. Recent breakthroughs in protein design have opened up opportunities to develop protein-based tools that can precisely manipulate and monitor cellular activities. These advancements will significantly accelerate the development and application of optogenetic tools. This article emphasizes the pivotal role of protein design in the development of optogenetic tools, offering insights into potential future directions. We begin by providing an introduction to the historical development and fundamental principles of optogenetics, followed by an exploration of the operational mechanisms of key photosensitive domains, which includes clarifying the conformational changes they undergo in response to light, such as allosteric modulation and dimerization processes. Building on this foundation, we reveal the development of protein design tools that will enable the creation of even more sophisticated optogenetic techniques.
Collapse
Affiliation(s)
| | | | - Xiaomin Wu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Guohua Wu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Guohao Zhang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Chuanyang Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Shaowei Zhang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| |
Collapse
|
5
|
Tago T, Ogawa T, Goto Y, Toyooka K, Tojima T, Nakano A, Satoh T, Satoh AK. RudLOV is an optically synchronized cargo transport method revealing unexpected effects of dynasore. EMBO Rep 2025; 26:613-634. [PMID: 39658747 PMCID: PMC11811055 DOI: 10.1038/s44319-024-00342-z] [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: 09/24/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Live imaging of secretory cargoes is a powerful method for understanding the mechanisms of membrane trafficking. Inducing the synchronous release of cargoes from an organelle is key for enhancing microscopic observation. We developed an optical cargo-releasing method, 'retention using dark state of LOV2' (RudLOV), which enables precise spatial, temporal, and quantity control during cargo release. A limited amount of cargo-release using RudLOV is able to visualize cargo cisternal-movement and cargo-specific exit sites on the Golgi/trans-Golgi network. Moreover, by controlling the timing of cargo-release using RudLOV, we reveal the canonical and non-canonical effects of the well-known dynamin inhibitor dynasore, which inhibits early- but not late-Golgi transport and exits from the trans-Golgi network where dynamin-2 is active. Accumulation of COPI vesicles at the cis-side of the Golgi stacks in dynasore-treated cells suggests that dynasore targets COPI-uncoating/tethering/fusion machinery in the early-Golgi cisternae or endoplasmic reticulum but not in the late-Golgi cisternae. These results provide insight into the cisternal maturation of Golgi stacks.
Collapse
Affiliation(s)
- Tatsuya Tago
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Takumi Ogawa
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Yumi Goto
- Technology Platform Division, Mass Spectrometry and Microscopy Unit, RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa, 230-0045, Japan
| | - Kiminori Toyooka
- Technology Platform Division, Mass Spectrometry and Microscopy Unit, RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa, 230-0045, Japan
| | - Takuro Tojima
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Akihiko Nakano
- Live Cell Super-Resolution Imaging Research Team, RIKEN Center for Advanced Photonics, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Takunori Satoh
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan.
| | - Akiko K Satoh
- Program of Life and Environmental Science, Graduate School of Integral Science for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan.
| |
Collapse
|
6
|
Muench P, Fiumara M, Southern N, Coda D, Aschenbrenner S, Correia B, Gräff J, Niopek D, Mathony J. A modular toolbox for the optogenetic deactivation of transcription. Nucleic Acids Res 2025; 53:gkae1237. [PMID: 39676667 PMCID: PMC11797043 DOI: 10.1093/nar/gkae1237] [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: 11/24/2023] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Light-controlled transcriptional activation is a commonly used optogenetic strategy that allows researchers to regulate gene expression with high spatiotemporal precision. The vast majority of existing tools are, however, limited to light-triggered induction of gene expression. Here, we inverted this mode of action and created optogenetic systems capable of efficiently terminating transcriptional activation in response to blue light. First, we designed highly compact regulators by photo-controlling the VP16 (pcVP16) transactivation peptide. Then, applying a two-hybrid strategy, we engineered LOOMINA (light off-operated modular inductor of transcriptional activation), a versatile transcriptional control platform for mammalian cells that is compatible with various effector proteins. Leveraging the flexibility of CRISPR systems, we combined LOOMINA with dCas9 to control transcription with blue light from endogenous promoters with exceptionally high dynamic ranges in multiple cell lines. Functionally and mechanistically, the versatile LOOMINA platform and the exceptionally compact pcVP16 transactivator represent valuable additions to the optogenetic repertoire for transcriptional regulation.
Collapse
Affiliation(s)
- Philipp Muench
- Department of Biology, Technical University of Darmstadt, Schnittspahnstraße 10, Darmstadt 64287, Germany
| | - Matteo Fiumara
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), SV 2513 (Bâtiment SV) - Station 19, Lausanne CH-1015, Switzerland
| | - Nicholas Southern
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| | - Davide Coda
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), SV 2513 (Bâtiment SV) - Station 19, Lausanne CH-1015, Switzerland
| | - Sabine Aschenbrenner
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| | - Bruno Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, AI 3138 (Bâtiment AI) – Station 19, Lausanne CH-1015, Switzerland
- Swiss Institute of Bioinformatics (SIB), AI 3138 (Bâtiment AI) – Station 19, Lausanne CH-1015, Switzerland
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), SV 2513 (Bâtiment SV) - Station 19, Lausanne CH-1015, Switzerland
| | - Dominik Niopek
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| | - Jan Mathony
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 364, Heidelberg 69120, Germany
| |
Collapse
|
7
|
Lindner F, Grossmann S, Diepold A. Light-Controlled Secretion and Injection of Proteins into Eukaryotic Cells by Optogenetic Control of the Bacterial Type III Secretion System. Methods Mol Biol 2025; 2840:115-131. [PMID: 39724348 DOI: 10.1007/978-1-0716-4047-0_9] [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
Gram-negative bacteria can use the type III secretion system (T3SS) to inject effector proteins into eukaryotic target cells. In this chapter, we describe the application of a light-controlled T3SS, based on the targeted sequestration of an essential dynamic T3SS component with the help of optogenetic interaction switches. This method enables to control the secretion or injection into eukaryotic cells for a wide range of protein cargos with high temporal and spatial precision.
Collapse
Affiliation(s)
- Florian Lindner
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Sebastian Grossmann
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Andreas Diepold
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany.
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany.
| |
Collapse
|
8
|
Lei J, Li H. The Photocleavable Protein PhoCl-Based Dynamic Hydrogels. ACS Biomater Sci Eng 2024; 10:7404-7412. [PMID: 39545597 DOI: 10.1021/acsbiomaterials.4c01584] [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: 11/17/2024]
Abstract
Dynamic protein hydrogels have attracted increasing attention owing to their tunable physiochemical and mechanical properties, customized functionality, and biocompatibility. Among the different types of dynamic hydrogels, photoresponsive hydrogels are of particular interest. Here, we report the engineering of a photoresponsive protein hydrogel by using the photocleavable protein PhoCl. We employed the well-developed SpyTag and SpyCatcher chemistry to engineer PhoCl-containing covalently cross-linked hydrogels. In the hydrogel network, PhoCl, which can be cleaved into two fragments upon violet irradiation, is employed as a dynamic structural motif to regulate the cross-linking density of the hydrogel network. The resultant PhoCl-containing hydrogels showed photoresponsive viscoelastic properties. Upon violet irradiation, the PhoCl hydrogels soften, leading to an irreversible reduction in the storage moduli. However, no gel-sol transition was observed. Leveraging this light-induced stiffness change, we employed this hydrogel as a cell culture substrate to investigate the mechanobiological response of NIH-3T3 fibroblast cells. Our results showed that 3T3 cells can change their morphologies in response to the stiffness change of the PhoCl hydrogel substrate dynamically, rendering PhoCl-based hydrogels a useful substrate for other mechanobiological studies.
Collapse
Affiliation(s)
- Jingqi Lei
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Hongbin Li
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| |
Collapse
|
9
|
Ma Y, Winegar PH, Figg CA, Ramani N, Anderson AJ, Ngo K, Ahrens JF, Chellam NS, Kim YJ, Mirkin CA. DNA-Regulated Multi-Protein Complement Control. J Am Chem Soc 2024; 146:32912-32918. [PMID: 39569872 PMCID: PMC11755408 DOI: 10.1021/jacs.4c11315] [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] [Indexed: 11/22/2024]
Abstract
In nature, the interactions between proteins and their complements/substrates can dictate complex functions. Herein, we explore how DNA on nucleic acid modified proteins can be used as scaffolds to deliberately control interactions with a peptide complement (by adjusting length, sequence, and rigidity). As model systems, split GFPs were covalently connected through DNA scaffolds (36-58 bp). Increasing the length or decreasing the rigidity of the DNA scaffold (through removal of the duplex) increases the extent of intramolecular protein binding (up to 7.5-fold) between these GFP fragments. Independent and dynamic control over functional outputs can also be regulated by DNA hybridization; a multi-protein (split CFP and YFP) architecture was synthesized and characterized by fluorescence. This ternary construct shows that DNA displacement strands in different stoichiometric ratios can be used deliberately to regulate competitive binding between two unique sets of proteins. These studies establish a foundation for creating new classes of biological machinery based upon the concept of DNA-regulated multi-protein complement control.
