1
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
2
|
Gerrard EJ, Tichy AM, Janovjak H. Automated Plate Reader-Based Assays of Light-Activated GPCRs. Methods Mol Biol 2025; 2840:217-229. [PMID: 39724355 DOI: 10.1007/978-1-0716-4047-0_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
In the emerging field of optogenetics, light-sensitive G-protein coupled receptors (GPCRs) allow for the temporally precise control of canonical cell signaling pathways. Expressing, stimulating, and measuring the activity of light-sensitive GPCRs (e.g., opsins or chimeric OptoXRs) in mammalian cells is a nontrivial task as many standard assay practices are not compatible with light-sensitive molecular tools. In this chapter, we present a method for quantifying opsin activity in automated plate reader-based assays without the need for additional optical hardware (i.e., light sources). The protocol is applied to assess cAMP levels downstream of a chimeric OptoXR but can be expanded to other opsins and second messengers, such as Ca2+ mobilization. We describe how the internal optical components in commonly available plate readers can be utilized to both activate and detect kinetic and dose-response relationships, as well as provide general guidance for optimizing assays with light-sensitive molecular tools.
Collapse
Affiliation(s)
- Elliot J Gerrard
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton, VIC, Australia
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton, VIC, Australia
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton, VIC, Australia
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
3
|
Zhang SX, Kim A, Madara JC, Zhu PK, Christenson LF, Lutas A, Kalugin PN, Sunkavalli PS, Jin Y, Pal A, Tian L, Lowell BB, Andermann ML. Stochastic neuropeptide signals compete to calibrate the rate of satiation. Nature 2025; 637:137-144. [PMID: 39506113 PMCID: PMC11981016 DOI: 10.1038/s41586-024-08164-8] [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: 07/19/2023] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Neuropeptides have important roles in neural plasticity, spiking and behaviour1. Yet, many fundamental questions remain regarding their spatiotemporal transmission, integration and functions in the awake brain. Here we examined how MC4R-expressing neurons in the paraventricular nucleus of the hypothalamus (PVHMC4R) integrate neuropeptide signals to modulate feeding-related fast synaptic transmission and titrate the transition to satiety2-6. We show that hunger-promoting AgRP axons release the neuropeptide NPY to decrease the second messenger cAMP in PVHMC4R neurons, while satiety-promoting POMC axons release the neuropeptide αMSH to increase cAMP. Each release event is all-or-none, stochastic and can impact multiple neurons within an approximately 100-µm-diameter region. After release, NPY and αMSH peptides compete to control cAMP-the amplitude and persistence of NPY signalling is blunted by high αMSH in the fed state, while αMSH signalling is blunted by high NPY in the fasted state. Feeding resolves this competition by simultaneously elevating αMSH release and suppressing NPY release7,8, thereby sustaining elevated cAMP in PVHMC4R neurons throughout a meal. In turn, elevated cAMP facilitates potentiation of feeding-related excitatory inputs with each bite to gradually promote satiation across many minutes. Our findings highlight biochemical modes of peptide signal integration and information accumulation to guide behavioural state transitions.
Collapse
Affiliation(s)
- Stephen X Zhang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Angela Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joseph C Madara
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Paula K Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lauren F Christenson
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrew Lutas
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter N Kalugin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Praneel S Sunkavalli
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yihan Jin
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Neuroscience Graduate Group, Center for Neuroscience, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Akash Pal
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- Max Planck Florida Institute for Neuroscience, One Max Planck Way, Jupiter, FL, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mark L Andermann
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
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
|
5
|
Alfihed S, Majrashi M, Ansary M, Alshamrani N, Albrahim SH, Alsolami A, Alamari HA, Zaman A, Almutairi D, Kurdi A, Alzaydi MM, Tabbakh T, Al-Otaibi F. Non-Invasive Brain Sensing Technologies for Modulation of Neurological Disorders. BIOSENSORS 2024; 14:335. [PMID: 39056611 PMCID: PMC11274405 DOI: 10.3390/bios14070335] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
The non-invasive brain sensing modulation technology field is experiencing rapid development, with new techniques constantly emerging. This study delves into the field of non-invasive brain neuromodulation, a safer and potentially effective approach for treating a spectrum of neurological and psychiatric disorders. Unlike traditional deep brain stimulation (DBS) surgery, non-invasive techniques employ ultrasound, electrical currents, and electromagnetic field stimulation to stimulate the brain from outside the skull, thereby eliminating surgery risks and enhancing patient comfort. This study explores the mechanisms of various modalities, including transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS), highlighting their potential to address chronic pain, anxiety, Parkinson's disease, and depression. We also probe into the concept of closed-loop neuromodulation, which personalizes stimulation based on real-time brain activity. While we acknowledge the limitations of current technologies, our study concludes by proposing future research avenues to advance this rapidly evolving field with its immense potential to revolutionize neurological and psychiatric care and lay the foundation for the continuing advancement of innovative non-invasive brain sensing technologies.
