1
|
Yin Y, Cheng X, Xie R, Fan D, Li H, Zhong S, Wegner SV, Zeng W, Chen F. Empowering bacteria with light: Optogenetically engineered bacteria for light-controlled disease theranostics and regulation. J Control Release 2025; 383:113787. [PMID: 40311686 DOI: 10.1016/j.jconrel.2025.113787] [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: 02/19/2025] [Revised: 04/19/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
Bacterial therapy has emerged as a promising approach for disease treatment due to its environmental sensitivity, immunogenicity, and modifiability. However, the clinical application of engineered bacteria is limited by differences of expression levels in patients and possible off-targeting. Optogenetics, which combines optics and genetics, offers key advantages such as remote controllability, non-invasiveness, and precise spatiotemporal control. By utilizing optogenetic tools, the behavior of engineered bacteria can be finely regulated, enabling on-demand control of the dosage and location of their therapeutic products. In this review, we highlight the latest advancements in the optogenetic engineering of bacteria for light-controlled disease theranostics and therapeutic regulation. By constructing a three-dimensional analytical framework of "sense-produce-apply", we begin by discussing the key components of bacterial optogenetic systems, categorizing them based on their photosensitive protein response to blue, green, and red light. Next, we introduce innovative light-producing tools that extend beyond traditional light sources. Then, special emphasis is placed on the biomedical applications of optogenetically engineered bacteria in treating diseases such as cancer, intestinal inflammation and systemic disease regulation. Finally, we address the challenges and future prospects of bacterial optogenetics, outlining potential directions for enhancing the safety and efficacy of light-controlled bacterial therapies. This review aims to provide insights and strategies for researchers working to advance the application of optogenetically engineered bacteria in drug delivery, precision medicine and therapeutic regulation.
Collapse
Affiliation(s)
- Ying Yin
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiang Cheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Ruyan Xie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Haohan Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Shibo Zhong
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster 48149, Germany
| | - Seraphine V Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster 48149, Germany
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
2
|
Liem N, Spreen A, Silapētere A, Hegemann P. Characterization of a rhodopsin-phosphodiesterase from Choanoeca flexa to be combined with rhodopsin-cyclases for bidirectional optogenetic cGMP control. J Biol Chem 2025; 301:108401. [PMID: 40081574 PMCID: PMC12004702 DOI: 10.1016/j.jbc.2025.108401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Rhodopsin phosphodiesterases (RhPDEs) were first discovered in the choanoflagellate Salpingoeca rosetta, but their physiological role remained unknown. Their light-dependent modulation was found to be low, limiting optogenetic application. However, recent in vivo studies in the choanoflagellate Choanoeca flexa revealed a strong linkage of RhPDE to the actomyosin-mediated contraction and colony sheet inversion and identified downstream cGMP effectors. Through screening various RhPDE variants from C. flexa, we identified four photomodulated PDEs of which C. flexa RhPDE1 (CfRhPDE1) revealed the highest cGMP affinity and the most pronounced light regulation with Km values of 1.9 and 4.4 μM in light and darkness. By coexpressing CfRhPDE1 with the rhodopsin-guanylyl-cyclase from the fungus Catenaria anguillulae and a cyclic nucleotide-gated ion channel from olfactory neurons in ND7/23 cells, we demonstrate bidirectional dual-color modulation of cGMP levels and ion channel conductance. Together with spectroscopic characterization, our fast functional recordings suggest that the M-state of the photocycle initiates functional changes in the phosphodiesterase domain via rapid rhodopsin-PDE coupling. With efficient expression and 3.5 s lifetime of the active state, this protein provides high photosensitivity to the host cells. This demonstrates that RhPDEs can regulate cGMP signaling in mammalian cells on a subsecond timescale, closing a present gap in optogenetics and assisting researchers in setting up multicomponent optogenetic systems for bidirectional control of cyclic nucleotides.
Collapse
Affiliation(s)
- Nicolas Liem
- Institut für Biologie, Humboldt University of Berlin, Berlin, Germany.
| | - Anika Spreen
- Institut für Biologie, Humboldt University of Berlin, Berlin, Germany
| | - Arita Silapētere
- Institut für Biologie, Humboldt University of Berlin, Berlin, Germany
| | - Peter Hegemann
- Institut für Biologie, Humboldt University of Berlin, Berlin, Germany
| |
Collapse
|
3
|
Tian Y, Zhang L, Wang Z, He Z, Shu L. Light Affects Host-Symbiont Dynamics in the Non-Photosynthetic Social Amoeba Symbiosis. Ecol Evol 2025; 15:e71320. [PMID: 40256268 PMCID: PMC12008035 DOI: 10.1002/ece3.71320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025] Open
Abstract
Light significantly influences phototactic behaviors and host-bacterial interactions of photosynthetic microorganisms such as algae. The non-photosynthetic slime mound amoeba Dictyostelium discoideum as the host shows phototaxis in the multicellular slugs, but the impact of light on amoeba-bacteria interactions remains unclear. Here we utilized two different clades of symbiotic Paraburkholderia species, namely Paraburkholderia agricolaris B1QS70 and Paraburkholderia hayleyella B2QS11, to investigate the light-induced symbiosis between the host amoebae and symbiotic bacteria. Our findings propose two light-induced symbiotic types (type I and type II termed from this study) likely due to amoebae metabolites or bacterial infection efficiency. The type I symbiosis reveals increased symbiotic B1QS70 amount in amoebae QS9 under light, while stable amounts persist in amoebae QS11 and QS70, both of which are native hosts of symbiotic Paraburkholderia species. Furthermore, the transcriptomics analysis suggests that certain upregulated genes, such as lectin genes, may play crucial roles in inducing the symbiosis of P. agricolaris B1QS70 in amoebae QS9 and QS70 under light stimulation. Conversely, the type II symbiosis enhances interactions between P. hayleyella B2QS11 and three individual amoebae clones (QS9, QS11, or QS70) in dark conditions due to the strong infection capability and high growth rates of B2QS11. Transcriptomic data show that a cluster of heat shock genes is upregulated in amoebae QS9 with B2QS11 under dark, indicating an immune response to the non-native host QS9, rather than that of in QS11 as the native host of B2QS11. Blue-light sensors like Cryptochrome/DNA photolyase in Paraburkholderia species might regulate the growth rate by light stimulation. These findings highlight light-regulated symbiosis between amoebae and two distinct Paraburkholderia species, indicating that light may be crucial for regulating amoebae-symbionts dynamics.
Collapse
Affiliation(s)
- Yuehui Tian
- School of Life SciencesGuangzhou UniversityGuangzhouChina
- School of Environmental Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Guangdong Provincial Key Laboratory of GuangzhouGuangzhouChina
| | - Lin Zhang
- School of Environmental Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Guangdong Provincial Key Laboratory of GuangzhouGuangzhouChina
| | - Zihe Wang
- School of Environmental Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Guangdong Provincial Key Laboratory of GuangzhouGuangzhouChina
| | - Zhili He
- School of Environmental Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Guangdong Provincial Key Laboratory of GuangzhouGuangzhouChina
| | - Longfei Shu
- School of Environmental Science and Engineering, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Guangdong Provincial Key Laboratory of GuangzhouGuangzhouChina
| |
Collapse
|
4
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
5
|
Ovechkina VS, Andrianova SK, Shimanskaia IO, Suvorova PS, Ryabinina AY, Blagonravov ML, Belousov VV, Mozhaev AA. Advances in Optogenetics and Thermogenetics for Control of Non-Neuronal Cells and Tissues in Biomedical Research. ACS Chem Biol 2025; 20:553-572. [PMID: 40056098 DOI: 10.1021/acschembio.4c00842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Optogenetics and chemogenetics are relatively new biomedical technologies that emerged 20 years ago and have been evolving rapidly since then. This has been made possible by the combined use of genetic engineering, optics, and electrophysiology. With the development of optogenetics and thermogenetics, the molecular tools for cellular control are continuously being optimized, studied, and modified, expanding both their applications and their biomedical uses. The most notable changes have occurred in the basic life sciences, especially in neurobiology and the activation of neurons to control behavior. Currently, these methods of activation have gone far beyond neurobiology and are being used in cardiovascular research, for potential cancer therapy, to control metabolism, etc. In this review, we provide brief information on the types of molecular tools for optogenetic and thermogenetic methods─microbial rhodopsins and proteins of the TRP superfamily─and also consider their applications in the field of activation of non-neuronal tissues and mammalian cells. We also consider the potential of these technologies and the prospects for the use of optogenetics and thermogenetics in biomedical research.
Collapse
Affiliation(s)
- Vera S Ovechkina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Sofya K Andrianova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Iana O Shimanskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Polina S Suvorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Anna Y Ryabinina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Mikhail L Blagonravov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117513, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, 121205, Russia
| | - Andrey A Mozhaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| |
Collapse
|
6
|
Nilsson CI, Dumral Ö, Sanchez G, Xie B, Müller A, Solimena M, Ren H, Idevall-Hagren O. Somatostatin triggers local cAMP and Ca 2+ signaling in primary cilia to modulate pancreatic β-cell function. EMBO J 2025; 44:1663-1691. [PMID: 39939781 PMCID: PMC11914567 DOI: 10.1038/s44318-025-00383-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
Somatostatin, released from δ-cells within pancreatic islets of Langerhans, is one of the most important negative regulators of islet hormone secretion. We find that islet δ-cells are positioned near, and release somatostatin onto, primary cilia of the other islet cell types, including insulin-secreting β-cells. Somatostatin activates ciliary somatostatin receptors, resulting in rapid lowering of the ciliary cAMP concentration which in turn promotes more sustained nuclear translocation of the cilia-dependent transcription factor GLI2 through a mechanism that operates in parallel with the canonical Hedgehog pathway and depends on ciliary Ca2+ signaling. We also find that primary cilia length is reduced in islets from human donors with type-2 diabetes, which is associated with a reduction in interactions between δ-cells and cilia. Our findings show that islet cell primary cilia constitute an important target of somatostatin action, which endows somatostatin with the ability to regulate islet cell function beyond acute suppression of hormone release.
Collapse
Affiliation(s)
- Ceren Incedal Nilsson
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 75123, Uppsala, Sweden
| | - Özge Dumral
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 75123, Uppsala, Sweden
| | - Gonzalo Sanchez
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 75123, Uppsala, Sweden
| | - Beichen Xie
- Center for Quantitative Biology, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
| | - Andreas Müller
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michele Solimena
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Huixia Ren
- Center for Quantitative Biology, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, 100871, Beijing, China
| | - Olof Idevall-Hagren
- Department of Medical Cell Biology, Uppsala University, BMC Box 571, 75123, Uppsala, Sweden.
| |
Collapse
|
7
|
Itkis DG, Brooks FP, Davis HC, Hotter R, Wong-Campos JD, Qi Y, Jia BZ, Howell M, Xiong M, Hayward RF, Lee BH, Wang Y, Perelman RT, Cohen AE. Luminos: open-source software for bidirectional microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639658. [PMID: 40060643 PMCID: PMC11888241 DOI: 10.1101/2025.02.22.639658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Bidirectional microscopy (BDM) combines simultaneous targeted optical perturbation and imaging of biophysical or biochemical signals (e.g. membrane voltage, Ca2+, or signaling molecules). A core challenge in BDM is precise spatial and temporal alignment of stimulation, imaging, and other experimental parameters. Here we present Luminos, an open-source MATLAB library for modular and precisely synchronized control of BDM experiments. The system supports hardware-triggered synchronization across stimulation, recording, and imaging channels with microsecond accuracy. Source code and documentation for Luminos are available online at https://www.luminosmicroscopy.com and https://github.com/adamcohenlab/luminos-microscopy. This library will facilitate development of bidirectional microscopy methods across the biological sciences.