Collapse
Affiliation(s)
- Yinglun Ma
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Peter H. Winegar
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - C. Adrian Figg
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Namrata Ramani
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Alex J. Anderson
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Kathleen Ngo
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - John F. Ahrens
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Nikhil S. Chellam
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Young Jun Kim
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| | - Chad. A. Mirkin
- Department of Chemistry, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, 60208, United States
- Interdisciplinary Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois, 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois, 60208, United States
| |
Collapse
|
10
|
Huang ZD, Bugaj LJ. Optogenetic Control of Condensates: Principles and Applications. J Mol Biol 2024; 436:168835. [PMID: 39454749 DOI: 10.1016/j.jmb.2024.168835] [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: 08/15/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 10/28/2024]
Abstract
Biomolecular condensates appear throughout cell physiology and pathology, but the specific role of condensation or its dynamics is often difficult to determine. Optogenetics offers an expanding toolset to address these challenges, providing tools to directly control condensation of arbitrary proteins with precision over their formation, dissolution, and patterning in space and time. In this review, we describe the current state of the field for optogenetic control of condensation. We survey the proteins and their derivatives that form the foundation of this toolset, and we discuss the factors that distinguish them to enable appropriate selection for a given application. We also describe recent examples of the ways in which optogenetic condensation has been used in both basic and applied studies. Finally, we discuss important design considerations when engineering new proteins for optogenetic condensation, and we preview future innovations that will further empower this toolset in the coming years.
Collapse
Affiliation(s)
- Zikang Dennis Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lukasz J Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Janssen V, Huveneers S. Cell-cell junctions in focus - imaging junctional architectures and dynamics at high resolution. J Cell Sci 2024; 137:jcs262041. [PMID: 39480660 DOI: 10.1242/jcs.262041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Studies utilizing electron microscopy and live fluorescence microscopy have significantly enhanced our understanding of the molecular mechanisms that regulate junctional dynamics during homeostasis, development and disease. To fully grasp the enormous complexity of cell-cell adhesions, it is crucial to study the nanoscale architectures of tight junctions, adherens junctions and desmosomes. It is important to integrate these junctional architectures with the membrane morphology and cellular topography in which the junctions are embedded. In this Review, we explore new insights from studies using super-resolution and volume electron microscopy into the nanoscale organization of these junctional complexes as well as the roles of the junction-associated cytoskeleton, neighboring organelles and the plasma membrane. Furthermore, we provide an overview of junction- and cytoskeletal-related biosensors and optogenetic probes that have contributed to these advances and discuss how these microscopy tools enhance our understanding of junctional dynamics across cellular environments.
Collapse
Affiliation(s)
- Vera Janssen
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, 1105 AZ, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Jia L, Gao S, Qiao Y. Optical Control over Liquid–Liquid Phase Separation. SMALL METHODS 2024; 8:e2301724. [PMID: 38530063 DOI: 10.1002/smtd.202301724] [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: 12/12/2023] [Revised: 03/12/2024] [Indexed: 03/27/2024]
Abstract
Liquid-liquid phase separation (LLPS) is responsible for the emergence of intracellular membrane-less organelles and the development of coacervate protocells. Benefitting from the advantages of simplicity, precision, programmability, and noninvasiveness, light has become an effective tool to regulate the assembly dynamics of LLPS, and mediate various biochemical processes associated with LLPS. In this review, recent advances in optically controlling membrane-less organelles within living organisms are summarized, thereby modulating a series of biological processes including irreversible protein aggregation pathologies, transcription activation, metabolic flux, genomic rearrangements, and enzymatic reactions. Among these, the intracellular systems (i.e., optoDroplet, Corelet, PixELL, CasDrop, and other optogenetic systems) that enable the photo-mediated control over biomolecular condensation are highlighted. The design of photoactive complex coacervate protocells in laboratory settings by utilizing photochromic molecules such as azobenzene and diarylethene is further discussed. This review is expected to provide in-depth insights into phase separation-associated biochemical processes, bio-metabolism, and diseases.
Collapse
Affiliation(s)
- Liyan Jia
- Beijing National Laboratory for Molecular Sciences (BNLMS), Laboratory of Polymer Physics and Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shan Gao
- Department of Orthopedic, Peking University Third Hospital, Beijing, 100191, China
| | - Yan Qiao
- Beijing National Laboratory for Molecular Sciences (BNLMS), Laboratory of Polymer Physics and Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
13
|
Gotthard G, Mous S, Weinert T, Maia RNA, James D, Dworkowski F, Gashi D, Furrer A, Ozerov D, Panepucci E, Wang M, Schertler GFX, Heberle J, Standfuss J, Nogly P. Capturing the blue-light activated state of the Phot-LOV1 domain from Chlamydomonas reinhardtii using time-resolved serial synchrotron crystallography. IUCRJ 2024; 11:792-808. [PMID: 39037420 PMCID: PMC11364019 DOI: 10.1107/s2052252524005608] [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: 10/30/2023] [Accepted: 06/11/2024] [Indexed: 07/23/2024]
Abstract
Light-oxygen-voltage (LOV) domains are small photosensory flavoprotein modules that allow the conversion of external stimuli (sunlight) into intracellular signals responsible for various cell behaviors (e.g. phototropism and chloroplast relocation). This ability relies on the light-induced formation of a covalent thioether adduct between a flavin chromophore and a reactive cysteine from the protein environment, which triggers a cascade of structural changes that result in the activation of a serine/threonine (Ser/Thr) kinase. Recent developments in time-resolved crystallography may allow the activation cascade of the LOV domain to be observed in real time, which has been elusive. In this study, we report a robust protocol for the production and stable delivery of microcrystals of the LOV domain of phototropin Phot-1 from Chlamydomonas reinhardtii (CrPhotLOV1) with a high-viscosity injector for time-resolved serial synchrotron crystallography (TR-SSX). The detailed process covers all aspects, from sample optimization to data collection, which may serve as a guide for soluble protein preparation for TR-SSX. In addition, we show that the crystals obtained preserve the photoreactivity using infrared spectroscopy. Furthermore, the results of the TR-SSX experiment provide high-resolution insights into structural alterations of CrPhotLOV1 from Δt = 2.5 ms up to Δt = 95 ms post-photoactivation, including resolving the geometry of the thioether adduct and the C-terminal region implicated in the signal transduction process.
Collapse
Affiliation(s)
- Guillaume Gotthard
- Institute of Molecular Biology and Biophysics, Department of BiologyETH Zurich8093ZürichSwitzerland
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Sandra Mous
- Institute of Molecular Biology and Biophysics, Department of BiologyETH Zurich8093ZürichSwitzerland
| | - Tobias Weinert
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Raiza Nara Antonelli Maia
- Experimental Molecular Biophysics, Department of PhysicsFreie Universität BerlinArnimallee 1414195BerlinGermany
| | - Daniel James
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Florian Dworkowski
- Macromolecular Crystallography, Swiss Light SourcePaul Scherrer Institute5232Villigen PSISwitzerland
| | - Dardan Gashi
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
- Laboratory of Femtochemistry, Photon Science DivisionPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Antonia Furrer
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Dmitry Ozerov
- Science ITPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Ezequiel Panepucci
- Laboratory for Macromolecules and Bioimaging, Photon Science DivisionPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Meitian Wang
- Laboratory for Macromolecules and Bioimaging, Photon Science DivisionPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Gebhard F. X. Schertler
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
- Department of BiologyETH Zürich8093ZürichSwitzerland
| | - Joachim Heberle
- Experimental Molecular Biophysics, Department of PhysicsFreie Universität BerlinArnimallee 1414195BerlinGermany
| | - Joerg Standfuss
- Laboratory of Biomolecular Research, Division of Biology and ChemistryPaul Scherrer Institute5232Villigen PSISwitzerland
| | - Przemyslaw Nogly
- Institute of Molecular Biology and Biophysics, Department of BiologyETH Zurich8093ZürichSwitzerland
- Dioscuri Center For Structural Dynamics of Receptors, Faculty of Biochemistry, Biophysics and BiotechnologyJagiellonian University in Kraków30-387KrakówPoland
| |
Collapse
|
14
|
Nagpal S, Swaminathan K, Beaudet D, Verdier M, Wang S, Berger CL, Berger F, Hendricks AG. Optogenetic control of kinesin-1, -2, -3 and dynein reveals their specific roles in vesicular transport. Cell Rep 2024; 43:114649. [PMID: 39159044 PMCID: PMC11416726 DOI: 10.1016/j.celrep.2024.114649] [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: 06/30/2023] [Revised: 07/04/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Each cargo in a cell employs a unique set of motor proteins for its transport. To dissect the roles of each type of motor, we developed optogenetic inhibitors of endogenous kinesin-1, -2, -3 and dynein motors and examined their effect on the transport of early endosomes, late endosomes, and lysosomes. While kinesin-1, -3, and dynein transport vesicles at all stages of endocytosis, kinesin-2 primarily drives late endosomes and lysosomes. Transient optogenetic inhibition of kinesin-1 or dynein causes both early and late endosomes to move more processively by relieving competition with opposing motors. Kinesin-2 and -3 support long-range transport, and optogenetic inhibition reduces the distances that their cargoes move. These results suggest that the directionality of transport is controlled through regulating kinesin-1 and dynein activity. On vesicles transported by several kinesin and dynein motors, modulating the activity of a single type of motor on the cargo is sufficient to direct motility.