Collapse
Affiliation(s)
- Salman Alfihed
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Majed Majrashi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Muhammad Ansary
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Naif Alshamrani
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Shahad H. Albrahim
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Abdulrahman Alsolami
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Hala A. Alamari
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Adnan Zaman
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Dhaifallah Almutairi
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Abdulaziz Kurdi
- Advanced Materials Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Mai M. Alzaydi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Thamer Tabbakh
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Faisal Al-Otaibi
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
6
|
Lee J, Jeong Y, Park S, Kim S, Oh H, Jin JA, Sohn JW, Kim D, Shin HS, Do Heo W. Phospholipase C beta 1 in the dentate gyrus gates fear memory formation through regulation of neuronal excitability. SCIENCE ADVANCES 2024; 10:eadj4433. [PMID: 38959322 PMCID: PMC11221510 DOI: 10.1126/sciadv.adj4433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/28/2024] [Indexed: 07/05/2024]
Abstract
Memory processes rely on a molecular signaling system that balances the interplay between positive and negative modulators. Recent research has focused on identifying memory-regulating genes and their mechanisms. Phospholipase C beta 1 (PLCβ1), highly expressed in the hippocampus, reportedly serves as a convergence point for signal transduction through G protein-coupled receptors. However, the detailed role of PLCβ1 in memory function has not been elucidated. Here, we demonstrate that PLCβ1 in the dentate gyrus functions as a memory suppressor. We reveal that mice lacking PLCβ1 in the dentate gyrus exhibit a heightened fear response and impaired memory extinction, and this excessive fear response is repressed by upregulation of PLCβ1 through its overexpression or activation using a newly developed optogenetic system. Last, our results demonstrate that PLCβ1 overexpression partially inhibits exaggerated fear response caused by traumatic experience. Together, PLCβ1 is crucial in regulating contextual fear memory formation and potentially enhancing the resilience to trauma-related conditions.
Collapse
Affiliation(s)
- Jinsu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Yeonji Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seahyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Hyunsik Oh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Ju-Ae Jin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon 34141, Korea
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- Department of Brain and Cognitive Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
- KAIST Institute for the BioCentury (KIB), Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
7
|
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
|
8
|
Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The Impact of Neurotransmitters on the Neurobiology of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:15340. [PMID: 37895020 PMCID: PMC10607327 DOI: 10.3390/ijms242015340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide. Neurodegenerative diseases result from progressive damage to nerve cells in the brain or peripheral nervous system connections that are essential for cognition, coordination, strength, sensation, and mobility. Dysfunction of these brain and nerve functions is associated with Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and motor neuron disease. In addition to these, 50% of people living with HIV develop a spectrum of cognitive, motor, and/or mood problems collectively referred to as HIV-Associated Neurocognitive Disorders (HAND) despite the widespread use of a combination of antiretroviral therapies. Neuroinflammation and neurotransmitter systems have a pathological correlation and play a critical role in developing neurodegenerative diseases. Each of these diseases has a unique pattern of dysregulation of the neurotransmitter system, which has been attributed to different forms of cell-specific neuronal loss. In this review, we will focus on a discussion of the regulation of dopaminergic and cholinergic systems, which are more commonly disturbed in neurodegenerative disorders. Additionally, we will provide evidence for the hypothesis that disturbances in neurotransmission contribute to the neuronal loss observed in neurodegenerative disorders. Further, we will highlight the critical role of dopamine as a mediator of neuronal injury and loss in the context of NeuroHIV. This review will highlight the need to further investigate neurotransmission systems for their role in the etiology of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (S.E.D.); (A.B.C.); (A.C.J.-T.)
| |
Collapse
|
9
|
Mitroshina E, Kalinina E, Vedunova M. Optogenetics in Alzheimer's Disease: Focus on Astrocytes. Antioxidants (Basel) 2023; 12:1856. [PMID: 37891935 PMCID: PMC10604138 DOI: 10.3390/antiox12101856] [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: 09/04/2023] [Revised: 09/27/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, resulting in disability and mortality. The global incidence of AD is consistently surging. Although numerous therapeutic agents with promising potential have been developed, none have successfully treated AD to date. Consequently, the pursuit of novel methodologies to address neurodegenerative processes in AD remains a paramount endeavor. A particularly promising avenue in this search is optogenetics, enabling the manipulation of neuronal activity. In recent years, research attention has pivoted from neurons to glial cells. This review aims to consider the potential of the optogenetic correction of astrocyte metabolism as a promising strategy for correcting AD-related disorders. The initial segment of the review centers on the role of astrocytes in the genesis of neurodegeneration. Astrocytes have been implicated in several pathological processes associated with AD, encompassing the clearance of β-amyloid, neuroinflammation, excitotoxicity, oxidative stress, and lipid metabolism (along with a critical role in apolipoprotein E function). The effect of astrocyte-neuronal interactions will also be scrutinized. Furthermore, the review delves into a number of studies indicating that changes in cellular calcium (Ca2+) signaling are one of the causes of neurodegeneration. The review's latter section presents insights into the application of various optogenetic tools to manipulate astrocytic function as a means to counteract neurodegenerative changes.