Collapse
Affiliation(s)
- Daniel G Itkis
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - F Phil Brooks
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Hunter C Davis
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Raphael Hotter
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - J David Wong-Campos
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Yitong Qi
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Bill Z Jia
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Madeleine Howell
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Marley Xiong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Rebecca Frank Hayward
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Byung Hun Lee
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Yangdong Wang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Rebecca T Perelman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge MA 02138
| |
Collapse
|
8
|
Jewlikar SS, Collado JT, Ali MI, Sabbah A, He Y, Iuliano JN, Hall CR, Adamczyk K, Greetham GM, Lukacs A, Meech SR, Tonge PJ. Probing the Signal Transduction Mechanism of the Light-Activated Adenylate Cyclase OaPAC Using Unnatural Amino Acid Mutagenesis. ACS Chem Biol 2025; 20:369-377. [PMID: 39844630 PMCID: PMC12117572 DOI: 10.1021/acschembio.4c00627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
OaPAC, the photoactivated adenylyl cyclase from Oscillatoria acuminata, is composed of a blue light using FAD (BLUF) domain fused to an adenylate cyclase (AC) domain. Since both the BLUF and AC domains are part of the same protein, OaPAC is a model for understanding how the ultrafast modulation of the chromophore binding pocket caused by photoexcitation results in the activation of the output domain on the μs-s time scale. In the present work, we use unnatural amino acid mutagenesis to identify specific sites in the protein that are involved in transducing the signal from the FAD binding site to the ATP binding site. To provide insight into site-specific structural dynamics, we replaced W90 which is close to the chromophore pocket, F103 which interacts with W90 across the dimer interface, and F180 in the central core of the AC domain, with the infrared probe azido-Phe (AzPhe). Using ultrafast IR, we show that AzPhe at position 90 responds on multiple time scales following photoexcitation. In contrast, the light minus dark IR spectrum of AzPhe103 shows only a minor perturbation in environment between the dark and light states, while replacement of F180 with AzPhe resulted in a protein with no catalytic activity. We also replaced Y125, which hydrogen bonds with N256 across the dimer interface, with fluoro-Tyr residues. All the fluoro-Tyr substituted proteins retained the light-induced red shift in the flavin absorption spectrum; however, only the 3-FY125 OaPAC retained photoinduced catalytic activity. The loss of activity in 3,5-F2Y125 and 2,3,5-F3Y125 OaPAC, which potentially increase the acidity of the Y125 phenol by more than 1000-fold, suggests that deprotonation of Y125 disrupts the signal transduction pathway from the BLUF to the AC domain.
Collapse
Affiliation(s)
- Samruddhi S. Jewlikar
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
| | | | - Madeeha I. Ali
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
| | - Aya Sabbah
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
| | - YongLe He
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
| | - James N. Iuliano
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
| | - Christopher R. Hall
- School of Chemistry, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Katrin Adamczyk
- School of Chemistry, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Gregory M. Greetham
- Central Laser Facility, Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot, OX11 0QX, United Kingdom
| | - Andras Lukacs
- Department of Biophysics, Medical School, University of Pecs, Szigeti ut 12, 7624 Pecs, Hungary
| | - Stephen R. Meech
- School of Chemistry, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| | - Peter J. Tonge
- Department of Chemistry, Stony Brook University, Stony Brook, NY, 11794-3400, United States
- School of Chemistry, University of East Anglia, Norwich, NR4 7TJ, United Kingdom
| |
Collapse
|
9
|
Huang J, Fussenegger M. Programming mammalian cell behaviors by physical cues. Trends Biotechnol 2025; 43:16-42. [PMID: 39179464 DOI: 10.1016/j.tibtech.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/26/2024]
Abstract
In recent decades, the field of synthetic biology has witnessed remarkable progress, driving advances in both research and practical applications. One pivotal area of development involves the design of transgene switches capable of precisely regulating specified outputs and controlling cell behaviors in response to physical cues, which encompass light, magnetic fields, temperature, mechanical forces, ultrasound, and electricity. In this review, we delve into the cutting-edge progress made in the field of physically controlled protein expression in engineered mammalian cells, exploring the diverse genetic tools and synthetic strategies available for engineering targeting cells to sense these physical cues and generate the desired outputs accordingly. We discuss the precision and efficiency limitations inherent in these tools, while also highlighting their immense potential for therapeutic applications.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Klingelbergstrasse 48, CH-4056 Basel, Switzerland; Faculty of Science, University of Basel, Klingelbergstrasse 48, CH-4056 Basel, Switzerland.
| |
Collapse
|
10
|
Yamashita K, Shimane K, Muramoto T. Optogenetic control of cAMP oscillations reveals frequency-selective transcription factor dynamics in Dictyostelium. Development 2025; 152:dev204403. [PMID: 39775856 PMCID: PMC11829771 DOI: 10.1242/dev.204403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Oscillatory dynamics and their modulation are crucial for cellular decision-making; however, analysing these dynamics remains challenging. Here, we present a tool that combines the light-activated adenylate cyclase mPAC with the cAMP biosensor Pink Flamindo, enabling precise manipulation and real-time monitoring of cAMP oscillation frequencies in Dictyostelium. High-frequency modulation of cAMP oscillations induced cell aggregation and multicellular formation, even at low cell densities, such as a few dozen cells. At the population level, chemotactic aggregation is driven by modulated frequency signals. Additionally, modulation of cAMP frequency significantly reduced the amplitude of the shuttling behaviour of the transcription factor GtaC, demonstrating low-pass filter characteristics capable of converting subtle oscillation changes, such as from 6 min to 4 min, into gene expression. These findings enhance our understanding of frequency-selective cellular decoding and its role in cellular signalling and development.
Collapse
Affiliation(s)
- Kensuke Yamashita
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Kazuya Shimane
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| | - Tetsuya Muramoto
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba 274-8510, Japan
| |
Collapse
|
11
|
Vogt A, Paulat R, Parthier D, Just V, Szczepek M, Scheerer P, Xu Q, Möglich A, Schmitz D, Rost BR, Wenger N. Simultaneous spectral illumination of microplates for high-throughput optogenetics and photobiology. Biol Chem 2024; 405:751-763. [PMID: 39303162 DOI: 10.1515/hsz-2023-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
The biophysical characterization and engineering of optogenetic tools and photobiological systems has been hampered by the lack of efficient methods for spectral illumination of microplates for high-throughput analysis of action spectra. Current methods to determine action spectra only allow the sequential spectral illumination of individual wells. Here we present the open-source RainbowCap-system, which combines LEDs and optical filters in a standard 96-well microplate format for simultaneous and spectrally defined illumination. The RainbowCap provides equal photon flux for each wavelength, with the output of the LEDs narrowed by optical bandpass filters. We validated the RainbowCap for photoactivatable G protein-coupled receptors (opto-GPCRs) and enzymes for the control of intracellular downstream signaling. The simultaneous, spectrally defined illumination provides minimal interruption during time-series measurements, while resolving 10 nm differences in the action spectra of optogenetic proteins under identical experimental conditions. The RainbowCap is also suitable for studying the spectral dependence of light-regulated gene expression in bacteria, which requires illumination over several hours. In summary, the RainbowCap provides high-throughput spectral illumination of microplates, while its modular, customizable design allows easy adaptation to a wide range of optogenetic and photobiological applications.
Collapse
Affiliation(s)
- Arend Vogt
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Raik Paulat
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Faculty of Energy and Information, HTW-Berlin University for Applied Sciences, D-10318 Berlin, Germany
| | - Daniel Parthier
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Verena Just
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Faculty of Energy and Information, HTW-Berlin University for Applied Sciences, D-10318 Berlin, Germany
| | - Michal Szczepek
- Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Qianzhao Xu
- Department of Biochemistry, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Andreas Möglich
- Department of Biochemistry, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- 638588 German Center for Neurodegenerative Diseases (DZNE) , D-10117 Berlin, Germany
| | - Benjamin R Rost
- Neuroscience Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- 638588 German Center for Neurodegenerative Diseases (DZNE) , D-10117 Berlin, Germany
| | - Nikolaus Wenger
- Department of Neurology with Experimental Neurology, Translational Neuromodulation Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| |
Collapse
|
12
|
Castillo-Ramírez LA, Herget U, Ryu S, De Marco RJ. Early-life challenge enhances cortisol regulation in zebrafish larvae. Biol Open 2024; 13:bio061684. [PMID: 39607018 PMCID: PMC11625891 DOI: 10.1242/bio.061684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis in mammals and the hypothalamic-pituitary-interrenal (HPI) axis in fish are open systems that adapt to the environment during development. Little is known about how this adaptation begins and regulates early stress responses. We used larval zebrafish to examine the impact of prolonged forced swimming at 5 days post-fertilization (dpf), termed early-life challenge (ELC), on cortisol responses, neuropeptide expression in the nucleus preopticus (NPO), and gene transcript levels. At 6 dpf, ELC-exposed larvae showed normal baseline cortisol but reduced reactivity to an initial stressor. Conversely, they showed increased reactivity to a second stressor within the 30-min refractory period, when cortisol responses are typically suppressed. ELC larvae had fewer corticotropin-releasing hormone (crh), arginine vasopressin (avp), and oxytocin (oxt)-positive cells in the NPO, with reduced crh and avp co-expression. Gene expression analysis revealed upregulation of genes related to cortisol metabolism (hsd11b2, cyp11c1), steroidogenesis (star), and stress modulation (crh, avp, oxt). These results suggest that early environmental challenge initiates adaptive plasticity in the HPI axis, tuning cortisol regulation to balance responsiveness and protection during repeated stress. Future studies should explore the broader physiological effects of prolonged forced swimming and its long-term impact on cortisol regulation and stress-related circuits.