Collapse
Affiliation(s)
- Sahil Nagpal
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | | | - Daniel Beaudet
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Maud Verdier
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada; Department of Biomedical Engineering and Health, Episen, Université Paris-Est Créteil, 94010 Créteil Cedex, France
| | - Samuel Wang
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Christopher L Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405-0075, USA
| | - Florian Berger
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Utrecht University, Utrecht, the Netherlands
| | - Adam G Hendricks
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada.
| |
Collapse
|
15
|
Yang H, Chen W. Protease-Responsive Toolkit for Conditional Targeted Protein Degradation. ACS Synth Biol 2024; 13:2073-2080. [PMID: 38889440 DOI: 10.1021/acssynbio.4c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
BioPROTACs are heterobifunctional proteins designed for targeted protein degradation. While they offer a potential therapeutic avenue for modulating disease-related proteins, the current strategies are static in nature and lack the ability to modulate protein degradation dynamically. Here, we introduce a synthetic framework for dynamic fine-tuning of target protein levels using protease control switches. The idea is to utilize proteases as an interfacing layer between exogenous inputs and protein degradation by modulating the recruitment of target proteins to E3 ligase by separating the two binding domains on bioPROTACs. By decoupling the external inputs from the primary protease layer, new conditional degradation phenotypes can be readily adapted with minimal modifications to the design. We demonstrate the adaptability of this approach using two highly efficient "bioPROTAC" systems: AdPROM and IpaH9.8-based Ubiquibodies. Using the TEV protease as the transducer, we can interface small-molecule and optogenetic inputs for conditional targeted protein degradation. Our findings highlight the potential of bioPROTACs with protease-responsive linkers as a versatile tool for conditional targeted protein degradation.
Collapse
Affiliation(s)
- Hopen Yang
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
16
|
Lian YL, Lin YC. The emerging tools for precisely manipulating microtubules. Curr Opin Cell Biol 2024; 88:102360. [PMID: 38640790 DOI: 10.1016/j.ceb.2024.102360] [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: 11/28/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/21/2024]
Abstract
Cells generate a highly diverse microtubule network to carry out different activities. This network is comprised of distinct tubulin isotypes, tubulins with different post-translational modifications, and many microtubule-based structures. Defects in this complex system cause numerous human disorders. However, how different microtubule subtypes in this network regulate cellular architectures and activities remains largely unexplored. Emerging tools such as photosensitive pharmaceuticals, chemogenetics, and optogenetics enable the spatiotemporal manipulation of structures, dynamics, post-translational modifications, and cross-linking with actin filaments in target microtubule subtypes. This review summarizes the design rationale and applications of these new approaches and aims to provide a roadmap for researchers navigating the intricacies of microtubule dynamics and their post-translational modifications in cellular contexts, thereby opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Yen-Ling Lian
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
17
|
Zhu Y, Ding W, Chen Y, Shan Y, Liu C, Fan X, Lin S, Chen PR. Genetically encoded bioorthogonal tryptophan decaging in living cells. Nat Chem 2024; 16:533-542. [PMID: 38418535 DOI: 10.1038/s41557-024-01463-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/29/2024] [Indexed: 03/01/2024]
Abstract
Tryptophan (Trp) plays a critical role in the regulation of protein structure, interactions and functions through its π system and indole N-H group. A generalizable method for blocking and rescuing Trp interactions would enable the gain-of-function manipulation of various Trp-containing proteins in vivo, but generating such a platform remains challenging. Here we develop a genetically encoded N1-vinyl-caged Trp capable of rapid and bioorthogonal decaging through an optimized inverse electron-demand Diels-Alder reaction, allowing site-specific activation of Trp on a protein of interest in living cells. This chemical activation of a genetically encoded caged-tryptophan (Trp-CAGE) strategy enables precise activation of the Trp of interest underlying diverse important molecular interactions. We demonstrate the utility of Trp-CAGE across various protein families, such as catalase-peroxidases and kinases, as translation initiators and posttranslational modification readers, allowing the modulation of epigenetic signalling in a temporally controlled manner. Coupled with computer-aided prediction, our strategy paves the way for bioorthogonal Trp activation on more than 28,000 candidate proteins within their native cellular settings.
Collapse
Affiliation(s)
- Yuchao Zhu
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Wenlong Ding
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yulin Chen
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Ye Shan
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chao Liu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Xinyuan Fan
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
| | - Shixian Lin
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China.
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Peng R Chen
- New Cornerstone Science Laboratory, Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
18
|
Shkarina K, Broz P. Selective induction of programmed cell death using synthetic biology tools. Semin Cell Dev Biol 2024; 156:74-92. [PMID: 37598045 DOI: 10.1016/j.semcdb.2023.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/21/2023]
Abstract
Regulated cell death (RCD) controls the removal of dispensable, infected or malignant cells, and is thus essential for development, homeostasis and immunity of multicellular organisms. Over the last years different forms of RCD have been described (among them apoptosis, necroptosis, pyroptosis and ferroptosis), and the cellular signaling pathways that control their induction and execution have been characterized at the molecular level. It has also become apparent that different forms of RCD differ in their capacity to elicit inflammation or an immune response, and that RCD pathways show a remarkable plasticity. Biochemical and genetic studies revealed that inhibition of a given pathway often results in the activation of back-up cell death mechanisms, highlighting close interconnectivity based on shared signaling components and the assembly of multivalent signaling platforms that can initiate different forms of RCD. Due to this interconnectivity and the pleiotropic effects of 'classical' cell death inducers, it is challenging to study RCD pathways in isolation. This has led to the development of tools based on synthetic biology that allow the targeted induction of RCD using chemogenetic or optogenetic methods. Here we discuss recent advances in the development of such toolset, highlighting their advantages and limitations, and their application for the study of RCD in cells and animals.
Collapse
Affiliation(s)
- Kateryna Shkarina
- Institute of Innate Immunity, University Hospital Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Switzerland.
| |
Collapse
|
19
|
Fenelon KD, Krause J, Koromila T. Opticool: Cutting-edge transgenic optical tools. PLoS Genet 2024; 20:e1011208. [PMID: 38517915 PMCID: PMC10959397 DOI: 10.1371/journal.pgen.1011208] [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] [Indexed: 03/24/2024] Open
Abstract
Only a few short decades have passed since the sequencing of GFP, yet the modern repertoire of transgenically encoded optical tools implies an exponential proliferation of ever improving constructions to interrogate the subcellular environment. A myriad of tags for labeling proteins, RNA, or DNA have arisen in the last few decades, facilitating unprecedented visualization of subcellular components and processes. Development of a broad array of modern genetically encoded sensors allows real-time, in vivo detection of molecule levels, pH, forces, enzyme activity, and other subcellular and extracellular phenomena in ever expanding contexts. Optogenetic, genetically encoded optically controlled manipulation systems have gained traction in the biological research community and facilitate single-cell, real-time modulation of protein function in vivo in ever broadening, novel applications. While this field continues to explosively expand, references are needed to assist scientists seeking to use and improve these transgenic devices in new and exciting ways to interrogate development and disease. In this review, we endeavor to highlight the state and trajectory of the field of in vivo transgenic optical tools.
Collapse
Affiliation(s)
- Kelli D. Fenelon
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Julia Krause
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
| | - Theodora Koromila
- Department of Biology, University of Texas at Arlington, Arlington, Texas, United States of America
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
20
|
Benisch M, Aoki SK, Khammash M. Unlocking the potential of optogenetics in microbial applications. Curr Opin Microbiol 2024; 77:102404. [PMID: 38039932 DOI: 10.1016/j.mib.2023.102404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/07/2023] [Accepted: 11/06/2023] [Indexed: 12/03/2023]
Abstract
Optogenetics is a powerful approach that enables researchers to use light to dynamically manipulate cellular behavior. Since the first published use of optogenetics in synthetic biology, the field has expanded rapidly, yielding a vast array of tools and applications. Despite its immense potential for achieving high spatiotemporal precision, optogenetics has predominantly been employed as a substitute for conventional chemical inducers. In this short review, we discuss key features of microbial optogenetics and highlight applications for understanding biology, cocultures, bioproduction, biomaterials, and therapeutics, in which optogenetics is more fully utilized to realize goals not previously possible by other methods.