Collapse
Affiliation(s)
- Elena Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia (M.V.)
| | | | | |
Collapse
|
10
|
Park B, Bang S, Hwang KS, Cha YK, Kwak J, Tran NL, Kim HS, Park S, Oh SJ, Im M, Chung S, Kim J, Park TH, Song HS, Kim HN, Kim JH. Eye-Mimicked Neural Network Composed of Photosensitive Neural Spheroids with Human Opsin Proteins. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302996. [PMID: 37377148 DOI: 10.1002/adma.202302996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023]
Abstract
An in vitro model, composed of the short-wavelength human opsins and rhodopsins, is created. Two types of photosensitive neural spheroids are transfected for selective reaction under bluish-purple and green lights. These are employed to two devices with intact neuron and neural-spheroid to study the interaction. By photostimulation, the photosensitive spheroid initiated photoactivation, and the signal generated from its body is transmitted to adjacent neural networks. Specifically, the signal traveled through the axon bundle in narrow gap from photosensitive spheroid to intact spheroid as an eye-to-brain model including optic nerve. The whole process with photosensitive spheroid is monitored by calcium ion detecting fluorescence images. The results of this study can be applied to examine vision restoration and novel photosensitive biological systems with spectral sensitivity.
Collapse
Affiliation(s)
- Byeongho Park
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Department of Biomedical Engineering, Dongguk University, Goyang, 10326, Republic of Korea
| | - Kyeong Seob Hwang
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yeon Kyung Cha
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jisung Kwak
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Na Ly Tran
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyo-Suk Kim
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Subeen Park
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seung Ja Oh
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Maesoon Im
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sooyoung Chung
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Tai Hyun Park
- Department of Nutritional Science and Food Management, Ewha Womans University, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Hyun Seok Song
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hong Nam Kim
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Hun Kim
- Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| |
Collapse
|
11
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Sugihara T, Shimizu T, Koyanagi M, Terakita A, Hibi M. Optogenetic manipulation of Gq- and Gi/o-coupled receptor signaling in neurons and heart muscle cells. eLife 2023; 12:e83974. [PMID: 37589544 PMCID: PMC10435233 DOI: 10.7554/elife.83974] [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: 10/05/2022] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) transmit signals into cells depending on the G protein type. To analyze the functions of GPCR signaling, we assessed the effectiveness of animal G-protein-coupled bistable rhodopsins that can be controlled into active and inactive states by light application using zebrafish. We expressed Gq- and Gi/o-coupled bistable rhodopsins in hindbrain reticulospinal V2a neurons, which are involved in locomotion, or in cardiomyocytes. Light stimulation of the reticulospinal V2a neurons expressing Gq-coupled spider Rh1 resulted in an increase in the intracellular Ca2+ level and evoked swimming behavior. Light stimulation of cardiomyocytes expressing the Gi/o-coupled mosquito Opn3, pufferfish TMT opsin, or lamprey parapinopsin induced cardiac arrest, and the effect was suppressed by treatment with pertussis toxin or barium, suggesting that Gi/o-dependent regulation of inward-rectifier K+ channels controls cardiac function. These data indicate that these rhodopsins are useful for optogenetic control of GPCR-mediated signaling in zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya UniversityNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | | | - Akihisa Terakita
- Graduate School of Science, Osaka Metropolitan UniversityOsakaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| |
Collapse
|
12
|
Tsukamoto H, Kubo Y. A self-inactivating invertebrate opsin optically drives biased signaling toward Gβγ-dependent ion channel modulation. Proc Natl Acad Sci U S A 2023; 120:e2301269120. [PMID: 37186850 PMCID: PMC10214182 DOI: 10.1073/pnas.2301269120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Animal opsins, light-sensitive G protein-coupled receptors, have been used for optogenetic tools to control G protein-dependent signaling pathways. Upon G protein activation, the Gα and Gβγ subunits drive different intracellular signaling pathways, leading to complex cellular responses. For some purposes, Gα- and Gβγ-dependent signaling needs to be separately modulated, but these responses are simultaneously evoked due to the 1:1 stoichiometry of Gα and Gβγ Nevertheless, we show temporal activation of G protein using a self-inactivating invertebrate opsin, Platynereis c-opsin1, drives biased signaling for Gβγ-dependent GIRK channel activation in a light-dependent manner by utilizing the kinetic difference between Gβγ-dependent and Gα-dependent responses. The opsin-induced transient Gi/o activation preferentially causes activation of the kinetically fast Gβγ-dependent GIRK channels rather than slower Gi/oα-dependent adenylyl cyclase inhibition. Although similar Gβγ-biased signaling properties were observed in a self-inactivating vertebrate visual pigment, Platynereis c-opsin1 requires fewer retinal molecules to evoke cellular responses. Furthermore, the Gβγ-biased signaling properties of Platynereis c-opsin1 are enhanced by genetically fusing with RGS8 protein, which accelerates G protein inactivation. The self-inactivating invertebrate opsin and its RGS8-fusion protein can function as optical control tools biased for Gβγ-dependent ion channel modulation.