Collapse
Affiliation(s)
- Luis A. Castillo-Ramírez
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| | - Ulrich Herget
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, California 91125, USA
| | - Soojin Ryu
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road EX4 4QD, Exeter, UK
| | - Rodrigo J. De Marco
- Developmental Genetics of the Nervous System, Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- School of Biological and Environmental Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, L3 3AF Liverpool, UK
| |
Collapse
|
13
|
Kapetanaki SM, Coquelle N, von Stetten D, Byrdin M, Rios-Santacruz R, Bean R, Bielecki J, Boudjelida M, Fekete Z, Grime GW, Han H, Hatton C, Kantamneni S, Kharitonov K, Kim C, Kloos M, Koua FHM, de Diego Martinez I, Melo D, Rane L, Round A, Round E, Sarma A, Schubert R, Schulz J, Sikorski M, Vakili M, Valerio J, Vitas J, de Wijn R, Wrona A, Zala N, Pearson A, Dörner K, Schirò G, Garman EF, Lukács A, Weik M. Crystal structure of a bacterial photoactivated adenylate cyclase determined by serial femtosecond and serial synchrotron crystallography. IUCRJ 2024; 11:991-1006. [PMID: 39470573 PMCID: PMC11533990 DOI: 10.1107/s2052252524010170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
OaPAC is a recently discovered blue-light-using flavin adenosine dinucleotide (BLUF) photoactivated adenylate cyclase from the cyanobacterium Oscillatoria acuminata that uses adenosine triphosphate and translates the light signal into the production of cyclic adenosine monophosphate. Here, we report crystal structures of the enzyme in the absence of its natural substrate determined from room-temperature serial crystallography data collected at both an X-ray free-electron laser and a synchrotron, and we compare these structures with cryo-macromolecular crystallography structures obtained at a synchrotron by us and others. These results reveal slight differences in the structure of the enzyme due to data collection at different temperatures and X-ray sources. We further investigate the effect of the Y6W mutation in the BLUF domain, a mutation which results in a rearrangement of the hydrogen-bond network around the flavin and a notable rotation of the side chain of the critical Gln48 residue. These studies pave the way for picosecond-millisecond time-resolved serial crystallography experiments at X-ray free-electron lasers and synchrotrons in order to determine the early structural intermediates and correlate them with the well studied picosecond-millisecond spectroscopic intermediates.
Collapse
Affiliation(s)
- Sofia M. Kapetanaki
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Nicolas Coquelle
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - David von Stetten
- European Molecular Biology Laboratory (EMBL)Hamburg Unit c/o DESYNotkestrasse 8522607HamburgGermany
| | - Martin Byrdin
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Ronald Rios-Santacruz
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | | | | | - Mohamed Boudjelida
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Zsuzsana Fekete
- Department of Biophysics, Medical SchoolUniversity of PecsSzigeti Street 127624PécsHungary
| | - Geoffrey W. Grime
- Surrey Ion Beam CentreUniversity of SurreyGuildfordGU2 7XHUnited Kingdom
| | - Huijong Han
- European XFELHolzkoppel 422869SchenefeldGermany
| | - Caitlin Hatton
- Institute for Nanostructure and Solid-State PhysicsUniversität HamburgHARBOR, Luruper Chaussee 14922761HamburgGermany
| | | | | | - Chan Kim
- European XFELHolzkoppel 422869SchenefeldGermany
| | - Marco Kloos
- European XFELHolzkoppel 422869SchenefeldGermany
| | | | | | - Diogo Melo
- European XFELHolzkoppel 422869SchenefeldGermany
| | - Lukas Rane
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Adam Round
- European XFELHolzkoppel 422869SchenefeldGermany
| | | | | | | | | | | | | | | | - Jovana Vitas
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | | | | | - Ninon Zala
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Arwen Pearson
- Institute for Nanostructure and Solid-State PhysicsUniversität HamburgHARBOR, Luruper Chaussee 14922761HamburgGermany
| | | | - Giorgio Schirò
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| | - Elspeth F. Garman
- Department of BiochemistryUniversity of OxfordDorothy Crowfoot Hodgkin Building, South Parks RoadOxfordOX1 3QUUnited Kingdom
| | - András Lukács
- Department of Biophysics, Medical SchoolUniversity of PecsSzigeti Street 127624PécsHungary
| | - Martin Weik
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale38044GrenobleFrance
| |
Collapse
|
14
|
Schoofs A, Miroschnikow A, Schlegel P, Zinke I, Schneider-Mizell CM, Cardona A, Pankratz MJ. Serotonergic modulation of swallowing in a complete fly vagus nerve connectome. Curr Biol 2024; 34:4495-4512.e6. [PMID: 39270641 PMCID: PMC7616834 DOI: 10.1016/j.cub.2024.08.025] [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: 02/06/2024] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
How the body interacts with the brain to perform vital life functions, such as feeding, is a fundamental issue in physiology and neuroscience. Here, we use a whole-animal scanning transmission electron microscopy volume of Drosophila to map the neuronal circuits that connect the entire enteric nervous system to the brain via the insect vagus nerve at synaptic resolution. We identify a gut-brain feedback loop in which Piezo-expressing mechanosensory neurons in the esophagus convey food passage information to a cluster of six serotonergic neurons in the brain. Together with information on food value, these central serotonergic neurons enhance the activity of serotonin receptor 7-expressing motor neurons that drive swallowing. This elemental circuit architecture includes an axo-axonic synaptic connection from the glutamatergic motor neurons innervating the esophageal muscles onto the mechanosensory neurons that signal to the serotonergic neurons. Our analysis elucidates a neuromodulatory sensory-motor system in which ongoing motor activity is strengthened through serotonin upon completion of a biologically meaningful action, and it may represent an ancient form of motor learning.
Collapse
Affiliation(s)
- Andreas Schoofs
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Anton Miroschnikow
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Philipp Schlegel
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 TN1, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK
| | - Ingo Zinke
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | | | - Albert Cardona
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK; Janelia Research Campus, Howard Hughes Medical Institute, Helix Drive, Ashburn, VA 20147, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Place, Cambridge CB2 3EL, UK
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany.
| |
Collapse
|
15
|
Phalip A, Netser S, Wagner S. Understanding the neurobiology of social behavior through exploring brain-wide dynamics of neural activity. Neurosci Biobehav Rev 2024; 165:105856. [PMID: 39159735 DOI: 10.1016/j.neubiorev.2024.105856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Social behavior is highly complex and adaptable. It can be divided into multiple temporal stages: detection, approach, and consummatory behavior. Each stage can be further divided into several cognitive and behavioral processes, such as perceiving social cues, evaluating the social and non-social contexts, and recognizing the internal/emotional state of others. Recent studies have identified numerous brain-wide circuits implicated in social behavior and suggested the existence of partially overlapping functional brain networks underlying various types of social and non-social behavior. However, understanding the brain-wide dynamics underlying social behavior remains challenging, and several brain-scale dynamics (macro-, meso-, and micro-scale levels) need to be integrated. Here, we suggest leveraging new tools and concepts to explore social brain networks and integrate those different levels. These include studying the expression of immediate-early genes throughout the entire brain to impartially define the structure of the neuronal networks involved in a given social behavior. Then, network dynamics could be investigated using electrode arrays or multi-channel fiber photometry. Finally, tools like high-density silicon probes and miniscopes can probe neural activity in specific areas and across neuronal populations at the single-cell level.
Collapse
Affiliation(s)
- Adèle Phalip
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Shai Netser
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
16
|
Dill H, Liewald JF, Becker M, Seidenthal M, Gottschalk A. Neuropeptidergic regulation of neuromuscular signaling in larval zebrafish alters swimming behavior and synaptic transmission. iScience 2024; 27:110687. [PMID: 39252958 PMCID: PMC11381845 DOI: 10.1016/j.isci.2024.110687] [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: 01/11/2024] [Revised: 04/13/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Chemical synaptic transmission is modulated to accommodate different activity levels, thus enabling homeostatic scaling in pre- and postsynaptic compartments. In nematodes, cholinergic neurons use neuropeptide signaling to modulate synaptic vesicle content. To explore if this mechanism is conserved in vertebrates, we studied the involvement of neuropeptides in cholinergic transmission at the neuromuscular junction of larval zebrafish. Optogenetic stimulation by photoactivated adenylyl cyclase evoked locomotion. We generated mutants lacking the neuropeptide-processing enzyme carboxypeptidase E (cpe), and the most abundant neuropeptide precursor in motor neurons, tachykinin (tac1). Both mutants showed exaggerated locomotion after photostimulation. Recording excitatory postsynaptic currents demonstrated overall larger amplitudes in the wild type. Exaggerated locomotion in the mutants thus reflected upscaling of postsynaptic excitability. Both mutant muscles expressed more nicotinic acetylcholine receptors (nAChRs) on their surface; thus, neuropeptide signaling regulates synaptic transmitter output in zebrafish motor neurons, and muscle cells homeostatically regulate nAChR surface expression, compensating reduced presynaptic input.
Collapse
Affiliation(s)
- Holger Dill
- Buchmann Institute, Goethe University, Max-von-Laue-Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Jana F Liewald
- Buchmann Institute, Goethe University, Max-von-Laue-Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Michelle Becker
- Buchmann Institute, Goethe University, Max-von-Laue-Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Marius Seidenthal
- Buchmann Institute, Goethe University, Max-von-Laue-Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| | - Alexander Gottschalk
- Buchmann Institute, Goethe University, Max-von-Laue-Strasse 15, D-60438 Frankfurt, Germany
- Institute for Biophysical Chemistry, Goethe University, Max-von-Laue-Strasse 9, D-60438 Frankfurt, Germany
| |
Collapse
|
17
|
Bolonduro OA, Chen Z, Fucetola CP, Lai YR, Cote M, Kajola RO, Rao AA, Liu H, Tzanakakis ES, Timko BP. An Integrated Optogenetic and Bioelectronic Platform for Regulating Cardiomyocyte Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402236. [PMID: 39054679 PMCID: PMC11423186 DOI: 10.1002/advs.202402236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/24/2024] [Indexed: 07/27/2024]
Abstract
Bioelectronic medicine is emerging as a powerful approach for restoring lost endogenous functions and addressing life-altering maladies such as cardiac disorders. Systems that incorporate both modulation of cellular function and recording capabilities can enhance the utility of these approaches and their customization to the needs of each patient. Here we report an integrated optogenetic and bioelectronic platform for stable and long-term stimulation and monitoring of cardiomyocyte function in vitro. Optical inputs are achieved through the expression of a photoactivatable adenylyl cyclase, that when irradiated with blue light causes a dose-dependent and time-limited increase in the secondary messenger cyclic adenosine monophosphate with subsequent rise in autonomous cardiomyocyte beating rate. Bioelectronic readouts are obtained through a multi-electrode array that measures real-time electrophysiological responses at 32 spatially-distinct locations. Irradiation at 27 µW mm-2 results in a 14% elevation of the beating rate within 20-25 min, which remains stable for at least 2 h. The beating rate can be cycled through "on" and "off" light states, and its magnitude is a monotonic function of irradiation intensity. The integrated platform can be extended to stretchable and flexible substrates, and can open new avenues in bioelectronic medicine, including closed-loop systems for cardiac regulation and intervention, for example, in the context of arrythmias.