Collapse
Affiliation(s)
- Moritz Benisch
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland.
| | - Stephanie K Aoki
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland.
| | - Mustafa Khammash
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Schanzenstrasse 44, 4056 Basel, Switzerland.
| |
Collapse
|
21
|
Tague N, Coriano-Ortiz C, Sheets MB, Dunlop MJ. Light-inducible protein degradation in E. coli with the LOVdeg tag. eLife 2024; 12:RP87303. [PMID: 38270583 PMCID: PMC10945698 DOI: 10.7554/elife.87303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
Molecular tools for optogenetic control allow for spatial and temporal regulation of cell behavior. In particular, light-controlled protein degradation is a valuable mechanism of regulation because it can be highly modular, used in tandem with other control mechanisms, and maintain functionality throughout growth phases. Here, we engineered LOVdeg, a tag that can be appended to a protein of interest for inducible degradation in Escherichia coli using blue light. We demonstrate the modularity of LOVdeg by using it to tag a range of proteins, including the LacI repressor, CRISPRa activator, and the AcrB efflux pump. Additionally, we demonstrate the utility of pairing the LOVdeg tag with existing optogenetic tools to enhance performance by developing a combined EL222 and LOVdeg system. Finally, we use the LOVdeg tag in a metabolic engineering application to demonstrate post-translational control of metabolism. Together, our results highlight the modularity and functionality of the LOVdeg tag system and introduce a powerful new tool for bacterial optogenetics.
Collapse
Affiliation(s)
- Nathan Tague
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
- Biological Design Center, Boston UniversityBostonUnited States
| | - Cristian Coriano-Ortiz
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
- Biological Design Center, Boston UniversityBostonUnited States
| | - Michael B Sheets
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
- Biological Design Center, Boston UniversityBostonUnited States
| | - Mary J Dunlop
- Department of Biomedical Engineering, Boston UniversityBostonUnited States
- Biological Design Center, Boston UniversityBostonUnited States
| |
Collapse
|
22
|
Fuyal M, James JR. Controlling the Potency of T Cell Activation Using an Optically Tunable Chimeric Antigen Receptor. Methods Mol Biol 2024; 2800:55-66. [PMID: 38709477 DOI: 10.1007/978-1-0716-3834-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
The ability of biological systems to convert inputs from their environment into information to guide future decisions is central to life and a matter of great importance. While we know the components of many of the signaling networks that make these decisions, our understanding of the dynamic flow of information between these parts remains far more limited. T cells are an essential white blood cell type of an adaptive immune response and can discriminate between healthy and infected cells with remarkable sensitivity. This chapter describes the use of a synthetic T-cell receptor (OptoCAR) that is optically tunable within cell conjugates, providing control over the duration, and intensity of intracellular T-cell signaling dynamics. Optical control can also provide control over signaling with high spatial precision, and the OptoCAR is likely to find application more generally when modulating T-cell function with imaging approaches.
Collapse
Affiliation(s)
- Muna Fuyal
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - John R James
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.
| |
Collapse
|
23
|
Nanda S, Calderon A, Sachan A, Duong TT, Koch J, Xin X, Solouk-Stahlberg D, Wu YW, Nalbant P, Dehmelt L. Rho GTPase activity crosstalk mediated by Arhgef11 and Arhgef12 coordinates cell protrusion-retraction cycles. Nat Commun 2023; 14:8356. [PMID: 38102112 PMCID: PMC10724141 DOI: 10.1038/s41467-023-43875-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023] Open
Abstract
Rho GTPases play a key role in the spatio-temporal coordination of cytoskeletal dynamics during cell migration. Here, we directly investigate crosstalk between the major Rho GTPases Rho, Rac and Cdc42 by combining rapid activity perturbation with activity measurements in mammalian cells. These studies reveal that Rac stimulates Rho activity. Direct measurement of spatio-temporal activity patterns show that Rac activity is tightly and precisely coupled to local cell protrusions, followed by Rho activation during retraction. Furthermore, we find that the Rho-activating Lbc-type GEFs Arhgef11 and Arhgef12 are enriched at transient cell protrusions and retractions and recruited to the plasma membrane by active Rac. In addition, their depletion reduces activity crosstalk, cell protrusion-retraction dynamics and migration distance and increases migration directionality. Thus, our study shows that Arhgef11 and Arhgef12 facilitate exploratory cell migration by coordinating cell protrusion and retraction by coupling the activity of the associated regulators Rac and Rho.
Collapse
Affiliation(s)
- Suchet Nanda
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Abram Calderon
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Arya Sachan
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
| | - Thanh-Thuy Duong
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Johannes Koch
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Xiaoyi Xin
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Djamschid Solouk-Stahlberg
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Yao-Wen Wu
- SciLifeLab and Department of Chemistry, Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, 45141, Essen, Germany.
| | - Leif Dehmelt
- Fakultät für Chemie und Chemische Biologie, TU Dortmund University, 44227, Dortmund, Germany.
| |
Collapse
|
24
|
Karasev MM, Verkhusha VV, Shcherbakova DM. Near-Infrared Optogenetic Module for Conditional Protein Splicing. J Mol Biol 2023; 435:168360. [PMID: 37949312 PMCID: PMC10842711 DOI: 10.1016/j.jmb.2023.168360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Optogenetics has emerged as a powerful tool for spatiotemporal control of biological processes. Near-infrared (NIR) light, with its low phototoxicity and deep tissue penetration, holds particular promise. However, the optogenetic control of polypeptide bond formation has not yet been developed. In this study, we introduce a NIR optogenetic module for conditional protein splicing (CPS) based on the gp41-1 intein. We optimized the module to minimize background signals in the darkness and to maximize the contrast between light and dark conditions. Next, we engineered a NIR CPS gene expression system based on the protein ligation of a transcription factor. We applied the NIR CPS for light-triggered protein cleavage to activate gasdermin D, a pore-forming protein that induces pyroptotic cell death. Our NIR CPS optogenetic module represents a promising tool for controlling molecular processes through covalent protein linkage and cleavage.
Collapse
Affiliation(s)
- Maksim M Karasev
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland
| | - Vladislav V Verkhusha
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland; Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Daria M Shcherbakova
- Department of Genetics, and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
25
|
Lu Y, Chen Y, Zhu Y, Zhao J, Ren K, Lu Z, Li J, Hao Z. Stimuli-Responsive Protein Hydrogels: Their Design, Properties, and Biomedical Applications. Polymers (Basel) 2023; 15:4652. [PMID: 38139904 PMCID: PMC10747532 DOI: 10.3390/polym15244652] [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: 10/31/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Protein-based hydrogels are considered ideal biomaterials due to their high biocompatibility, diverse structure, and their improved bioactivity and biodegradability. However, it remains challenging to mimic the native extracellular matrices that can dynamically respond to environmental stimuli. The combination of stimuli-responsive functionalities with engineered protein hydrogels has facilitated the development of new smart hydrogels with tunable biomechanics and biological properties that are triggered by cyto-compatible stimuli. This review summarizes the recent advancements of responsive hydrogels prepared from engineered proteins and integrated with physical, chemical or biological responsive moieties. We underscore the design principles and fabrication approaches of responsive protein hydrogels, and their biomedical applications in disease treatment, drug delivery, and tissue engineering are briefly discussed. Finally, the current challenges and future perspectives in this field are highlighted.
Collapse
Affiliation(s)
- Yuxuan Lu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.L.); (Y.C.)
| | - Yuhe Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (Y.L.); (Y.C.)
| | - Yuhan Zhu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| | - Jingyi Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| | - Ketong Ren
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| | - Zhao Lu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| | - Jun Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| | - Ziyang Hao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (Y.Z.); (J.Z.)
| |
Collapse
|
26
|
Joest EF, Tampé R. Design principles for engineering light-controlled antibodies. Trends Biotechnol 2023; 41:1501-1517. [PMID: 37507295 DOI: 10.1016/j.tibtech.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023]
Abstract
Engineered antibodies are essential tools for research and advanced pharmacy. In the development of therapeutics, antibodies are excellent candidates as they offer both target recognition and modulation. Thanks to the latest advances in biotechnology, light-activated antibody fragments can be constructed to control spontaneous antigen interaction with high spatiotemporal precision. To implement conditional antigen binding, several optogenetic and optochemical engineering concepts have recently been developed. Here, we highlight the various strategies and discuss the features of opto-conditional antibodies. Each concept offers intrinsic advantages beneficial to different applications. In summary, the novel design approaches constitute a complementary toolset to promote current and upcoming antibody technologies with ultimate precision.
Collapse
Affiliation(s)
- Eike F Joest
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt a.M., Germany.
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt a.M., Germany.
| |
Collapse
|
27
|
Nalbant P, Wagner J, Dehmelt L. Direct investigation of cell contraction signal networks by light-based perturbation methods. Pflugers Arch 2023; 475:1439-1452. [PMID: 37851146 DOI: 10.1007/s00424-023-02864-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 10/19/2023]
Abstract
Cell contraction plays an important role in many physiological and pathophysiological processes. This includes functions in skeletal, heart, and smooth muscle cells, which lead to highly coordinated contractions of multicellular assemblies, and functions in non-muscle cells, which are often highly localized in subcellular regions and transient in time. While the regulatory processes that control cell contraction in muscle cells are well understood, much less is known about cell contraction in non-muscle cells. In this review, we focus on the mechanisms that control cell contraction in space and time in non-muscle cells, and how they can be investigated by light-based methods. The review particularly focusses on signal networks and cytoskeletal components that together control subcellular contraction patterns to perform functions on the level of cells and tissues, such as directional migration and multicellular rearrangements during development. Key features of light-based methods that enable highly local and fast perturbations are highlighted, and how experimental strategies can capitalize on these features to uncover causal relationships in the complex signal networks that control cell contraction.