Collapse
Affiliation(s)
- Hisao Tsukamoto
- Department of Biology, Kobe University, Kobe657-8501, Japan
- Department of Life and Coordination-Complex Molecular Science, Institute for Molecular Science, Okazaki444-8585, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi 332-0012, Japan
| | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular Physiology, National Institute for Physiological Sciences, Okazaki444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama240-0193, Japan
| |
Collapse
|
13
|
Zhou G, Wan WW, Wang W. Modular Peroxidase-Based Reporters for Detecting Protease Activity and Protein Interactions with Temporal Gating. J Am Chem Soc 2022; 144:22933-22940. [PMID: 36511757 PMCID: PMC10026560 DOI: 10.1021/jacs.2c08280] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Enzymatic reporters have been widely applied to study various biological processes because they can amplify signal through enzymatic reactions and provide good sensitivity. However, there is still a need for modular motifs for designing a series of enzymatic reporters. Here, we report a modular peroxidase-based motif, named CLAPon, that features acid-base coil-caged enhanced ascorbate peroxidase (APEX). We demonstrate the modularity of CLAPon by designing a series of reporters for detecting protease activity and protein-protein interactions (PPIs). CLAPon for protease activity showed a 390-fold fluorescent signal increase upon tobacco etch virus protease cleavage. CLAPon for PPI detection (PPI-CLAPon) has two variants, PPI-CLAPon1.0 and 1.1. PPI-CLAPon1.0 showed a signal-to-noise ratio (SNR) of up to 107 for high-affinity PPI pairs and enabled imaging with sub-cellular spatial resolution. However, the more sensitive PPI-CLAPon1.1 is required for detecting low-affinity PPI pairs. PPI-CLAPon1.0 was further engineered to a reporter with light-dependent temporal gating, called LiPPI-CLAPon1.0, which can detect a 3-min calcium-dependent PPI with an SNR of 17. LiPPI-CLAPon enables PPI detection within a specific time window with rapid APEX activation and diverse readout. Lastly, PPI-CLAPon1.0 was designed to have chemical gating, providing more versatility to complement the LiPPI-CLAPon. These CLAPon-based reporter designs can be broadly applied to study various signaling processes that involve protease activity and PPIs and provide a versatile platform to design various genetically encoded reporters.
Collapse
Affiliation(s)
- Guanwei Zhou
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wei Wei Wan
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wenjing Wang
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Corresponding Author: Wenjing Wang,
| |
Collapse
|
14
|
Surdin T, Preissing B, Rohr L, Grömmke M, Böke H, Barcik M, Azimi Z, Jancke D, Herlitze S, Mark MD, Siveke I. Optogenetic activation of mGluR1 signaling in the cerebellum induces synaptic plasticity. iScience 2022; 26:105828. [PMID: 36632066 PMCID: PMC9826949 DOI: 10.1016/j.isci.2022.105828] [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: 04/12/2022] [Revised: 10/21/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Neuronal plasticity underlying cerebellar learning behavior is strongly associated with type 1 metabotropic glutamate receptor (mGluR1) signaling. Activation of mGluR1 leads to activation of the Gq/11 pathway, which is involved in inducing synaptic plasticity at the parallel fiber-Purkinje cell synapse (PF-PC) in form of long-term depression (LTD). To optogenetically modulate mGluR1 signaling we fused mouse melanopsin (OPN4) that activates the Gq/11 pathway to the C-termini of mGluR1 splice variants (OPN4-mGluR1a and OPN4-mGluR1b). Activation of both OPN4-mGluR1 variants showed robust Ca2+ increase in HEK cells and PCs of cerebellar slices. We provide the prove-of-concept approach to modulate synaptic plasticity via optogenetic activation of OPN4-mGluR1a inducing LTD at the PF-PC synapse in vitro. Moreover, we demonstrate that light activation of mGluR1a signaling pathway by OPN4-mGluR1a in PCs leads to an increase in intrinsic activity of PCs in vivo and improved cerebellum driven learning behavior.
Collapse
Affiliation(s)
- Tatjana Surdin
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Bianca Preissing
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Lennard Rohr
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Michelle Grömmke
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Hanna Böke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Maike Barcik
- Cardiovascular Research Institute Düsseldorf, Division of Cardiology, Pulmonology, and Vascular Medicine, University Duesseldorf, Medical Faculty, Duesseldorf, Germany
| | - Zohre Azimi
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Dirk Jancke
- Optical Imaging Group, Institut für Neuroinformatik, Ruhr-University Bochum, Bochum, Germany
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Corresponding author
| | - Melanie D. Mark
- Behavioral Neuroscience, Ruhr-University Bochum, Bochum, Germany
| | - Ida Siveke
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany,Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany,Corresponding author
| |
Collapse
|
15
|
A photocaged orexin-B for spatiotemporally precise control of orexin signaling. Cell Chem Biol 2022; 29:1729-1738.e8. [PMID: 36481097 PMCID: PMC9794195 DOI: 10.1016/j.chembiol.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/04/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Orexin neuropeptides carry out important neuromodulatory functions in the brain, yet tools to precisely control the activation of endogenous orexin signaling are lacking. Here, we developed a photocaged orexin-B (photo-OXB) through a C-terminal photocaging strategy. We show that photo-OXB is unable to activate its cognate receptors in the dark but releases functionally active native orexin-B upon uncaging by illumination with UV-visible (UV-vis) light (370-405 nm). We established an all-optical assay combining photo-OXB with a genetically encoded orexin biosensor and used it to characterize the efficiency and spatial profile of photo-OXB uncaging. Finally, we demonstrated that photo-OXB enables optical control over orexin signaling with fine temporal precision both in vitro and ex vivo. Thus, our photocaging strategy and photo-OXB advance the chemical biological toolkit by introducing a method for the optical control of peptide signaling and physiological function.