Collapse
Affiliation(s)
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
| | - Corey P Fucetola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Yan-Ru Lai
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Megan Cote
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Rofiat O Kajola
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Akshita A Rao
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Haitao Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China
| | - Emmanuel S Tzanakakis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Chemical and Biological Engineering, Tufts University, Medford, MA, 02155, USA
- Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, 02111, USA
| | - Brian P Timko
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
18
|
Mukherjee S, Klarenbeek J, El Oualid F, van den Broek B, Jalink K. "Radical" differences between two FLIM microscopes affect interpretation of cell signaling dynamics. iScience 2024; 27:110268. [PMID: 39036041 PMCID: PMC11257777 DOI: 10.1016/j.isci.2024.110268] [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: 09/12/2023] [Revised: 02/12/2024] [Accepted: 06/11/2024] [Indexed: 07/23/2024] Open
Abstract
The outcome of cell signaling depends not only on signal strength but also on temporal progression. We use Fluorescence Lifetime Imaging of Resonance Energy Transfer (FLIM/FRET) biosensors to investigate intracellular signaling dynamics. We examined the β1 receptor-Gαs-cAMP signaling axis using both widefield frequency domain FLIM (fdFLIM) and fast confocal time-correlated single photon counting (TCSPC) setups. Unexpectedly, we observed that fdFLIM revealed transient cAMP responses in HeLa and Cos7 cells, contrasting with sustained responses as detected with TCSPC. Investigation revealed no light-induced effects on cAMP generation or breakdown. Rather, folic acid present in the imaging medium appeared to be the culprit, as its excitation with blue light sensitized degradation of β1 agonists. Our findings highlight the impact of subtle phototoxicity on experimental outcomes, advocating confocal TCSPC for reliable analysis of response kinetics and stressing the need for full disclosure of chemical formulations by scientific vendors.
Collapse
Affiliation(s)
- Sravasti Mukherjee
- Department of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, the Netherlands
- Swammerdam Institute of Life Sciences, University of Amsterdam, Science Park 904, Amsterdam 1098 XH, the Netherlands
| | - Jeffrey Klarenbeek
- Department of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, the Netherlands
| | - Farid El Oualid
- UbiQ Bio B.V., Science Park 301, Amsterdam 1098 XH, the Netherlands
| | - Bram van den Broek
- Department of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, the Netherlands
- BioImaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, the Netherlands
| | - Kees Jalink
- Department of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066CX, the Netherlands
- Swammerdam Institute of Life Sciences, University of Amsterdam, Science Park 904, Amsterdam 1098 XH, the Netherlands
| |
Collapse
|
19
|
Joshi SHN, Jenkins C, Ulaeto D, Gorochowski TE. Accelerating Genetic Sensor Development, Scale-up, and Deployment Using Synthetic Biology. BIODESIGN RESEARCH 2024; 6:0037. [PMID: 38919711 PMCID: PMC11197468 DOI: 10.34133/bdr.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024] Open
Abstract
Living cells are exquisitely tuned to sense and respond to changes in their environment. Repurposing these systems to create engineered biosensors has seen growing interest in the field of synthetic biology and provides a foundation for many innovative applications spanning environmental monitoring to improved biobased production. In this review, we present a detailed overview of currently available biosensors and the methods that have supported their development, scale-up, and deployment. We focus on genetic sensors in living cells whose outputs affect gene expression. We find that emerging high-throughput experimental assays and evolutionary approaches combined with advanced bioinformatics and machine learning are establishing pipelines to produce genetic sensors for virtually any small molecule, protein, or nucleic acid. However, more complex sensing tasks based on classifying compositions of many stimuli and the reliable deployment of these systems into real-world settings remain challenges. We suggest that recent advances in our ability to precisely modify nonmodel organisms and the integration of proven control engineering principles (e.g., feedback) into the broader design of genetic sensing systems will be necessary to overcome these hurdles and realize the immense potential of the field.
Collapse
Affiliation(s)
| | - Christopher Jenkins
- CBR Division, Defence Science and Technology Laboratory, Porton Down, Wiltshire SP4 0JQ, UK
| | - David Ulaeto
- CBR Division, Defence Science and Technology Laboratory, Porton Down, Wiltshire SP4 0JQ, UK
| | - Thomas E. Gorochowski
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
- BrisEngBio,
School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| |
Collapse
|
20
|
Nagpal J, Eachus H, Lityagina O, Ryu S. Optogenetic induction of chronic glucocorticoid exposure in early-life leads to blunted stress-response in larval zebrafish. Eur J Neurosci 2024; 59:3134-3146. [PMID: 38602078 DOI: 10.1111/ejn.16301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 04/12/2024]
Abstract
Early life stress (ELS) exposure alters stress susceptibility in later life and affects vulnerability to stress-related disorders, but how ELS changes the long-lasting responsiveness of the stress system is not well understood. Zebrafish provides an opportunity to study conserved mechanisms underlying the development and function of the stress response that is regulated largely by the neuroendocrine hypothalamus-pituitary-adrenal/interrenal (HPA/I) axis, with glucocorticoids (GC) as the final effector. In this study, we established a method to chronically elevate endogenous GC levels during early life in larval zebrafish. To this end, we employed an optogenetic actuator, beggiatoa photoactivated adenylyl cyclase, specifically expressed in the interrenal cells of zebrafish and demonstrate that its chronic activation leads to hypercortisolaemia and dampens the acute-stress evoked cortisol levels, across a variety of stressor modalities during early life. This blunting of stress-response was conserved in ontogeny at a later developmental stage. Furthermore, we observe a strong reduction of proopiomelanocortin (pomc)-expression in the pituitary as well as upregulation of fkbp5 gene expression. Going forward, we propose that this model can be leveraged to tease apart the mechanisms underlying developmental programming of the HPA/I axis by early-life GC exposure and its implications for vulnerability and resilience to stress in adulthood.
Collapse
Affiliation(s)
- Jatin Nagpal
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- APC Microbiome Ireland and School of Pharmacy and Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Olga Lityagina
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Soojin Ryu
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
21
|
Ohlendorf R, Li N, Phi Van VD, Schwalm M, Ke Y, Dawson M, Jiang Y, Das S, Stallings B, Zheng WT, Jasanoff A. Imaging bioluminescence by detecting localized haemodynamic contrast from photosensitized vasculature. Nat Biomed Eng 2024; 8:775-786. [PMID: 38730257 PMCID: PMC12121613 DOI: 10.1038/s41551-024-01210-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/30/2024] [Indexed: 05/12/2024]
Abstract
Bioluminescent probes are widely used to monitor biomedically relevant processes and cellular targets in living animals. However, the absorption and scattering of visible light by tissue drastically limit the depth and resolution of the detection of luminescence. Here we show that bioluminescent sources can be detected with magnetic resonance imaging by leveraging the light-mediated activation of vascular cells expressing a photosensitive bacterial enzyme that causes the conversion of bioluminescent emission into local changes in haemodynamic contrast. In the brains of rats with photosensitized vasculature, we used magnetic resonance imaging to volumetrically map bioluminescent xenografts and cell populations virally transduced to express luciferase. Detecting bioluminescence-induced haemodynamic signals from photosensitized vasculature will extend the applications of bioluminescent probes.
Collapse
Affiliation(s)
- Robert Ohlendorf
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Max Planck Institute for Biological Cybernetics, Tubingen, Germany
| | - Nan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Advanced Imaging Research Center and Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Valerie Doan Phi Van
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miriam Schwalm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yuting Ke
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miranda Dawson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ying Jiang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sayani Das
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brenna Stallings
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Wen Ting Zheng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alan Jasanoff
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
22
|
Sakai K, Aoki K, Goto Y. Live-cell fluorescence imaging and optogenetic control of PKA kinase activity in fission yeast Schizosaccharomyces pombe. Yeast 2024; 41:349-363. [PMID: 38583078 DOI: 10.1002/yea.3937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/21/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024] Open
Abstract
The cAMP-PKA signaling pathway plays a crucial role in sensing and responding to nutrient availability in the fission yeast Schizosaccharomyces pombe. This pathway monitors external glucose levels to control cell growth and sexual differentiation. However, the temporal dynamics of the cAMP-PKA pathway in response to external stimuli remains unclear mainly due to the lack of tools to quantitatively visualize the activity of the pathway. Here, we report the development of the kinase translocation reporter (KTR)-based biosensor spPKA-KTR1.0, which allows us to measure the dynamics of PKA activity in fission yeast cells. The spPKA-KTR1.0 is derived from the transcription factor Rst2, which translocates from the nucleus to the cytoplasm upon PKA activation. We found that spPKA-KTR1.0 translocates between the nucleus and cytoplasm in a cAMP-PKA pathway-dependent manner, indicating that the spPKA-KTR1.0 is a reliable indicator of the PKA activity in fission yeast cells. In addition, we implemented a system that simultaneously visualizes and manipulates the cAMP-PKA signaling dynamics by introducing bPAC, a photoactivatable adenylate cyclase, in combination with spPKA-KTR1.0. This system offers an opportunity for investigating the role of the signaling dynamics of the cAMP-PKA pathway in fission yeast cells with higher temporal resolution.
Collapse
Affiliation(s)
- Keiichiro Sakai
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuhiro Aoki
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
- Center for Living Systems Information Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Division of Integrated Life Science, Department of Gene Mechanisms, Laboratory of Cell Cycle Regulation, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yuhei Goto
- Quantitative Biology Research Group, Department of Creative Research, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Division of Quantitative Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| |
Collapse
|
23
|
Çoban B, Poppinga H, Rachad EY, Geurten B, Vasmer D, Rodriguez Jimenez FJ, Gadgil Y, Deimel SH, Alyagor I, Schuldiner O, Grunwald Kadow IC, Riemensperger TD, Widmann A, Fiala A. The caloric value of food intake structurally adjusts a neuronal mushroom body circuit mediating olfactory learning in Drosophila. Learn Mem 2024; 31:a053997. [PMID: 38862177 PMCID: PMC11199950 DOI: 10.1101/lm.053997.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/10/2024] [Indexed: 06/13/2024]
Abstract
Associative learning enables the adaptive adjustment of behavioral decisions based on acquired, predicted outcomes. The valence of what is learned is influenced not only by the learned stimuli and their temporal relations, but also by prior experiences and internal states. In this study, we used the fruit fly Drosophila melanogaster to demonstrate that neuronal circuits involved in associative olfactory learning undergo restructuring during extended periods of low-caloric food intake. Specifically, we observed a decrease in the connections between specific dopaminergic neurons (DANs) and Kenyon cells at distinct compartments of the mushroom body. This structural synaptic plasticity was contingent upon the presence of allatostatin A receptors in specific DANs and could be mimicked optogenetically by expressing a light-activated adenylate cyclase in exactly these DANs. Importantly, we found that this rearrangement in synaptic connections influenced aversive, punishment-induced olfactory learning but did not impact appetitive, reward-based learning. Whether induced by prolonged low-caloric conditions or optogenetic manipulation of cAMP levels, this synaptic rearrangement resulted in a reduction of aversive associative learning. Consequently, the balance between positive and negative reinforcing signals shifted, diminishing the ability to learn to avoid odor cues signaling negative outcomes. These results exemplify how a neuronal circuit required for learning and memory undergoes structural plasticity dependent on prior experiences of the nutritional value of food.