Collapse
Affiliation(s)
- Perihan Nalbant
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Room T03 R01 D33, Universitätsstrasse 2, 45141, Essen, Germany.
| | - Jessica Wagner
- Department of Molecular Cell Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Room T03 R01 D33, Universitätsstrasse 2, 45141, Essen, Germany
| | - Leif Dehmelt
- Department of Systemic Cell Biology, Fakultät für Chemie und Chemische Biologie, Max Planck Institute of Molecular Physiology, and Dortmund University of Technology, Room CP-02-157, Otto-Hahn-Str. 4a, 44227, Dortmund, Germany.
| |
Collapse
|
28
|
Tague N, Coriano-Ortiz C, Sheets MB, Dunlop MJ. Light inducible protein degradation in E. coli with the LOVdeg tag. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530042. [PMID: 36865169 PMCID: PMC9980293 DOI: 10.1101/2023.02.25.530042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Molecular tools for optogenetic control allow for spatial and temporal regulation of cell behavior. In particular, light controlled protein degradation is a valuable mechanism of regulation because it can be highly modular, used in tandem with other control mechanisms, and maintain functionality throughout growth phases. Here, we engineered LOVdeg, a tag that can be appended to a protein of interest for inducible degradation in Escherichia coli using blue light. We demonstrate the modularity of LOVdeg by using it to tag a range of proteins, including the LacI repressor, CRISPRa activator, and the AcrB efflux pump. Additionally, we demonstrate the utility of pairing the LOVdeg tag with existing optogenetic tools to enhance performance by developing a combined EL222 and LOVdeg system. Finally, we use the LOVdeg tag in a metabolic engineering application to demonstrate post-translational control of metabolism. Together, our results highlight the modularity and functionality of the LOVdeg tag system, and introduce a powerful new tool for bacterial optogenetics.
Collapse
|
29
|
Zhou W, Ryan A, Janosko CP, Shoger KE, Haugh JM, Gottschalk RA, Deiters A. Isoform-specific optical activation of kinase function reveals p38-ERK signaling crosstalk. RSC Chem Biol 2023; 4:765-773. [PMID: 37799579 PMCID: PMC10549237 DOI: 10.1039/d2cb00157h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/08/2023] [Indexed: 10/07/2023] Open
Abstract
Evolution has diversified the mammalian proteome by the generation of protein isoforms that originate from identical genes, e.g., through alternative gene splicing or post-translational modifications, or very similar genes found in gene families. Protein isoforms can have either overlapping or unique functions and traditional chemical, biochemical, and genetic techniques are often limited in their ability to differentiate between isoforms due to their high similarity. This is particularly true in the context of highly dynamic cell signaling cascades, which often require acute spatiotemporal perturbation to assess mechanistic details. To that end, we describe a method for the selective perturbation of the individual protein isoforms of the mitogen-activated protein kinase (MAPK) p38. The genetic installation of a photocaging group at a conserved active site lysine enables the precise light-controlled initiation of kinase signaling, followed by investigation of downstream events. Through optical control, we have identified a novel point of crosstalk between two major signaling cascades: the p38/MAPK pathway and the extracellular signal-regulated kinase (ERK)/MAPK pathway. Specifically, using the photoactivated p38 isoforms, we have found the p38γ and p38δ variants to be positive regulators of the ERK signaling cascade, while confirming the p38α and p38β variants as negative regulators.
Collapse
Affiliation(s)
- Wenyuan Zhou
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Amy Ryan
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Chasity P Janosko
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
| | - Karsen E Shoger
- Department of Immunology, University of Pittsburgh School of Medicine Pittsburgh PA 15260 USA
- Center for Systems Immunology, University of Pittsburgh Pittsburgh PA 15261 USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University Raleigh NC 27606 USA
| | - Rachel A Gottschalk
- Department of Immunology, University of Pittsburgh School of Medicine Pittsburgh PA 15260 USA
- Center for Systems Immunology, University of Pittsburgh Pittsburgh PA 15261 USA
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh Pittsburgh PA 15260 USA
- Center for Systems Immunology, University of Pittsburgh Pittsburgh PA 15261 USA
| |
Collapse
|
30
|
Zalejski J, Sun J, Sharma A. Unravelling the Mystery inside Cells by Using Single-Molecule Fluorescence Imaging. J Imaging 2023; 9:192. [PMID: 37754956 PMCID: PMC10532472 DOI: 10.3390/jimaging9090192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Live-cell imaging is a powerful technique to study the dynamics and mechanics of various biological molecules like proteins, organelles, DNA, and RNA. With the rapid evolution of optical microscopy, our understanding of how these molecules are implicated in the cells' most critical physiological roles deepens. In this review, we focus on how spatiotemporal nanoscale live-cell imaging at the single molecule level allows for profound contributions towards new discoveries in life science. This review will start by summarizing how single-molecule tracking has been used to analyze membrane dynamics, receptor-ligand interactions, protein-protein interactions, inner- and extra-cellular transport, gene expression/transcription, and whole organelle tracking. We then move on to how current authors are trying to improve single-molecule tracking and overcome current limitations by offering new ways of labeling proteins of interest, multi-channel/color detection, improvements in time-lapse imaging, and new methods and programs to analyze the colocalization and movement of targets. We later discuss how single-molecule tracking can be a beneficial tool used for medical diagnosis. Finally, we wrap up with the limitations and future perspectives of single-molecule tracking and total internal reflection microscopy.
Collapse
Affiliation(s)
| | | | - Ashutosh Sharma
- Department of Chemistry, University of Illinois Chicago, Chicago, IL 60607, USA; (J.Z.); (J.S.)
| |
Collapse
|
31
|
Vide U, Kasapović D, Fuchs M, Heimböck MP, Totaro MG, Zenzmaier E, Winkler A. Illuminating the inner workings of a natural protein switch: Blue-light sensing in LOV-activated diguanylate cyclases. SCIENCE ADVANCES 2023; 9:eadh4721. [PMID: 37531459 PMCID: PMC10396304 DOI: 10.1126/sciadv.adh4721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/28/2023] [Indexed: 08/04/2023]
Abstract
Regulatory proteins play a crucial role in adaptation to environmental cues. Especially for lifestyle transitions, such as cell proliferation or apoptosis, switch-like characteristics are desirable. While nature frequently uses regulatory circuits to amplify or dampen signals, stand-alone protein switches are interesting for applications like biosensors, diagnostic tools, or optogenetics. However, such stand-alone systems frequently feature limited dynamic and operational ranges and suffer from slow response times. Here, we characterize a LOV-activated diguanylate cyclase (LadC) that offers precise temporal and spatial control of enzymatic activity with an exceptionally high dynamic range over four orders of magnitude. To establish this pronounced activation, the enzyme exhibits a two-stage activation process in which its activity is inhibited in the dark by caging its effector domains and stimulated upon illumination by the formation of an extended coiled-coil. These switch-like characteristics of the LadC system can be used to develop new optogenetic tools with tight regulation.
Collapse
Affiliation(s)
- Uršula Vide
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Dženita Kasapović
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Maximilian Fuchs
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Martin P. Heimböck
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Massimo G. Totaro
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Elfriede Zenzmaier
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
| | - Andreas Winkler
- Institute of Biochemistry, Graz University of Technology, Petersgasse 12/II, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
32
|
Mattedi F, Lloyd-Morris E, Hirth F, Vagnoni A. Optogenetic cleavage of the Miro GTPase reveals the direct consequences of real-time loss of function in Drosophila. PLoS Biol 2023; 21:e3002273. [PMID: 37590319 PMCID: PMC10465005 DOI: 10.1371/journal.pbio.3002273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 08/29/2023] [Accepted: 07/22/2023] [Indexed: 08/19/2023] Open
Abstract
Miro GTPases control mitochondrial morphology, calcium homeostasis, and regulate mitochondrial distribution by mediating their attachment to the kinesin and dynein motor complex. It is not clear, however, how Miro proteins spatially and temporally integrate their function as acute disruption of protein function has not been performed. To address this issue, we have developed an optogenetic loss of function "Split-Miro" allele for precise control of Miro-dependent mitochondrial functions in Drosophila. Rapid optogenetic cleavage of Split-Miro leads to a striking rearrangement of the mitochondrial network, which is mediated by mitochondrial interaction with the microtubules. Unexpectedly, this treatment did not impact the ability of mitochondria to buffer calcium or their association with the endoplasmic reticulum. While Split-Miro overexpression is sufficient to augment mitochondrial motility, sustained photocleavage shows that Split-Miro is surprisingly dispensable to maintain elevated mitochondrial processivity. In adult fly neurons in vivo, Split-Miro photocleavage affects both mitochondrial trafficking and neuronal activity. Furthermore, functional replacement of endogenous Miro with Split-Miro identifies its essential role in the regulation of locomotor activity in adult flies, demonstrating the feasibility of tuning animal behaviour by real-time loss of protein function.