Collapse
|
16
|
Girven KS, Mangieri L, Bruchas MR. Emerging approaches for decoding neuropeptide transmission. Trends Neurosci 2022; 45:899-912. [PMID: 36257845 PMCID: PMC9671847 DOI: 10.1016/j.tins.2022.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Neuropeptides produce robust effects on behavior across species, and recent research has benefited from advances in high-resolution techniques to investigate peptidergic transmission and expression throughout the brain in model systems. Neuropeptides exhibit distinct characteristics which includes their post-translational processing, release from dense core vesicles, and ability to activate G-protein-coupled receptors (GPCRs). These complex properties have driven the need for development of specialized tools that can sense neuropeptide expression, cell activity, and release. Current research has focused on isolating when and how neuropeptide transmission occurs, as well as the conditions in which neuropeptides directly mediate physiological and adaptive behavioral states. Here we describe the current technological landscape in which the field is operating to decode key questions regarding these dynamic neuromodulators.
Collapse
Affiliation(s)
- Kasey S Girven
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Leandra Mangieri
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA; University of Washington Center for the Neurobiology of Addiction, Pain, and Emotion, Seattle, WA, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
18
|
Heshmati M, Bruchas MR. Historical and Modern Evidence for the Role of Reward Circuitry in Emergence. Anesthesiology 2022; 136:997-1014. [PMID: 35362070 PMCID: PMC9467375 DOI: 10.1097/aln.0000000000004148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Increasing evidence supports a role for brain reward circuitry in modulating arousal along with emergence from anesthesia. Emergence remains an important frontier for investigation, since no drug exists in clinical practice to initiate rapid and smooth emergence. This review discusses clinical and preclinical evidence indicating a role for two brain regions classically considered integral components of the mesolimbic brain reward circuitry, the ventral tegmental area and the nucleus accumbens, in emergence from propofol and volatile anesthesia. Then there is a description of modern systems neuroscience approaches to neural circuit investigations that will help span the large gap between preclinical and clinical investigation with the shared aim of developing therapies to promote rapid emergence without agitation or delirium. This article proposes that neuroscientists include models of whole-brain network activity in future studies to inform the translational value of preclinical investigations and foster productive dialogues with clinician anesthesiologists.
Collapse
Affiliation(s)
- Mitra Heshmati
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Biological Structure, University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Pharmacology, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
Structure-guided optimization of light-activated chimeric G-protein-coupled receptors. Structure 2022; 30:1075-1087.e4. [PMID: 35588733 DOI: 10.1016/j.str.2022.04.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 01/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest human receptor family and involved in virtually every physiological process. One hallmark of their function is specific coupling to selected signaling pathways. The ability to tune this coupling would make development of receptors with new capabilities possible. Complexes of GPCRs and G-proteins have recently been resolved at high resolution, but this information was in only few cases harnessed for rational receptor engineering. Here, we demonstrate structure-guided optimization of light-activated OptoXRs. Our hypothesis was that incorporation of GPCR-Gα contacts would lead to improved coupling. We first evaluated structure-based alignments for chimeric receptor fusion. We then show in a light-activated β2AR that including Gα contacts increased signaling 7- to 20-fold compared with other designs. In turn, contact elimination diminished function. Finally, this platform allowed optimization of a further OptoXR and spectral tuning. Our work exemplifies structure-based OptoXR development for targeted cell and network manipulation.
Collapse
|
20
|
Smith SJ, von Zastrow M. A Molecular Landscape of Mouse Hippocampal Neuromodulation. Front Neural Circuits 2022; 16:836930. [PMID: 35601530 PMCID: PMC9120848 DOI: 10.3389/fncir.2022.836930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adaptive neuronal circuit function requires a continual adjustment of synaptic network parameters known as “neuromodulation.” This process is now understood to be based primarily on the binding of myriad secreted “modulatory” ligands such as dopamine, serotonin and the neuropeptides to G protein-coupled receptors (GPCRs) that, in turn, regulate the function of the ion channels that establish synaptic weights and membrane excitability. Many of the basic molecular mechanisms of neuromodulation are now known, but the organization of neuromodulation at a network level is still an enigma. New single-cell RNA sequencing data and transcriptomic neurotaxonomies now offer bright new lights to shine on this critical “dark matter” of neuroscience. Here we leverage these advances to explore the cell-type-specific expression of genes encoding GPCRs, modulatory ligands, ion channels and intervening signal transduction molecules in mouse hippocampus area CA1, with the goal of revealing broad outlines of this well-studied brain structure’s neuromodulatory network architecture.