Collapse
Affiliation(s)
- Büşra Çoban
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | - Haiko Poppinga
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | - El Yazid Rachad
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | - Bart Geurten
- Department of Zoology, Otago University, Dunedin 9016, New Zealand
| | - David Vasmer
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | | | - Yogesh Gadgil
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | | | - Idan Alyagor
- Department of Molecular Cell Biology, Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | | | - Annekathrin Widmann
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| | - André Fiala
- Molecular Neurobiology of Behavior, University of Göttingen, 37077 Göttingen, Germany
| |
Collapse
|
24
|
Nagase M, Nagashima T, Hamada S, Morishima M, Tohyama S, Arima-Yoshida F, Hiyoshi K, Hirano T, Ohtsuka T, Watabe AM. All-optical presynaptic plasticity induction by photoactivated adenylyl cyclase targeted to axon terminals. CELL REPORTS METHODS 2024; 4:100740. [PMID: 38521059 PMCID: PMC11045876 DOI: 10.1016/j.crmeth.2024.100740] [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: 04/12/2023] [Revised: 10/08/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024]
Abstract
Intracellular signaling plays essential roles in various cell types. In the central nervous system, signaling cascades are strictly regulated in a spatiotemporally specific manner to govern brain function; for example, presynaptic cyclic adenosine monophosphate (cAMP) can enhance the probability of neurotransmitter release. In the last decade, channelrhodopsin-2 has been engineered for subcellular targeting using localization tags, but optogenetic tools for intracellular signaling are not well developed. Therefore, we engineered a selective presynaptic fusion tag for photoactivated adenylyl cyclase (bPAC-Syn1a) and found its high localization at presynaptic terminals. Furthermore, an all-optical electrophysiological method revealed rapid and robust short-term potentiation by bPAC-Syn1a at brain stem-amygdala synapses in acute brain slices. Additionally, bPAC-Syn1a modulated mouse immobility behavior. These results indicate that bPAC-Syn1a can manipulate presynaptic cAMP signaling in vitro and in vivo. The all-optical manipulation technique developed in this study can help further elucidate the dynamic regulation of various cellular functions.
Collapse
Affiliation(s)
- Masashi Nagase
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Takashi Nagashima
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Shun Hamada
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Mieko Morishima
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Suguru Tohyama
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Fumiko Arima-Yoshida
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Kanae Hiyoshi
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan
| | - Tomoha Hirano
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan.
| | - Ayako M Watabe
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba 277-8567, Japan.
| |
Collapse
|
25
|
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: 6] [Impact Index Per Article: 6.0] [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
|
26
|
Chen Z, Stoukides DM, Tzanakakis ES. Light-Mediated Enhancement of Glucose-Stimulated Insulin Release of Optogenetically Engineered Human Pancreatic Beta-Cells. ACS Synth Biol 2024; 13:825-836. [PMID: 38377949 PMCID: PMC10949932 DOI: 10.1021/acssynbio.3c00653] [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: 10/27/2023] [Revised: 01/27/2024] [Accepted: 02/05/2024] [Indexed: 02/22/2024]
Abstract
Enhancement of glucose-stimulated insulin secretion (GSIS) in exogenously delivered pancreatic β-cells is desirable, for example, to overcome the insulin resistance manifested in type 2 diabetes or to reduce the number of β-cells for supporting homeostasis of blood sugar in type 1 diabetes. Optogenetically engineered cells can potentiate their function with exposure to light. Given that cyclic adenosine monophosphate (cAMP) mediates GSIS, we surmised that optoamplification of GSIS is feasible in human β-cells carrying a photoactivatable adenylyl cyclase (PAC). To this end, human EndoC-βH3 cells were engineered to express a blue-light-activated PAC, and a workflow was established combining the scalable manufacturing of pseudoislets (PIs) with efficient adenoviral transduction, resulting in over 80% of cells carrying PAC. Changes in intracellular cAMP and GSIS were determined with the photoactivation of PAC in vitro as well as after encapsulation and implantation in mice with streptozotocin-induced diabetes. cAMP rapidly rose in β-cells expressing PAC with illumination and quickly declined upon its termination. Light-induced amplification in cAMP was concomitant with a greater than 2-fold GSIS vs β-cells without PAC in elevated glucose. The enhanced GSIS retained its biphasic pattern, and the rate of oxygen consumption remained unchanged. Diabetic mice receiving the engineered β-cell PIs exhibited improved glucose tolerance upon illumination compared to those kept in the dark or not receiving cells. The findings support the use of optogenetics for molecular customization of the β-cells toward better treatments for diabetes without the adverse effects of pharmacological approaches.
Collapse
Affiliation(s)
- Zijing Chen
- Department
of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Demetrios M. Stoukides
- Department
of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Emmanuel S. Tzanakakis
- Department
of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Department
of Developmental, Molecular and Cell Biology, Tufts University School of Medicine, Boston, Massachusetts 02111, United States
- Graduate
Program in Pharmacology and Experimental Therapeutics and Pharmacology
and Drug Development, Tufts University School
of Medicine, Boston, Massachusetts 02111, United States
- Clinical
and Translational Science Institute, Tufts
Medical Center, Boston, Massachusetts 02111, United States
| |
Collapse
|
27
|
Xu Q, Vogt A, Frechen F, Yi C, Küçükerden M, Ngum N, Sitjà-Roqueta L, Greiner A, Parri R, Masana M, Wenger N, Wachten D, Möglich A. Engineering Bacteriophytochrome-coupled Photoactivated Adenylyl Cyclases for Enhanced Optogenetic cAMP Modulation. J Mol Biol 2024; 436:168257. [PMID: 37657609 DOI: 10.1016/j.jmb.2023.168257] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Sensory photoreceptors abound in nature and enable organisms to adapt behavior, development, and physiology to environmental light. In optogenetics, photoreceptors allow spatiotemporally precise, reversible, and non-invasive control by light of cellular processes. Notwithstanding the development of numerous optogenetic circuits, an unmet demand exists for efficient systems sensitive to red light, given its superior penetration of biological tissue. Bacteriophytochrome photoreceptors sense the ratio of red and far-red light to regulate the activity of enzymatic effector modules. The recombination of bacteriophytochrome photosensor modules with cyclase effectors underlies photoactivated adenylyl cyclases (PAC) that catalyze the synthesis of the ubiquitous second messenger 3', 5'-cyclic adenosine monophosphate (cAMP). Via homologous exchanges of the photosensor unit, we devised novel PACs, with the variant DmPAC exhibiting 40-fold activation of cyclase activity under red light, thus surpassing previous red-light-responsive PACs. Modifications of the PHY tongue modulated the responses to red and far-red light. Exchanges of the cyclase effector offer an avenue to further enhancing PACs but require optimization of the linker to the photosensor. DmPAC and a derivative for 3', 5'-cyclic guanosine monophosphate allow the manipulation of cyclic-nucleotide-dependent processes in mammalian cells by red light. Taken together, we advance the optogenetic control of second-messenger signaling and provide insight into the signaling and design of bacteriophytochrome receptors.
Collapse
Affiliation(s)
- Qianzhao Xu
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | - Arend Vogt
- Charité - University Medicine Berlin, Department of Neurology with Experimental Neurology, 10117 Berlin, Germany. https://twitter.com/ArendVogt
| | - Fabian Frechen
- Institute of Innate Immunity, University of Bonn, 53127 Bonn, Germany
| | - Chengwei Yi
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany
| | - Melike Küçükerden
- Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Neville Ngum
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, United Kingdom
| | - Laia Sitjà-Roqueta
- Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Andreas Greiner
- Macromolecular Chemistry and Bavarian Polymer Institute, University of Bayreuth, Bayreuth 95440, Germany
| | - Rhein Parri
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, United Kingdom
| | - Mercè Masana
- Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain. https://twitter.com/mercemasana
| | - Nikolaus Wenger
- Charité - University Medicine Berlin, Department of Neurology with Experimental Neurology, 10117 Berlin, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, University of Bonn, 53127 Bonn, Germany. https://twitter.com/DagmarWachten
| | - Andreas Möglich
- Department of Biochemistry, University of Bayreuth, 95447 Bayreuth, Germany; Bayreuth Center for Biochemistry & Molecular Biology, Universität Bayreuth, 95447 Bayreuth, Germany; North-Bavarian NMR Center, Universität Bayreuth, 95447 Bayreuth, Germany.
| |
Collapse
|
28
|
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
|
29
|
Chretien A, Nagel MF, Botha S, de Wijn R, Brings L, Dörner K, Han H, Koliyadu JCP, Letrun R, Round A, Sato T, Schmidt C, Secareanu RC, von Stetten D, Vakili M, Wrona A, Bean R, Mancuso A, Schulz J, Pearson AR, Kottke T, Lorenzen K, Schubert R. Light-induced Trp in/Met out Switching During BLUF Domain Activation in ATP-bound Photoactivatable Adenylate Cyclase OaPAC. J Mol Biol 2024; 436:168439. [PMID: 38185322 DOI: 10.1016/j.jmb.2024.168439] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/28/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
The understanding of signal transduction mechanisms in photoreceptor proteins is essential for elucidating how living organisms respond to light as environmental stimuli. In this study, we investigated the ATP binding, photoactivation and signal transduction process in the photoactivatable adenylate cyclase from Oscillatoria acuminata (OaPAC) upon blue light excitation. Structural models with ATP bound in the active site of native OaPAC at cryogenic as well as room temperature are presented. ATP is found in one conformation at cryogenic- and in two conformations at ambient-temperature, and is bound in an energetically unfavorable conformation for the conversion to cAMP. However, FTIR spectroscopic experiments confirm that this conformation is the native binding mode in dark state OaPAC and that transition to a productive conformation for ATP turnover only occurs after light activation. A combination of time-resolved crystallography experiments at synchrotron and X-ray Free Electron Lasers sheds light on the early events around the Flavin Adenine Dinucleotide (FAD) chromophore in the light-sensitive BLUF domain of OaPAC. Early changes involve the highly conserved amino acids Tyr6, Gln48 and Met92. Crucially, the Gln48 side chain performs a 180° rotation during activation, leading to the stabilization of the FAD chromophore. Cryo-trapping experiments allowed us to investigate a late light-activated state of the reaction and revealed significant conformational changes in the BLUF domain around the FAD chromophore. In particular, a Trpin/Metout transition upon illumination is observed for the first time in the BLUF domain and its role in signal transmission via α-helix 3 and 4 in the linker region between sensor and effector domain is discussed.