Collapse
Affiliation(s)
- Francesca Mattedi
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Ethlyn Lloyd-Morris
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Frank Hirth
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Alessio Vagnoni
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
33
|
Sadhanasatish T, Augustin K, Windgasse L, Chrostek-Grashoff A, Rief M, Grashoff C. A molecular optomechanics approach reveals functional relevance of force transduction across talin and desmoplakin. SCIENCE ADVANCES 2023; 9:eadg3347. [PMID: 37343090 DOI: 10.1126/sciadv.adg3347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
Many mechanobiological processes that govern development and tissue homeostasis are regulated on the level of individual molecular linkages, and a number of proteins experiencing piconewton-scale forces in cells have been identified. However, under which conditions these force-bearing linkages become critical for a given mechanobiological process is often still unclear. Here, we established an approach to revealing the mechanical function of intracellular molecules using molecular optomechanics. When applied to the integrin activator talin, the technique provides direct evidence that its role as a mechanical linker is indispensable for the maintenance of cell-matrix adhesions and overall cell integrity. Applying the technique to desmoplakin shows that mechanical engagement of desmosomes to intermediate filaments is expendable under homeostatic conditions yet strictly required for preserving cell-cell adhesion under stress. These results reveal a central role of talin and desmoplakin as mechanical linkers in cell adhesion structures and demonstrate that molecular optomechanics is a powerful tool to investigate the molecular details of mechanobiological processes.
Collapse
Affiliation(s)
- Tanmay Sadhanasatish
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Katharina Augustin
- Center for Protein Assemblies and Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Lukas Windgasse
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Anna Chrostek-Grashoff
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| | - Matthias Rief
- Center for Protein Assemblies and Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Carsten Grashoff
- University of Münster, Institute of Integrative Cell Biology and Physiology, Münster D-48149, Germany
| |
Collapse
|
34
|
Nagasawa Y, Ueda HH, Kawabata H, Murakoshi H. LOV2-based photoactivatable CaMKII and its application to single synapses: Local Optogenetics. Biophys Physicobiol 2023; 20:e200027. [PMID: 38496236 PMCID: PMC10941968 DOI: 10.2142/biophysico.bppb-v20.0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/02/2023] [Indexed: 03/19/2024] Open
Abstract
Optogenetic techniques offer a high spatiotemporal resolution to manipulate cellular activity. For instance, Channelrhodopsin-2 with global light illumination is the most widely used to control neuronal activity at the cellular level. However, the cellular scale is much larger than the diffraction limit of light (<1 μm) and does not fully exploit the features of the "high spatial resolution" of optogenetics. For instance, until recently, there were no optogenetic methods to induce synaptic plasticity at the level of single synapses. To address this, we developed an optogenetic tool named photoactivatable CaMKII (paCaMKII) by fusing a light-sensitive domain (LOV2) to CaMKIIα, which is a protein abundantly expressed in neurons of the cerebrum and hippocampus and essential for synaptic plasticity. Combining photoactivatable CaMKII with two-photon excitation, we successfully activated it in single spines, inducing synaptic plasticity (long-term potentiation) in hippocampal neurons. We refer to this method as "Local Optogenetics", which involves the local activation of molecules and measurement of cellular responses. In this review, we will discuss the characteristics of LOV2, the recent development of its derivatives, and the development and application of paCaMKII.
Collapse
Affiliation(s)
- Yutaro Nagasawa
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Hiromi H Ueda
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Haruka Kawabata
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Hideji Murakoshi
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| |
Collapse
|
35
|
Abstract
Lipids are key components of all organisms. We are well educated in their use as fuel and their essential role to form membranes. We also know much about their biosynthesis and metabolism. We are also aware that most lipids have signaling character meaning that a change in their concentration or location constitutes a signal that helps a living cell to respond to changes in the environment or to fulfill its specific function ranging from secretion to cell division. What is much less understood is how lipids change location in cells over time and what other biomolecules they interact with at each stage of their lifetime. Due to the large number of often quite similar lipid species and the sometimes very short lifetime of signaling lipids, we need highly specific tools to manipulate and visualize lipids and lipid-protein interactions. If successfully applied, these tools provide fabulous opportunities for discovery.In this Account, I summarize the development of synthetic tools from our lab that were designed to address crucial properties that allow them to function as tools in live cell experiments. Techniques to change the concentration of lipids by adding a small molecule or by light are described and complemented by examples of biological findings made when applying the tools. This ranges from chemical dimerizer-based systems to synthetic "caged" lipid derivatives. Furthermore, I discuss the problem of locating a lipid in an intact cell. Synthetic molecular probes are described that help to unravel the lipid location and to determine their binding proteins. These location studies require in-cell lipid tagging by click chemistry, photo-cross-linking to prevent further movement and the "caging" groups to avoid premature metabolism. The combination of these many technical features in a single tool allows for the analysis of not only lipid fluxes through metabolism but also lipid transport from one membrane to another as well as revealing the lipid interactome in a cell-dependent manner. This latter point is crucial because with these multifunctional tools in combination with lipidomics we can now address differences in healthy versus diseased cells and ultimately find the changes that are essential for disease development and new therapeutics that prevent these changes.
Collapse
Affiliation(s)
- Carsten Schultz
- Department of Chemical Physiology and
Biochemistry, Oregon Health & Science
University, Portland, Oregon 97239, United States
| |
Collapse
|
36
|
Panda R, Panda PK, Krishnamoorthy J, Kar RK. Network analysis of chromophore binding site in LOV domain. Comput Biol Med 2023; 161:106996. [PMID: 37201443 DOI: 10.1016/j.compbiomed.2023.106996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/16/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023]
Abstract
Photoreceptor proteins are versatile toolbox for developing biosensors for optogenetic applications. These molecular tools get activated upon illumination of blue light, which in turn offers a non-invasive method for gaining high spatiotemporal resolution and precise control of cellular signal transduction. The Light-Oxygen-Voltage (LOV) domain family of proteins is a well-recognized system for constructing optogenetic devices. Translation of these proteins into efficient cellular sensors is possible by tuning their photochemistry lifetime. However, the bottleneck is the need for more understanding of the relationship between the protein environment and photocycle kinetics. Significantly, the effect of the local environment also modulates the electronic structure of chromophore, which perturbs the electrostatic and hydrophobic interaction within the binding site. This work highlights the critical factors hidden in the protein networks, linking with their experimental photocycle kinetics. It presents an opportunity to quantitatively examine the alternation in chromophore's equilibrium geometry and identify details which have substantial implications in designing synthetic LOV constructs with desirable photocycle efficiency.
Collapse
Affiliation(s)
- Rishab Panda
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Punjab, India
| | - Pritam K Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20, Uppsala, Sweden; Division of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Rajiv K Kar
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
37
|
Diaz-Rohrer B, Castello-Serrano I, Chan SH, Wang HY, Shurer CR, Levental KR, Levental I. Rab3 mediates a pathway for endocytic sorting and plasma membrane recycling of ordered microdomains. Proc Natl Acad Sci U S A 2023; 120:e2207461120. [PMID: 36848577 PMCID: PMC10013782 DOI: 10.1073/pnas.2207461120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 01/31/2023] [Indexed: 03/01/2023] Open
Abstract
The composition of the plasma membrane (PM) must be tightly controlled despite constant, rapid endocytosis, which requires active, selective recycling of endocytosed membrane components. For many proteins, the mechanisms, pathways, and determinants of this PM recycling remain unknown. We report that association with ordered, lipid-driven membrane microdomains (known as rafts) is sufficient for PM localization of a subset of transmembrane proteins and that abrogation of raft association disrupts their trafficking and leads to degradation in lysosomes. Using orthogonal, genetically encoded probes with tunable raft partitioning, we screened for the trafficking machinery required for efficient recycling of engineered microdomain-associated cargo from endosomes to the PM. Using this screen, we identified the Rab3 family as an important mediator of PM localization of microdomain-associated proteins. Disruption of Rab3 reduced PM localization of raft probes and led to their accumulation in Rab7-positive endosomes, suggesting inefficient recycling. Abrogation of Rab3 function also mislocalized the endogenous raft-associated protein Linker for Activation of T cells (LAT), leading to its intracellular accumulation and reduced T cell activation. These findings reveal a key role for lipid-driven microdomains in endocytic traffic and suggest Rab3 as a mediator of microdomain recycling and PM composition.
Collapse
Affiliation(s)
- Barbara Diaz-Rohrer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Ivan Castello-Serrano
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Sze Ham Chan
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Hong-Yin Wang
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Carolyn R. Shurer
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Kandice R. Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA22904
| |
Collapse
|
38
|
Jang J, Tang K, Youn J, McDonald S, Beyer HM, Zurbriggen MD, Uppalapati M, Woolley GA. Engineering of bidirectional, cyanobacteriochrome-based light-inducible dimers (BICYCL)s. Nat Methods 2023; 20:432-441. [PMID: 36823330 DOI: 10.1038/s41592-023-01764-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 12/21/2022] [Indexed: 02/25/2023]
Abstract
Optogenetic tools for controlling protein-protein interactions (PPIs) have been developed from a small number of photosensory modules that respond to a limited selection of wavelengths. Cyanobacteriochrome (CBCR) GAF domain variants respond to an unmatched array of colors; however, their natural molecular mechanisms of action cannot easily be exploited for optogenetic control of PPIs. Here we developed bidirectional, cyanobacteriochrome-based light-inducible dimers (BICYCL)s by engineering synthetic light-dependent interactors for a red/green GAF domain. The systematic approach enables the future engineering of the broad chromatic palette of CBCRs for optogenetics use. BICYCLs are among the smallest optogenetic tools for controlling PPIs and enable either green-ON/red-OFF (BICYCL-Red) or red-ON/green-OFF (BICYCL-Green) control with up to 800-fold state selectivity. The access to green wavelengths creates new opportunities for multiplexing with existing tools. We demonstrate the utility of BICYCLs for controlling protein subcellular localization and transcriptional processes in mammalian cells and for multiplexing with existing blue-light tools.