Collapse
Affiliation(s)
- Stephen J Smith
- Allen Institute for Brain Science, Seattle, WA, United States
- *Correspondence: Stephen J Smith,
| | - Mark von Zastrow
- Departments of Psychiatry and Pharmacology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
21
|
Brofiga M, Massobrio P. Brain-on-a-Chip: Dream or Reality? Front Neurosci 2022; 16:837623. [PMID: 35310088 PMCID: PMC8924512 DOI: 10.3389/fnins.2022.837623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/04/2022] [Indexed: 01/01/2023] Open
Affiliation(s)
- Martina Brofiga
- Department of Informatics, Bioengineering, Robotics, Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- ScreenNeuroPharm, Sanremo, Italy
| | - Paolo Massobrio
- Department of Informatics, Bioengineering, Robotics, Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- National Institute for Nuclear Physics (INFN), Genova, Italy
- *Correspondence: Paolo Massobrio
| |
Collapse
|
22
|
Nakajima K, Nakabayashi H, Kawahara M. Cell fate‐inducing CARs orthogonally control multiple signaling pathways. Biotechnol J 2022; 17:e2100463. [DOI: 10.1002/biot.202100463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Kyoko Nakajima
- Laboratory of Cell Vaccine Center for Vaccine and Adjuvant Research (CVAR) National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) 7‐6‐8 Saito‐Asagi Ibaraki‐shi Osaka 567‐0085 Japan
| | - Hideto Nakabayashi
- Department of Chemistry and Biotechnology Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113–8656 Japan
| | - Masahiro Kawahara
- Laboratory of Cell Vaccine Center for Vaccine and Adjuvant Research (CVAR) National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) 7‐6‐8 Saito‐Asagi Ibaraki‐shi Osaka 567‐0085 Japan
- Department of Chemistry and Biotechnology Graduate School of Engineering The University of Tokyo 7‐3‐1 Hongo Bunkyo‐ku Tokyo 113–8656 Japan
| |
Collapse
|
23
|
Shen Y, Luchetti A, Fernandes G, Do Heo W, Silva AJ. The emergence of molecular systems neuroscience. Mol Brain 2022; 15:7. [PMID: 34983613 PMCID: PMC8728933 DOI: 10.1186/s13041-021-00885-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Systems neuroscience is focused on how ensemble properties in the brain, such as the activity of neuronal circuits, gives rise to internal brain states and behavior. Many of the studies in this field have traditionally involved electrophysiological recordings and computational approaches that attempt to decode how the brain transforms inputs into functional outputs. More recently, systems neuroscience has received an infusion of approaches and techniques that allow the manipulation (e.g., optogenetics, chemogenetics) and imaging (e.g., two-photon imaging, head mounted fluorescent microscopes) of neurons, neurocircuits, their inputs and outputs. Here, we will review novel approaches that allow the manipulation and imaging of specific molecular mechanisms in specific cells (not just neurons), cell ensembles and brain regions. These molecular approaches, with the specificity and temporal resolution appropriate for systems studies, promise to infuse the field with novel ideas, emphases and directions, and are motivating the emergence of a molecularly oriented systems neuroscience, a new discipline that studies how the spatial and temporal patterns of molecular systems modulate circuits and brain networks, and consequently shape the properties of brain states and behavior.
Collapse
Affiliation(s)
- Yang Shen
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Giselle Fernandes
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Cell-type-specific neuromodulation guides synaptic credit assignment in a spiking neural network. Proc Natl Acad Sci U S A 2021; 118:2111821118. [PMID: 34916291 PMCID: PMC8713766 DOI: 10.1073/pnas.2111821118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/27/2022] Open
Abstract
Synaptic connectivity provides the foundation for our present understanding of neuronal network function, but static connectivity cannot explain learning and memory. We propose a computational role for the diversity of cortical neuronal types and their associated cell-type–specific neuromodulators in improving the efficiency of synaptic weight adjustments for task learning in neuronal networks. Brains learn tasks via experience-driven differential adjustment of their myriad individual synaptic connections, but the mechanisms that target appropriate adjustment to particular connections remain deeply enigmatic. While Hebbian synaptic plasticity, synaptic eligibility traces, and top-down feedback signals surely contribute to solving this synaptic credit-assignment problem, alone, they appear to be insufficient. Inspired by new genetic perspectives on neuronal signaling architectures, here, we present a normative theory for synaptic learning, where we predict that neurons communicate their contribution to the learning outcome to nearby neurons via cell-type–specific local neuromodulation. Computational tests suggest that neuron-type diversity and neuron-type–specific local neuromodulation may be critical pieces of the biological credit-assignment puzzle. They also suggest algorithms for improved artificial neural network learning efficiency.