Collapse
Affiliation(s)
- Anaïs Chretien
- European XFEL GmbH, Schenefeld, Germany; Department of Chemistry, Universität Hamburg, Hamburg, Germany
| | - Marius F Nagel
- Department of Chemistry and Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Sabine Botha
- Department of Physics, Arizona State University, Tempe, AZ 85287-1504, USA; Biodesign Center for Applied Structural Discovery, Arizona State University, Tempe, AZ 85287-5001, USA
| | | | | | | | | | | | | | | | | | | | | | - David von Stetten
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | | | | | | | | | | | - Arwen R Pearson
- Institute for Nanostructure and Solid-State Physics, Universität Hamburg, Hamburg, Germany
| | - Tilman Kottke
- Department of Chemistry and Medical School OWL, Bielefeld University, Bielefeld, Germany
| | | | | |
Collapse
|
30
|
Zhang Y, Gao J, Li N, Xu P, Qu S, Cheng J, Wang M, Li X, Song Y, Xiao F, Yang X, Liu J, Hong H, Mu R, Li X, Wang Y, Xu H, Xie Y, Gao T, Wang G, Aa J. Targeting cAMP in D1-MSNs in the nucleus accumbens, a new rapid antidepressant strategy. Acta Pharm Sin B 2024; 14:667-681. [PMID: 38322327 PMCID: PMC10840425 DOI: 10.1016/j.apsb.2023.12.005] [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: 06/13/2023] [Revised: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 02/08/2024] Open
Abstract
Studies have suggested that the nucleus accumbens (NAc) is implicated in the pathophysiology of major depression; however, the regulatory strategy that targets the NAc to achieve an exclusive and outstanding anti-depression benefit has not been elucidated. Here, we identified a specific reduction of cyclic adenosine monophosphate (cAMP) in the subset of dopamine D1 receptor medium spiny neurons (D1-MSNs) in the NAc that promoted stress susceptibility, while the stimulation of cAMP production in NAc D1-MSNs efficiently rescued depression-like behaviors. Ketamine treatment enhanced cAMP both in D1-MSNs and dopamine D2 receptor medium spiny neurons (D2-MSNs) of depressed mice, however, the rapid antidepressant effect of ketamine solely depended on elevating cAMP in NAc D1-MSNs. We discovered that a higher dose of crocin markedly increased cAMP in the NAc and consistently relieved depression 24 h after oral administration, but not a lower dose. The fast onset property of crocin was verified through multicenter studies. Moreover, crocin specifically targeted at D1-MSN cAMP signaling in the NAc to relieve depression and had no effect on D2-MSN. These findings characterize a new strategy to achieve an exclusive and outstanding anti-depression benefit by elevating cAMP in D1-MSNs in the NAc, and provide a potential rapid antidepressant drug candidate, crocin.
Collapse
Affiliation(s)
- Yue Zhang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jingwen Gao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Na Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Xu
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Shimeng Qu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jinqian Cheng
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Mingrui Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xueru Li
- School of Foreign Languages, China Pharmaceutical University, Nanjing 211198, China
| | - Yaheng Song
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Xiao
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyu Yang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jihong Liu
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Ronghao Mu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Youmei Wang
- Key Laboratory of Drug Monitoring and Control, Drug Intelligence and Forensic Center, Ministry of Public Security, Beijing 100193, China
| | - Hui Xu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Xie
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Tianming Gao
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Jiye Aa
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, Research Unit of PK–PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
31
|
Pizzoni A, Naim N, Zhang X, Altschuler DL. Mapping the Cellular Distribution of an Optogenetic Protein Using a Light-Stimulation Grid. J Vis Exp 2024:10.3791/65471. [PMID: 38345221 PMCID: PMC11536799 DOI: 10.3791/65471] [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: 02/15/2024] Open
Abstract
Our goal was to accurately track the cellular distribution of an optogenetic protein and evaluate its functionality within a specific cytoplasmic location. To achieve this, we co-transfected cells with nuclear-targeted cAMP sensors and our laboratory-developed optogenetic protein, bacterial photoactivatable adenylyl cyclase-nanoluciferase (bPAC-nLuc). bPAC-nLuc, when stimulated with 445 nm light or luciferase substrates, generates adenosine 3',5'-cyclic monophosphate (cAMP). We employed a solid-state laser illuminator connected to a point scanning system that allowed us to create a grid/matrix pattern of small illuminated spots (~1 µm2) throughout the cytoplasm of HC-1 cells. By doing so, we were able to effectively track the distribution of nuclear-targeted bPAC-nLuc and generate a comprehensive cAMP response map. This map accurately represented the cellular distribution of bPAC-nLuc, and its response to light stimulation varied according to the amount of protein in the illuminated spot. This innovative approach contributes to the expanding toolkit of techniques available for investigating cellular optogenetic proteins. The ability to map its distribution and response with high precision has far-reaching potential and could advance various fields of research.
Collapse
Affiliation(s)
- Alejandro Pizzoni
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine
| | - Nyla Naim
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine; Addgene
| | - Xuefeng Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine
| | - Daniel L Altschuler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine;
| |
Collapse
|
32
|
Luyben TT, Rai J, Zhou B, Li H, Okamoto K. Two-Photon FRET/FLIM Imaging of Cerebral Neurons. Methods Mol Biol 2024; 2794:33-43. [PMID: 38630218 DOI: 10.1007/978-1-0716-3810-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Two-photon FRET (Förster resonance energy transfer) and FLIM (fluorescence lifetime imaging microscopy) enable the detection of FRET changes of fluorescence reporters in deep brain tissues, which provide a valuable approach for monitoring target molecular dynamics and functions. Here, we describe two-photon FRET and FLIM imaging techniques that allow us to visualize endogenous and optogenetically induced cAMP dynamics in living neurons with genetically engineered FRET-based cAMP reporters.
Collapse
Affiliation(s)
- Thomas T Luyben
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jayant Rai
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bingyue Zhou
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hang Li
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Kenichi Okamoto
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
33
|
Zhou F, Tichy AM, Imambocus BN, Sakharwade S, Rodriguez Jimenez FJ, González Martínez M, Jahan I, Habib M, Wilhelmy N, Burre V, Lömker T, Sauter K, Helfrich-Förster C, Pielage J, Grunwald Kadow IC, Janovjak H, Soba P. Optimized design and in vivo application of optogenetically functionalized Drosophila dopamine receptors. Nat Commun 2023; 14:8434. [PMID: 38114457 PMCID: PMC10730509 DOI: 10.1038/s41467-023-43970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Neuromodulatory signaling via G protein-coupled receptors (GPCRs) plays a pivotal role in regulating neural network function and animal behavior. The recent development of optogenetic tools to induce G protein-mediated signaling provides the promise of acute and cell type-specific manipulation of neuromodulatory signals. However, designing and deploying optogenetically functionalized GPCRs (optoXRs) with accurate specificity and activity to mimic endogenous signaling in vivo remains challenging. Here we optimize the design of optoXRs by considering evolutionary conserved GPCR-G protein interactions and demonstrate the feasibility of this approach using two Drosophila Dopamine receptors (optoDopRs). These optoDopRs exhibit high signaling specificity and light sensitivity in vitro. In vivo, we show receptor and cell type-specific effects of dopaminergic signaling in various behaviors, including the ability of optoDopRs to rescue the loss of the endogenous receptors. This work demonstrates that optoXRs can enable optical control of neuromodulatory receptor-specific signaling in functional and behavioral studies.
Collapse
Affiliation(s)
- Fangmin Zhou
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
| | - Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Shreyas Sakharwade
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Francisco J Rodriguez Jimenez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Marco González Martínez
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Ishrat Jahan
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
| | - Margarita Habib
- Neurobiology and Genetics, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Nina Wilhelmy
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Vanessa Burre
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Tatjana Lömker
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Kathrin Sauter
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | | | - Jan Pielage
- Division of Neurobiology and Zoology, RPTU University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Ilona C Grunwald Kadow
- Institute of Physiology II, University Clinic Bonn (UKB), University of Bonn, 53115, Bonn, Germany
- ZIEL-Institute of Life and Health, Technical University of Munich, School of Life Sciences, 85354, Freising, Germany
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 3800, Clayton, Victoria, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 3800, Clayton, Victoria, Australia
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 5042, Bedford Park, South Australia, Australia
| | - Peter Soba
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany.
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany.
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
34
|
Bolonduro OA, Chen Z, Lai YR, Cote M, Rao AA, Liu H, Tzanakakis ES, Timko BP. An Integrated Optogenetic and Bioelectronic Platform for Regulating Cardiomyocyte Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571704. [PMID: 38168441 PMCID: PMC10760153 DOI: 10.1101/2023.12.15.571704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
We report an integrated optogenetic and bioelectronic platform for stable and long-term modulation and monitoring of cardiomyocyte function in vitro. Optogenetic inputs were achieved through expression of a photoactivatable adenylyl cyclase (bPAC), that when activated by blue light caused a dose-dependent and time-limited increase in autonomous cardiomyocyte beat rate. Bioelectronic readouts were achieved through an integrated planar multi-electrode array (MEA) that provided real-time readouts of electrophysiological activity from 32 spatially-distinct locations. Irradiation at 27 μW/mm2 resulted in a ca. 14% increase in beat rate within 20-25 minutes, which remained stable for at least 2 hours. The beating rate could be cycled through repeated "on" and "off' states, and its magnitude was a monotonic function of irradiation intensity. Our integrated platform opens new avenues in bioelectronic medicine, including closed-loop feedback systems, with potential applications for cardiac regulation including arrhythmia diagnosis and intervention.
Collapse
Affiliation(s)
| | - Zijing Chen
- Department of Chemical and Biological Engineering, Tufts University
| | - Yan-Ru Lai
- Department of Biomedical Engineering, Tufts University
| | - Megan Cote
- Department of Biomedical Engineering, Tufts University
| | | | - Haitao Liu
- Department of Biomedical Engineering, Tufts University
- General Surgery Department, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University
- Cell, Molecular and Developmental Biology, Graduate School of Biomedical Sciences, Tufts University
- Clinical and Translational Science Institute, Tufts Medical Center
| | | |
Collapse
|
35
|
Atkins M, Wurmser M, Darmon M, Roche F, Nicol X, Métin C. CXCL12 targets the primary cilium cAMP/cGMP ratio to regulate cell polarity during migration. Nat Commun 2023; 14:8003. [PMID: 38049397 PMCID: PMC10695954 DOI: 10.1038/s41467-023-43645-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/15/2023] [Indexed: 12/06/2023] Open
Abstract
Directed cell migration requires sustained cell polarisation. In migrating cortical interneurons, nuclear movements are directed towards the centrosome that organises the primary cilium signalling hub. Primary cilium-elicited signalling, and how it affects migration, remain however ill characterised. Here, we show that altering cAMP/cGMP levels in the primary cilium by buffering cAMP, cGMP or by locally increasing cAMP, influences the polarity and directionality of migrating interneurons, whereas buffering cAMP or cGMP in the apposed centrosome compartment alters their motility. Remarkably, we identify CXCL12 as a trigger that targets the ciliary cAMP/cGMP ratio to promote sustained polarity and directed migration. We thereby uncover cAMP/cGMP levels in the primary cilium as a major target of extrinsic cues and as the steering wheel of neuronal migration.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM UMR-S 1270; Institut du Fer à Moulin, Sorbonne Université, F-75005, Paris, France.
| | - Maud Wurmser
- Institut de la Vision, Sorbonne Université, INSERM CNRS, F-75012, Paris, France
| | - Michèle Darmon
- INSERM UMR-S 1270; Institut du Fer à Moulin, Sorbonne Université, F-75005, Paris, France
| | - Fiona Roche
- Institut de la Vision, Sorbonne Université, INSERM CNRS, F-75012, Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM CNRS, F-75012, Paris, France
| | - Christine Métin
- INSERM UMR-S 1270; Institut du Fer à Moulin, Sorbonne Université, F-75005, Paris, France.
| |
Collapse
|
36
|
Westbrook ER, Lenn T, Chubb JR, Antolović V. Collective signalling drives rapid jumping between cell states. Development 2023; 150:dev201946. [PMID: 37921687 PMCID: PMC10730084 DOI: 10.1242/dev.201946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023]
Abstract
Development can proceed in 'fits and starts', with rapid transitions between cell states involving concerted transcriptome-wide changes in gene expression. However, it is not clear how these transitions are regulated in complex cell populations, in which cells receive multiple inputs. We address this issue using Dictyostelium cells undergoing development in their physiological niche. A continuous single cell transcriptomics time series identifies a sharp 'jump' in global gene expression marking functionally different cell states. By simultaneously imaging the physiological dynamics of transcription and signalling, we show the jump coincides with the onset of collective oscillations of cAMP. Optogenetic control of cAMP pulses shows that different jump genes respond to distinct dynamic features of signalling. Late jump gene expression changes are almost completely dependent on cAMP, whereas transcript changes at the onset of the jump require additional input. The coupling of collective signalling with gene expression is a potentially powerful strategy to drive robust cell state transitions in heterogeneous signalling environments. Based on the context of the jump, we also conclude that sharp gene expression transitions may not be sufficient for commitment.