Collapse
Affiliation(s)
- Jaewan Jang
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Kun Tang
- Institute of Synthetic Biology, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jeffrey Youn
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Sherin McDonald
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hannes M Beyer
- Institute of Synthetic Biology, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Matias D Zurbriggen
- Institute of Synthetic Biology, Heinrich-Heine-Universität, Düsseldorf, Germany. .,CEPLAS - Cluster of Excellence on Plant Science, Düsseldorf, Germany.
| | - Maruti Uppalapati
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - G Andrew Woolley
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
39
|
Younas T, Liu C, Struwe WB, Kukura P, He L. Engineer RNA-Protein Nanowires as Light-Responsive Biomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206513. [PMID: 36642821 DOI: 10.1002/smll.202206513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/15/2022] [Indexed: 06/17/2023]
Abstract
RNA molecules have emerged as increasingly attractive biomaterials with important applications such as RNA interference (RNAi) for cancer treatment and mRNA vaccines against infectious diseases. However, it remains challenging to engineer RNA biomaterials with sophisticated functions such as non-covalent light-switching ability. Herein, light-responsive RNA-protein nanowires are engineered to have such functions. It first demonstrates that the high affinity of RNA aptamer enables the formation of long RNA-protein nanowires through designing a dimeric RNA aptamer and an engineered green fluorescence protein (GFP) that contains two TAT-derived peptides at N- and C- termini. GFP is then replaced with an optogenetic protein pair system, LOV2 (light-oxygen-voltage) protein and its binding partner ZDK (Z subunit of protein A), to confer blue light-controlled photo-switching ability. The light-responsive nanowires are long (>500 nm) in the dark, but small (20-30 nm) when exposed to light. Importantly, the co-assembly of this RNA-protein hybrid biomaterial does not rely on the photochemistry commonly used for light-responsive biomaterials, such as bond formation, cleavage, and isomerization, and is thus reversible. These RNA-protein structures can serve as a new class of light-controlled biocompatible frameworks for incorporating versatile elements such as RNA, DNA, and enzymes.
Collapse
Affiliation(s)
- Tayyaba Younas
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Chang Liu
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Weston B Struwe
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| | - Philipp Kukura
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| | - Lizhong He
- Department of Chemical and Biological Engineering, Monash University, Clayton, VIC, 3800, Australia
| |
Collapse
|
40
|
Kunida K, Takagi N, Aoki K, Ikeda K, Nakamura T, Sakumura Y. Decoding cellular deformation from pseudo-simultaneously observed Rho GTPase activities. Cell Rep 2023; 42:112071. [PMID: 36764299 DOI: 10.1016/j.celrep.2023.112071] [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: 10/17/2022] [Revised: 10/31/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Limitations in simultaneously observing the activity of multiple molecules in live cells prevent researchers from elucidating how these molecules coordinate the dynamic regulation of cellular functions. Here, we propose the motion-triggered average (MTA) algorithm to characterize pseudo-simultaneous dynamic changes in arbitrary cellular deformation and molecular activities. Using MTA, we successfully extract a pseudo-simultaneous time series from individually observed activities of three Rho GTPases: Cdc42, Rac1, and RhoA. To verify that this time series encoded information on cell-edge movement, we use a mathematical regression model to predict the edge velocity from the activities of the three molecules. The model accurately predicts the unknown edge velocity, providing numerical evidence that these Rho GTPases regulate edge movement. Data preprocessing using MTA combined with mathematical regression provides an effective strategy for reusing numerous individual observations of molecular activities.
Collapse
Affiliation(s)
- Katsuyuki Kunida
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 8916-5, Japan; School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Nobuhiro Takagi
- Graduate School of Information Science and Technology, Aichi Prefectural University, Nagakute, Aichi 480-1342, Japan
| | - Kazuhiro Aoki
- National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| | - Kazushi Ikeda
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 8916-5, Japan; Data Science Center, Nara Institute of Science and Technology, Ikoma, Nara 8916-5, Japan; RIKEN Center for Advanced Intelligence Project (RIKEN AIP), Kyoto 619-0288, Japan
| | - Takeshi Nakamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-0022, Japan
| | - Yuichi Sakumura
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 8916-5, Japan; Data Science Center, Nara Institute of Science and Technology, Ikoma, Nara 8916-5, Japan.
| |
Collapse
|
41
|
Fine spectral tuning of a flavin-binding fluorescent protein for multicolor imaging. J Biol Chem 2023; 299:102977. [PMID: 36738792 PMCID: PMC10023982 DOI: 10.1016/j.jbc.2023.102977] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Flavin-binding fluorescent proteins are promising genetically encoded tags for microscopy. However, spectral properties of their chromophores (riboflavin, flavin mononucleotide, and flavin adenine dinucleotide) are notoriously similar even between different protein families, which limits applications of flavoproteins in multicolor imaging. Here, we present a palette of 22 finely tuned fluorescent tags based on the thermostable LOV domain from Chloroflexus aggregans. We performed site saturation mutagenesis of three amino acid positions in the flavin-binding pocket, including the photoactive cysteine, to obtain variants with fluorescence emission maxima uniformly covering the wavelength range from 486 to 512 nm. We demonstrate three-color imaging based on spectral separation and two-color fluorescence lifetime imaging of bacteria, as well as two-color imaging of mammalian cells (HEK293T), using the proteins from the palette. These results highlight the possibility of fine spectral tuning of flavoproteins and pave the way for further applications of flavin-binding fluorescent proteins in fluorescence microscopy.
Collapse
|
42
|
Dema A, Charafeddine R, Rahgozar S, van Haren J, Wittmann T. Growth cone advance requires EB1 as revealed by genomic replacement with a light-sensitive variant. eLife 2023; 12:84143. [PMID: 36715499 PMCID: PMC9917429 DOI: 10.7554/elife.84143] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A challenge in analyzing dynamic intracellular cell biological processes is the dearth of methodologies that are sufficiently fast and specific to perturb intracellular protein activities. We previously developed a light-sensitive variant of the microtubule plus end-tracking protein EB1 by inserting a blue light-controlled protein dimerization module between functional domains. Here, we describe an advanced method to replace endogenous EB1 with this light-sensitive variant in a single genome editing step, thereby enabling this approach in human induced pluripotent stem cells (hiPSCs) and hiPSC-derived neurons. We demonstrate that acute and local optogenetic EB1 inactivation in developing cortical neurons induces microtubule depolymerization in the growth cone periphery and subsequent neurite retraction. In addition, advancing growth cones are repelled from areas of blue light exposure. These phenotypes were independent of the neuronal EB1 homolog EB3, revealing a direct dynamic role of EB1-mediated microtubule plus end interactions in neuron morphogenesis and neurite guidance.
Collapse
Affiliation(s)
- Alessandro Dema
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Rabab Charafeddine
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | - Shima Rahgozar
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| | | | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
43
|
de Seze J, Gatin J, Coppey M. RhoA regulation in space and time. FEBS Lett 2023; 597:836-849. [PMID: 36658753 DOI: 10.1002/1873-3468.14578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023]
Abstract
RhoGTPases are well known for being controllers of cell cytoskeleton and share common features in the way they act and are controlled. These include their switch from GDP to GTP states, their regulations by different guanine exchange factors (GEFs), GTPase-activating proteins and guanosine dissociation inhibitors (GDIs), and their similar structure of active sites/membrane anchors. These very similar features often lead to the common consideration that the differences in their biological effects mainly arise from the different types of regulators and specific effectors associated with each GTPase. Focusing on data obtained through biosensors, live cell microscopy and recent optogenetic approaches, we highlight in this review that the regulation of RhoA appears to depart from Cdc42 and Rac1 modes of regulation through its enhanced lability at the plasma membrane. RhoA presents a high dynamic turnover at the membrane that is regulated not only by GDIs but also by GEFs, effectors and a possible soluble conformational state. This peculiarity of RhoA regulation may be important for the specificities of its functions, such as the existence of activity waves or its putative dual role in the initiation of protrusions and contractions.