Collapse
|
25
|
Maltan L, Najjar H, Tiffner A, Derler I. Deciphering Molecular Mechanisms and Intervening in Physiological and Pathophysiological Processes of Ca 2+ Signaling Mechanisms Using Optogenetic Tools. Cells 2021; 10:3340. [PMID: 34943850 PMCID: PMC8699489 DOI: 10.3390/cells10123340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Calcium ion channels are involved in numerous biological functions such as lymphocyte activation, muscle contraction, neurotransmission, excitation, hormone secretion, gene expression, cell migration, memory, and aging. Therefore, their dysfunction can lead to a wide range of cellular abnormalities and, subsequently, to diseases. To date various conventional techniques have provided valuable insights into the roles of Ca2+ signaling. However, their limited spatiotemporal resolution and lack of reversibility pose significant obstacles in the detailed understanding of the structure-function relationship of ion channels. These drawbacks could be partially overcome by the use of optogenetics, which allows for the remote and well-defined manipulation of Ca2+-signaling. Here, we review the various optogenetic tools that have been used to achieve precise control over different Ca2+-permeable ion channels and receptors and associated downstream signaling cascades. We highlight the achievements of optogenetics as well as the still-open questions regarding the resolution of ion channel working mechanisms. In addition, we summarize the successes of optogenetics in manipulating many Ca2+-dependent biological processes both in vitro and in vivo. In summary, optogenetics has significantly advanced our understanding of Ca2+ signaling proteins and the used tools provide an essential basis for potential future therapeutic application.
Collapse
Affiliation(s)
| | | | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (L.M.); (H.N.); (A.T.)
| |
Collapse
|
26
|
Covey DP, Yocky AG. Endocannabinoid Modulation of Nucleus Accumbens Microcircuitry and Terminal Dopamine Release. Front Synaptic Neurosci 2021; 13:734975. [PMID: 34497503 PMCID: PMC8419321 DOI: 10.3389/fnsyn.2021.734975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
The nucleus accumbens (NAc) is located in the ventromedial portion of the striatum and is vital to valence-based predictions and motivated action. The neural architecture of the NAc allows for complex interactions between various cell types that filter incoming and outgoing information. Dopamine (DA) input serves a crucial role in modulating NAc function, but the mechanisms that control terminal DA release and its effect on NAc neurons continues to be elucidated. The endocannabinoid (eCB) system has emerged as an important filter of neural circuitry within the NAc that locally shapes terminal DA release through various cell type- and site-specific actions. Here, we will discuss how eCB signaling modulates terminal DA release by shaping the activity patterns of NAc neurons and their afferent inputs. We then discuss recent technological advancements that are capable of dissecting how distinct cell types, their afferent projections, and local neuromodulators influence valence-based actions.
Collapse
Affiliation(s)
- Dan P Covey
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| | - Alyssa G Yocky
- Department of Neuroscience, Lovelace Biomedical Research Institute, Albuquerque, NM, United States
| |
Collapse
|
27
|
Hicks JM, Yao YC, Barber S, Neate N, Watts JA, Noy A, Rawson FJ. Electric Field Induced Biomimetic Transmembrane Electron Transport Using Carbon Nanotube Porins. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102517. [PMID: 34269516 DOI: 10.1002/smll.202102517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/27/2021] [Indexed: 06/13/2023]
Abstract
Cells modulate their homeostasis through the control of redox reactions via transmembrane electron transport systems. These are largely mediated via oxidoreductase enzymes. Their use in biology has been linked to a host of systems including reprogramming for energy requirements in cancer. Consequently, the ability to modulate membrane redox systems may give rise to opportunities to modulate underlying biology. The current work aims to develop a wireless bipolar electrochemical approach to form on-demand electron transfer across biological membranes. To achieve this goal, it is shown that by using membrane inserted carbon nanotube porins (CNTPs) that can act as bipolar nanoelectrodes, one can control electron flow with externally applied electric fields across membranes. Before this work, bipolar electrochemistry has been thought to require high applied voltages not compatible with biological systems. It is shown that bipolar electrochemical reaction via gold reduction at the nanotubes can be modulated at low cell-friendly voltages, providing an opportunity to use bipolar electrodes to control electron flux across membranes. The authors provide new mechanistic insight into this newly describe phenomena at the nanoscale. The results presented give rise to a new method using CNTPs to modulate cell behavior via wireless control of membrane electron transfer.