Collapse
Affiliation(s)
- Elizabeth R. Westbrook
- UCL Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Tchern Lenn
- UCL Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jonathan R. Chubb
- UCL Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Vlatka Antolović
- UCL Laboratory for Molecular Cell Biology and Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
37
|
Berndt A, Cai D, Cohen A, Juarez B, Iglesias JT, Xiong H, Qin Z, Tian L, Slesinger PA. Current Status and Future Strategies for Advancing Functional Circuit Mapping In Vivo. J Neurosci 2023; 43:7587-7598. [PMID: 37940594 PMCID: PMC10634581 DOI: 10.1523/jneurosci.1391-23.2023] [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: 07/21/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 11/10/2023] Open
Abstract
The human brain represents one of the most complex biological systems, containing billions of neurons interconnected through trillions of synapses. Inherent to the brain is a biochemical complexity involving ions, signaling molecules, and peptides that regulate neuronal activity and allow for short- and long-term adaptations. Large-scale and noninvasive imaging techniques, such as fMRI and EEG, have highlighted brain regions involved in specific functions and visualized connections between different brain areas. A major shortcoming, however, is the need for more information on specific cell types and neurotransmitters involved, as well as poor spatial and temporal resolution. Recent technologies have been advanced for neuronal circuit mapping and implemented in behaving model organisms to address this. Here, we highlight strategies for targeting specific neuronal subtypes, identifying, and releasing signaling molecules, controlling gene expression, and monitoring neuronal circuits in real-time in vivo Combined, these approaches allow us to establish direct causal links from genes and molecules to the systems level and ultimately to cognitive processes.
Collapse
Affiliation(s)
| | - Denise Cai
- Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | | | | | | | - Zhenpeng Qin
- University of Texas-Dallas, Richardson, TX 75080
| | - Lin Tian
- University of California-Davis, Davis, CA 95616
| | | |
Collapse
|
38
|
Nakasone Y, Murakami H, Tokonami S, Oda T, Terazima M. Time-resolved study on signaling pathway of photoactivated adenylate cyclase and its nonlinear optical response. J Biol Chem 2023; 299:105285. [PMID: 37742920 PMCID: PMC10634658 DOI: 10.1016/j.jbc.2023.105285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Photoactivated adenylate cyclases (PACs) are multidomain BLUF proteins that regulate the cellular levels of cAMP in a light-dependent manner. The signaling route and dynamics of PAC from Oscillatoria acuminata (OaPAC), which consists of a light sensor BLUF domain, an adenylate cyclase domain, and a connector helix (α3-helix), were studied by detecting conformational changes in the protein moiety. Although circular dichroism and small-angle X-ray scattering measurements did not show significant changes upon light illumination, the transient grating method successfully detected light-induced changes in the diffusion coefficient (diffusion-sensitive conformational change (DSCC)) of full-length OaPAC and the BLUF domain with the α3-helix. DSCC of full-length OaPAC was observed only when both protomers in a dimer were photoconverted. This light intensity dependence suggests that OaPAC is a cyclase with a nonlinear light intensity response. The enzymatic activity indeed nonlinearly depends on light intensity, that is, OaPAC is activated under strong light conditions. It was also found that both DSCC and enzymatic activity were suppressed by a mutation in the W90 residue, indicating the importance of the highly conserved Trp in many BLUF domains for the function. Based on these findings, a reaction scheme was proposed together with the reaction dynamics.
Collapse
Affiliation(s)
- Yusuke Nakasone
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Hiroto Murakami
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Shunrou Tokonami
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Takashi Oda
- Department of Life Science and Research Center for Life Science, College of Science, Rikkyo University, Tokyo, Japan
| | - Masahide Terazima
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, Japan.
| |
Collapse
|
39
|
Marcus DJ, Bruchas MR. Optical Approaches for Investigating Neuromodulation and G Protein-Coupled Receptor Signaling. Pharmacol Rev 2023; 75:1119-1139. [PMID: 37429736 PMCID: PMC10595021 DOI: 10.1124/pharmrev.122.000584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/06/2023] [Accepted: 05/01/2023] [Indexed: 07/12/2023] Open
Abstract
Despite the fact that roughly 40% of all US Food and Drug Administration (FDA)-approved pharmacological therapeutics target G protein-coupled receptors (GPCRs), there remains a gap in our understanding of the physiologic and functional role of these receptors at the systems level. Although heterologous expression systems and in vitro assays have revealed a tremendous amount about GPCR signaling cascades, how these cascades interact across cell types, tissues, and organ systems remains obscure. Classic behavioral pharmacology experiments lack both the temporal and spatial resolution to resolve these long-standing issues. Over the past half century, there has been a concerted effort toward the development of optical tools for understanding GPCR signaling. From initial ligand uncaging approaches to more recent development of optogenetic techniques, these strategies have allowed researchers to probe longstanding questions in GPCR pharmacology both in vivo and in vitro. These tools have been employed across biologic systems and have allowed for interrogation of everything from specific intramolecular events to pharmacology at the systems level in a spatiotemporally specific manner. In this review, we present a historical perspective on the motivation behind and development of a variety of optical toolkits that have been generated to probe GPCR signaling. Here we highlight how these tools have been used in vivo to uncover the functional role of distinct populations of GPCRs and their signaling cascades at a systems level. SIGNIFICANCE STATEMENT: G protein-coupled receptors (GPCRs) remain one of the most targeted classes of proteins for pharmaceutical intervention, yet we still have a limited understanding of how their unique signaling cascades effect physiology and behavior at the systems level. In this review, we discuss a vast array of optical techniques that have been devised to probe GPCR signaling both in vitro and in vivo.
Collapse
Affiliation(s)
- David J Marcus
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain and Emotion (D.J.M., M.R.B.), Department of Anesthesiology and Pain Medicine (D.J.M., M.R.B.), Department of Pharmacology (M.R.B.), and Department of Bioengineering (M.R.B.), University of Washington, Seattle, Washington
| |
Collapse
|
40
|
Idstein V, Ehret AK, Yousefi OS, Schamel WW. Engineering of an Optogenetic T Cell Receptor Compatible with Fluorescence-Based Readouts. ACS Synth Biol 2023; 12:2857-2864. [PMID: 37781987 DOI: 10.1021/acssynbio.3c00429] [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: 10/03/2023]
Abstract
Optogenetics offers a set of tools for the precise manipulation of signaling pathways. Here we exploit optogenetics to experimentally change the kinetics of protein-protein interactions on demand. We had developed a system in which the interaction of a modified T cell receptor (TCR) with an engineered ligand can be controlled by light. The ligand was the plant photoreceptor phytochrome B (PhyB) and the TCR included a TCRβ chain fused to GFP and a mutated PhyB-interacting factor (PIFS), resulting in the GFP-PIFS-TCR. We failed to engineer a nonfluorescent PIFS-fused TCR, since PIFS did not bind to PhyB when omitting GFP. Here we tested nine different versions of PIFS-fused TCRs. We found that the SNAP-PIFS-TCR was expressed well on the surface, bound to PhyB, and subsequently elicited activation signals. This receptor could be combined with a GFP reporter system in which the expression of GFP is driven by the transcription factor NF-AT.
Collapse
Affiliation(s)
- Vincent Idstein
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Centre Freiburg, and Faculty of Medicine, University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstr. 19a, 79104 Freiburg, Germany
| | - Anna K Ehret
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Centre Freiburg, and Faculty of Medicine, University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstr. 19a, 79104 Freiburg, Germany
| | - O Sascha Yousefi
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
| | - Wolfgang W Schamel
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Schänzlestr. 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Centre Freiburg, and Faculty of Medicine, University of Freiburg, Breisacherstr. 115, 79106 Freiburg, Germany
| |
Collapse
|
41
|
Aslam M, Ladilov Y. Editorial: Advances in cAMP signaling research: basic and translational aspects. Front Physiol 2023; 14:1266718. [PMID: 37727656 PMCID: PMC10505720 DOI: 10.3389/fphys.2023.1266718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/21/2023] Open
Affiliation(s)
- Muhammad Aslam
- Experimental Cardiology, Department of Internal Medicine I, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Clinic GmbH, Bad Nauheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Rhein-Main, Bad Nauheim, Germany
| | - Yury Ladilov
- Department of Cardiovascular Surgery, Heart Center Brandenburg, Brandenburg Medical School Theodor Fontane, Bernau bei Berlin, Germany
| |
Collapse
|
42
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Shimizu T, Hososhima S, Tsunoda SP, Kandori H, Hibi M. Optogenetic manipulation of neuronal and cardiomyocyte functions in zebrafish using microbial rhodopsins and adenylyl cyclases. eLife 2023; 12:e83975. [PMID: 37589546 PMCID: PMC10435232 DOI: 10.7554/elife.83975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
Even though microbial photosensitive proteins have been used for optogenetics, their use should be optimized to precisely control cell and tissue functions in vivo. We exploited GtCCR4 and KnChR, cation channelrhodopsins from algae, BeGC1, a guanylyl cyclase rhodopsin from a fungus, and photoactivated adenylyl cyclases (PACs) from cyanobacteria (OaPAC) or bacteria (bPAC), to control cell functions in zebrafish. Optical activation of GtCCR4 and KnChR in the hindbrain reticulospinal V2a neurons, which are involved in locomotion, induced swimming behavior at relatively short latencies, whereas activation of BeGC1 or PACs achieved it at long latencies. Activation of GtCCR4 and KnChR in cardiomyocytes induced cardiac arrest, whereas activation of bPAC gradually induced bradycardia. KnChR activation led to an increase in intracellular Ca2+ in the heart, suggesting that depolarization caused cardiac arrest. These data suggest that these optogenetic tools can be used to reveal the function and regulation of zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Takashi Shimizu
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shoko Hososhima
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Satoshi P Tsunoda
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| |
Collapse
|
43
|
Derderian C, Canales GI, Reiter JF. Seriously cilia: A tiny organelle illuminates evolution, disease, and intercellular communication. Dev Cell 2023; 58:1333-1349. [PMID: 37490910 PMCID: PMC10880727 DOI: 10.1016/j.devcel.2023.06.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/18/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
The borders between cell and developmental biology, which have always been permeable, have largely dissolved. One manifestation is the blossoming of cilia biology, with cell and developmental approaches (increasingly complemented by human genetics, structural insights, and computational analysis) fruitfully advancing understanding of this fascinating, multifunctional organelle. The last eukaryotic common ancestor probably possessed a motile cilium, providing evolution with ample opportunity to adapt cilia to many jobs. Over the last decades, we have learned how non-motile, primary cilia play important roles in intercellular communication. Reflecting their diverse motility and signaling functions, compromised cilia cause a diverse range of diseases collectively called "ciliopathies." In this review, we highlight how cilia signal, focusing on how second messengers generated in cilia convey distinct information; how cilia are a potential source of signals to other cells; how evolution may have shaped ciliary function; and how cilia research may address thorny outstanding questions.