Collapse
Affiliation(s)
- Jean de Seze
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France
| | - Joséphine Gatin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France
| | - Mathieu Coppey
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France
| |
Collapse
|
44
|
Lan TH, He L, Huang Y, Zhou Y. Optogenetics for transcriptional programming and genetic engineering. Trends Genet 2022; 38:1253-1270. [PMID: 35738948 PMCID: PMC10484296 DOI: 10.1016/j.tig.2022.05.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 01/24/2023]
Abstract
Optogenetics combines genetics and biophotonics to enable noninvasive control of biological processes with high spatiotemporal precision. When engineered into protein machineries that govern the cellular information flow as depicted in the central dogma, multiple genetically encoded non-opsin photosensory modules have been harnessed to modulate gene transcription, DNA or RNA modifications, DNA recombination, and genome engineering by utilizing photons emitting in the wide range of 200-1000 nm. We present herein generally applicable modular strategies for optogenetic engineering and highlight latest advances in the broad applications of opsin-free optogenetics to program transcriptional outputs and precisely manipulate the mammalian genome, epigenome, and epitranscriptome. We also discuss current challenges and future trends in opsin-free optogenetics, which has been rapidly evolving to meet the growing needs in synthetic biology and genetics research.
Collapse
Affiliation(s)
- Tien-Hung Lan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Lian He
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA.
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA; Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX 77030, USA.
| |
Collapse
|
45
|
Fan H, Barnes C, Hwang H, Zhang K, Yang J. Precise modulation of embryonic development through optogenetics. Genesis 2022; 60:e23505. [PMID: 36478118 PMCID: PMC9847014 DOI: 10.1002/dvg.23505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed enormous progress in optogenetics, which uses photo-sensitive proteins to control signal transduction in live cells and animals. The ever-increasing amount of optogenetic tools, however, could overwhelm the selection of appropriate optogenetic strategies. In this work, we summarize recent progress in this emerging field and highlight the application of opsin-free optogenetics in studying embryonic development, focusing on new insights gained into optical induction of morphogenesis, cell polarity, cell fate determination, tissue differentiation, neuronal regeneration, synaptic plasticity, and removal of cells during development.
Collapse
Affiliation(s)
- Huaxun Fan
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Collin Barnes
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hyojeong Hwang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA,Authors for correspondence: Kai Zhang, Ph.D., , 600 South Mathews Avenue, 314 B Roger Adams Laboratory,Urbana, Illinois 61801, USA, Phone: 1-217-300-0582; Jing Yang, Ph.D., , 2001 S Lincoln Ave, VMBSB3411, Urbana, Illinois 61802, USA, Phone: 1-217-333-6825
| | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA,Authors for correspondence: Kai Zhang, Ph.D., , 600 South Mathews Avenue, 314 B Roger Adams Laboratory,Urbana, Illinois 61801, USA, Phone: 1-217-300-0582; Jing Yang, Ph.D., , 2001 S Lincoln Ave, VMBSB3411, Urbana, Illinois 61802, USA, Phone: 1-217-333-6825
| |
Collapse
|
46
|
A red light-responsive photoswitch for deep tissue optogenetics. Nat Biotechnol 2022; 40:1672-1679. [PMID: 35697806 DOI: 10.1038/s41587-022-01351-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/04/2022] [Indexed: 12/30/2022]
Abstract
Red light penetrates deep into mammalian tissues and has low phototoxicity, but few optogenetic tools that use red light have been developed. Here we present MagRed, a red light-activatable photoswitch that consists of a red light-absorbing bacterial phytochrome incorporating a mammalian endogenous chromophore, biliverdin and a photo-state-specific binder that we developed using Affibody library selection. Red light illumination triggers the binding of the two components of MagRed and the assembly of split-proteins fused to them. Using MagRed, we developed a red light-activatable Cre recombinase, which enables light-activatable DNA recombination deep in mammalian tissues. We also created red light-inducible transcriptional regulators based on CRISPR-Cas9 that enable an up to 378-fold activation (average, 135-fold induction) of multiple endogenous target genes. MagRed will facilitate optogenetic applications deep in mammalian organisms in a variety of biological research areas.
Collapse
|
47
|
Plug-and-Display Photo-Switchable Systems on Plant Virus Nanoparticles. BIOTECH (BASEL (SWITZERLAND)) 2022; 11:biotech11040049. [PMID: 36278561 PMCID: PMC9589989 DOI: 10.3390/biotech11040049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/09/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Light can be used to regulate protein interactions with a high degree of spatial and temporal precision. Photo-switchable systems therefore allow the development of controllable protein complexes that can influence various cellular and molecular processes. Here, we describe a plant virus-based nanoparticle shuttle for the distribution of proteins that can be released when exposed to light. Potato virus X (PVX) is often used as a presentation system for heterologous proteins and epitopes, and has ideal properties for biomedical applications such as good tissue penetration and the ability to form hydrogels that present signaling molecules and promote cell adhesion. In this study, we describe three different systems attached to the surface of PVX particles: LOVTRAP, BphP1/QPAS1 and Dronpa145N. We demonstrated the functionality of all three photo-switchable protein complexes in vitro and the successful loading and unloading of PVX particles. The new systems provide the basis for promising applications in the biomedical and biomaterial sciences.
Collapse
|
48
|
Ohlendorf R, Möglich A. Light-regulated gene expression in Bacteria: Fundamentals, advances, and perspectives. Front Bioeng Biotechnol 2022; 10:1029403. [PMID: 36312534 PMCID: PMC9614035 DOI: 10.3389/fbioe.2022.1029403] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Numerous photoreceptors and genetic circuits emerged over the past two decades and now enable the light-dependent i.e., optogenetic, regulation of gene expression in bacteria. Prompted by light cues in the near-ultraviolet to near-infrared region of the electromagnetic spectrum, gene expression can be up- or downregulated stringently, reversibly, non-invasively, and with precision in space and time. Here, we survey the underlying principles, available options, and prominent examples of optogenetically regulated gene expression in bacteria. While transcription initiation and elongation remain most important for optogenetic intervention, other processes e.g., translation and downstream events, were also rendered light-dependent. The optogenetic control of bacterial expression predominantly employs but three fundamental strategies: light-sensitive two-component systems, oligomerization reactions, and second-messenger signaling. Certain optogenetic circuits moved beyond the proof-of-principle and stood the test of practice. They enable unprecedented applications in three major areas. First, light-dependent expression underpins novel concepts and strategies for enhanced yields in microbial production processes. Second, light-responsive bacteria can be optogenetically stimulated while residing within the bodies of animals, thus prompting the secretion of compounds that grant health benefits to the animal host. Third, optogenetics allows the generation of precisely structured, novel biomaterials. These applications jointly testify to the maturity of the optogenetic approach and serve as blueprints bound to inspire and template innovative use cases of light-regulated gene expression in bacteria. Researchers pursuing these lines can choose from an ever-growing, versatile, and efficient toolkit of optogenetic circuits.
Collapse
Affiliation(s)
- Robert Ohlendorf
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Andreas Möglich
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
- Bayreuth Center for Biochemistry and Molecular Biology, Universität Bayreuth, Bayreuth, Germany
- North-Bavarian NMR Center, Universität Bayreuth, Bayreuth, Germany
| |
Collapse
|
49
|
Caillaud MC. Tools for studying the cytoskeleton during plant cell division. TRENDS IN PLANT SCIENCE 2022; 27:1049-1062. [PMID: 35667969 DOI: 10.1016/j.tplants.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
The plant cytoskeleton regulates fundamental biological processes, including cell division. How to experimentally perturb the cytoskeleton is a key question if one wants to understand the role of both actin filaments (AFs) and microtubules (MTs) in a given biological process. While a myriad of mutants are available, knock-out in cytoskeleton regulators, when nonlethal, often produce little or no phenotypic perturbation because such regulators are often part of a large family, leading to functional redundancy. In this review, alternative techniques to modify the plant cytoskeleton during plant cell division are outlined. The different pharmacological and genetic approaches already developed in cell culture, transient assays, or in whole organisms are presented. Perspectives on the use of optogenetics to perturb the plant cytoskeleton are also discussed.
Collapse
Affiliation(s)
- Marie-Cécile Caillaud
- Laboratoire Reproduction et Développement des Plantes, Université de Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France.
| |
Collapse
|
50
|
Shikata H, Denninger P. Plant optogenetics: Applications and perspectives. CURRENT OPINION IN PLANT BIOLOGY 2022; 68:102256. [PMID: 35780691 DOI: 10.1016/j.pbi.2022.102256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
To understand cell biological processes, like signalling pathways, protein movements, or metabolic processes, precise tools for manipulation are desired. Optogenetics allows to control cellular processes by light and can be applied at a high temporal and spatial resolution. In the last three decades, various optogenetic applications have been developed for animal, fungal, and prokaryotic cells. However, using optogenetics in plants has been difficult due to biological and technical issues, like missing cofactors, the presence of endogenous photoreceptors, or the necessity of light for photosynthesis, which potentially activates optogenetic tools constitutively. Recently developed tools overcome these limitations, making the application of optogenetics feasible also in plants. Here, we highlight the most useful recent applications in plants and give a perspective for future optogenetic approaches in plants science.
Collapse
Affiliation(s)
- Hiromasa Shikata
- Division of Plant Environmental Responses, National Institute for Basic Biology, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8, Honcho, Kawaguchi, Japan.
| | - Philipp Denninger
- Technical University of Munich, School of Life Sciences, Plant Systems Biology, Emil-Ramann-Strasse 8, 85354 Freising, Germany.
| |
Collapse
|