Collapse
Affiliation(s)
- Camille Derderian
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriela I Canales
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
44
|
Baudet S, Zagar Y, Roche F, Gomez-Bravo C, Couvet S, Bécret J, Belle M, Vougny J, Uthayasuthan S, Ros O, Nicol X. Subcellular second messenger networks drive distinct repellent-induced axon behaviors. Nat Commun 2023; 14:3809. [PMID: 37369692 PMCID: PMC10300027 DOI: 10.1038/s41467-023-39516-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Second messengers, including cAMP, cGMP and Ca2+ are often placed in an integrating position to combine the extracellular cues that orient growing axons in the developing brain. This view suggests that axon repellents share the same set of cellular messenger signals and that axon attractants evoke opposite cAMP, cGMP and Ca2+ changes. Investigating the confinement of these second messengers in cellular nanodomains, we instead demonstrate that two repellent cues, ephrin-A5 and Slit1, induce spatially segregated signals. These guidance molecules activate subcellular-specific second messenger crosstalk, each signaling network controlling distinct axonal morphology changes in vitro and pathfinding decisions in vivo.
Collapse
Affiliation(s)
- Sarah Baudet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Fiona Roche
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Claudia Gomez-Bravo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Sandrine Couvet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Johann Bécret
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Morgane Belle
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Juliette Vougny
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | | | - Oriol Ros
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028, Barcelona, Catalonia, Spain
| | - Xavier Nicol
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France.
| |
Collapse
|
45
|
Ramirez-Garcia PD, Veldhuis NA, Bunnett NW, Davis TP. Targeting endosomal receptors, a new direction for polymers in nanomedicine. J Mater Chem B 2023; 11:5390-5399. [PMID: 37219363 PMCID: PMC10641892 DOI: 10.1039/d3tb00156c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In this perspective, we outline a new opportunity for exploiting nanoparticle delivery of antagonists to target G-protein coupled receptors localized in intracellular compartments. We discuss the specific example of antagonizing endosomal receptors involved in pain to develop long-lasting analgesics but also outline the broader application potential of this delivery approach. We discuss the materials used to target endosomal receptors and indicate the design requirements for future successful applications.
Collapse
Affiliation(s)
- Paulina D Ramirez-Garcia
- Dentistry Translational Research Center, New York University College of Dentistry, New York, 10010, USA.
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone School of Medicine, New York, NY 10010, USA
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
46
|
Konrad KR, Gao S, Zurbriggen MD, Nagel G. Optogenetic Methods in Plant Biology. ANNUAL REVIEW OF PLANT BIOLOGY 2023; 74:313-339. [PMID: 37216203 DOI: 10.1146/annurev-arplant-071122-094840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Optogenetics is a technique employing natural or genetically engineered photoreceptors in transgene organisms to manipulate biological activities with light. Light can be turned on or off, and adjusting its intensity and duration allows optogenetic fine-tuning of cellular processes in a noninvasive and spatiotemporally resolved manner. Since the introduction of Channelrhodopsin-2 and phytochrome-based switches nearly 20 years ago, optogenetic tools have been applied in a variety of model organisms with enormous success, but rarely in plants. For a long time, the dependence of plant growth on light and the absence of retinal, the rhodopsin chromophore, prevented the establishment of plant optogenetics until recent progress overcame these difficulties. We summarize the recent results of work in the field to control plant growth and cellular motion via green light-gated ion channels and present successful applications to light-control gene expression with single or combined photoswitches in plants. Furthermore, we highlight the technical requirements and options for future plant optogenetic research.
Collapse
Affiliation(s)
- Kai R Konrad
- Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute for Biosciences, Biocenter, University of Würzburg, Würzburg, Germany;
| | - Shiqiang Gao
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, Würzburg, Germany; ,
| | - Matias D Zurbriggen
- Institute of Synthetic Biology and CEPLAS, University of Düsseldorf, Düsseldorf, Germany;
| | - Georg Nagel
- Department of Neurophysiology, Institute of Physiology, Biocenter, University of Würzburg, Würzburg, Germany; ,
| |
Collapse
|
47
|
Paolocci E, Zaccolo M. Compartmentalised cAMP signalling in the primary cilium. Front Physiol 2023; 14:1187134. [PMID: 37256063 PMCID: PMC10226274 DOI: 10.3389/fphys.2023.1187134] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
cAMP is a universal second messenger that relies on precise spatio-temporal regulation to control varied, and often opposing, cellular functions. This is achieved via selective activation of effectors embedded in multiprotein complexes, or signalosomes, that reside at distinct subcellular locations. cAMP is also one of many pathways known to operate within the primary cilium. Dysfunction of ciliary signaling leads to a class of diseases known as ciliopathies. In Autosomal Dominant Polycystic Kidney Disease (ADPKD), a ciliopathy characterized by the formation of fluid-filled kidney cysts, upregulation of cAMP signaling is known to drive cystogenesis. For decades it has been debated whether the primary cilium is an independent cAMP sub-compartment, or whether it shares a diffusible pool of cAMP with the cell body. Recent studies now suggest it is a specific pool of cAMP generated in the cilium that propels cyst formation in ADPKD, supporting the notion that this antenna-like organelle is a compartment within which cAMP signaling occurs independently from cAMP signaling in the bulk cytosol. Here we present examples of cAMP function in the cilium which suggest this mysterious organelle is home to more than one cAMP signalosome. We review evidence that ciliary membrane localization of G-Protein Coupled Receptors (GPCRs) determines their downstream function and discuss how optogenetic tools have contributed to establish that cAMP generated in the primary cilium can drive cystogenesis.
Collapse
|
48
|
Choi J, Shin E, Lee J, Devarasou S, Kim D, Shin JH, Choi JH, Heo WD, Han YM. Light-stimulated insulin secretion from pancreatic islet-like organoids derived from human pluripotent stem cells. Mol Ther 2023; 31:1480-1495. [PMID: 36932674 PMCID: PMC10188912 DOI: 10.1016/j.ymthe.2023.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/06/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Optogenetic techniques permit non-invasive, spatiotemporal, and reversible modulation of cellular activities. Here, we report a novel optogenetic regulatory system for insulin secretion in human pluripotent stem cell (hPSC)-derived pancreatic islet-like organoids using monSTIM1 (monster-opto-Stromal interaction molecule 1), an ultra-light-sensitive OptoSTIM1 variant. The monSTIM1 transgene was incorporated at the AAVS1 locus in human embryonic stem cells (hESCs) by CRISPR-Cas9-mediated genome editing. Not only were we able to elicit light-induced intracellular Ca2+ concentration ([Ca2+]i) transients from the resulting homozygous monSTIM1+/+-hESCs, but we also successfully differentiated them into pancreatic islet-like organoids (PIOs). Upon light stimulation, the β-cells in these monSTIM1+/+-PIOs displayed reversible and reproducible [Ca2+]i transient dynamics. Furthermore, in response to photoexcitation, they secreted human insulin. Light-responsive insulin secretion was similarly observed in monSTIM1+/+-PIOs produced from neonatal diabetes (ND) patient-derived induced pluripotent stem cells (iPSCs). Under LED illumination, monSTIM1+/+-PIO-transplanted diabetic mice produced human c-peptide. Collectively, we developed a cellular model for the optogenetic control of insulin secretion using hPSCs, with the potential to be applied to the amelioration of hyperglycemic disorders.
Collapse
Affiliation(s)
- Jieun Choi
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Eunji Shin
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Jinsu Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | | | - Dongkyu Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Jin-Ho Choi
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea.
| | - Yong-Mahn Han
- Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
49
|
Doucette CC, Nguyen DC, Barteselli D, Blanchard S, Pelletier M, Kesharwani D, Jachimowicz E, Su S, Karolak M, Brown AC. Optogenetic activation of UCP1-dependent thermogenesis in brown adipocytes. iScience 2023; 26:106560. [PMID: 37123235 PMCID: PMC10139976 DOI: 10.1016/j.isci.2023.106560] [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: 10/13/2022] [Revised: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Brown adipocytes are unique in that they expend energy and produce heat to maintain euthermia through expression of uncoupling protein-1 (UCP1). Given their propensity to stimulate weight loss and promote resistance to obesity, they are a compelling interventional target for obesity-related disorders. Here, we tested whether an optogenetic approach could be used to activate UCP1-dependent thermogenesis in brown adipocytes. We generated brown adipocytes expressing a bacterial-derived photoactivatable adenylyl cyclase (bPAC) that, upon blue light stimulation, increases UCP1 expression, fuel uptake and thermogenesis. This unique system allows for precise, chemical free, temporal control of UCP1-dependent thermogenesis, which can aid in our understanding of brown adipocyte biology and development of therapies that target obesity-related disorders.
Collapse
Affiliation(s)
- Chad C. Doucette
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Daniel C. Nguyen
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Davide Barteselli
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Sophia Blanchard
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Masen Pelletier
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Devesh Kesharwani
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Ed Jachimowicz
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Su Su
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Michele Karolak
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
| | - Aaron C. Brown
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
- School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
- Tufts University School of Medicine, 145 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
50
|
Harada K, Takashima M, Kitaguchi T, Tsuboi T. F-actin determines the time-dependent shift in docking dynamics of glucagon-like peptide-1 granules upon stimulation of secretion. FEBS Lett 2023; 597:657-671. [PMID: 36694275 DOI: 10.1002/1873-3468.14580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
Although exocytosis can be categorized into several forms based on docking dynamics, temporal regulatory mechanisms of the exocytotic forms are unclear. We explored the dynamics of glucagon-like peptide-1 (GLP-1) exocytosis in murine GLUTag cells (GLP-1-secreting enteroendocrine L-cells) upon stimulation with deoxycholic acid (DCA) or high K+ to elucidate the mechanisms regulating the balance between the different types of exocytotic forms (pre-docked with the plasma membrane before stimulation; docked after stimulation and subsequently fused; or rapidly recruited and fused after stimulation, without stable docking). GLP-1 exocytosis showed a biphasic pattern, and we found that most exocytosis was from the pre-docked granules with the plasma membrane before stimulation, or granules rapidly fused to the plasma membrane without docking after stimulation. In contrast, granules docked with the plasma membrane after stimuli and eventually fused were predominant thereafter. Inhibition of actin polymerization suppressed exocytosis of the pre-docked granules. These results suggest that the docking dynamics of GLP-1 granules shows a time-dependent biphasic shift, which is determined by interaction with F-actin.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| | - Maoko Takashima
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| | - Tetsuya Kitaguchi
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Japan
| |
Collapse
|