Collapse
Affiliation(s)
- Jacqueline M Hicks
- Biodiscovery Institute, School of Pharmacy, Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Yun-Chiao Yao
- School of Natural Sciences, University of California Merced, Merced, 95343, USA
- Materials Science Division, Lawrence Livermore National Laboratory, Livermore, 94550, USA
| | - Sydney Barber
- Materials Science Division, Lawrence Livermore National Laboratory, Livermore, 94550, USA
- United States Naval Academy, Annapolis, 21402, USA
| | - Nigel Neate
- Nanoscale and Microscale Research Centre, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Julie A Watts
- Biodiscovery Institute, School of Pharmacy, Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Aleksandr Noy
- School of Natural Sciences, University of California Merced, Merced, 95343, USA
- Materials Science Division, Lawrence Livermore National Laboratory, Livermore, 94550, USA
| | - Frankie J Rawson
- Biodiscovery Institute, School of Pharmacy, Division of Regenerative Medicine and Cellular Therapies, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
28
|
Optogenetically-inspired neuromodulation: Translating basic discoveries into therapeutic strategies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 159:187-219. [PMID: 34446246 DOI: 10.1016/bs.irn.2021.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Optogenetic tools allow for the selective activation, inhibition or modulation of genetically-defined neural circuits with incredible temporal precision. Over the past decade, application of these tools in preclinical models of psychiatric disease has advanced our understanding the neural circuit basis of maladaptive behaviors in these disorders. Despite their power as an investigational tool, optogenetics cannot yet be applied in the clinical for the treatment of neurological and psychiatric disorders. To date, deep brain stimulation (DBS) is the only clinical treatment that can be used to achieve circuit-specific neuromodulation in the context of psychiatric. Despite its increasing clinical indications, the mechanisms underlying the therapeutic effects of DBS for psychiatric disorders are poorly understood, which makes optimization difficult. We discuss the variety of optogenetic tools available for preclinical research, and how these tools have been leveraged to reverse-engineer the mechanisms underlying DBS for movement and compulsive disorders. We review studies that have used optogenetics to induce plasticity within defined basal ganglia circuits, to alter neural circuit function and evaluate the corresponding effects on motor and compulsive behaviors. While not immediately applicable to patient populations, the translational power of optogenetics is in inspiring novel DBS protocols by providing a rationale for targeting defined neural circuits to ameliorate specific behavioral symptoms, and by establishing optimal stimulation paradigms that could selectively compensate for pathological synaptic plasticity within these defined neural circuits.
Collapse
|
29
|
Wang T, Ulrich H, Semyanov A, Illes P, Tang Y. Optical control of purinergic signaling. Purinergic Signal 2021; 17:385-392. [PMID: 34156578 PMCID: PMC8410941 DOI: 10.1007/s11302-021-09799-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 06/07/2021] [Indexed: 12/29/2022] Open
Abstract
Purinergic signaling plays a pivotal role in physiological processes and pathological conditions. Over the past decades, conventional pharmacological, biochemical, and molecular biology techniques have been utilized to investigate purinergic signaling cascades. However, none of them is capable of spatially and temporally manipulating purinergic signaling cascades. Currently, optical approaches, including optopharmacology and optogenetic, enable controlling purinergic signaling with low invasiveness and high spatiotemporal precision. In this mini-review, we discuss optical approaches for controlling purinergic signaling and their applications in basic and translational science.
Collapse
Affiliation(s)
- Tao Wang
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Henning Ulrich
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Sechenov First Moscow State Medical University, Moscow, Russia
| | - Peter Illes
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China. .,Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany.
| | - Yong Tang
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China. .,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
30
|
Abstract
Optobiochemical control of protein activities allows the investigation of protein functions in living cells with high spatiotemporal resolution. Over the last two decades, numerous natural photosensory domains have been characterized and synthetic domains engineered and assembled into photoregulatory systems to control protein function with light. Here, we review the field of optobiochemistry, categorizing photosensory domains by chromophore, describing photoregulatory systems by mechanism of action, and discussing protein classes frequently investigated using optical methods. We also present examples of how spatial or temporal control of proteins in living cells has provided new insights not possible with traditional biochemical or cell biological techniques.
Collapse
Affiliation(s)
- Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, South Korea;
| | - Michael Z Lin
- Department of Neurobiology, Stanford University, Stanford, California 94305, USA;
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
31
|
Abstract
Reliable optogenetic tools for sustained, projection-specific presynaptic silencing have been elusive. Recently in Neuron, Mahn et al. (2021) and Copits et al. (2021) describe how the light-activated inhibitory GPCRs eOPN3 and PPO can be used to reversibly suppress synaptic transmission in mice.
Collapse
|
32
|
Francioni V, Harnett MT. Rethinking Single Neuron Electrical Compartmentalization: Dendritic Contributions to Network Computation In Vivo. Neuroscience 2021; 489:185-199. [PMID: 34116137 DOI: 10.1016/j.neuroscience.2021.05.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/11/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Decades of experimental and theoretical work support a now well-established theory that active dendritic processing contributes to the computational power of individual neurons. This theory is based on the high degree of electrical compartmentalization observed in the dendrites of single neurons in ex vivo preparations. Compartmentalization allows dendrites to conduct semi-independent operations on their inputs before final integration and output at the axon, producing a "network-in-a-neuron." However, recent in vivo functional imaging experiments in mouse cortex have reported surprisingly little evidence for strong dendritic compartmentalization. In this review, we contextualize these new findings and discuss their impact on the future of the field. Specifically, we consider how highly coordinated, and thus less compartmentalized, activity in soma and dendrites can contribute to cortical computations including nonlinear mixed selectivity, prediction/expectation, multiplexing, and credit assignment.
Collapse
Affiliation(s)
- Valerio Francioni
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Mark T Harnett
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
33
|
Copits BA, Gowrishankar R, O'Neill PR, Li JN, Girven KS, Yoo JJ, Meshik X, Parker KE, Spangler SM, Elerding AJ, Brown BJ, Shirley SE, Ma KKL, Vasquez AM, Stander MC, Kalyanaraman V, Vogt SK, Samineni VK, Patriarchi T, Tian L, Gautam N, Sunahara RK, Gereau RW, Bruchas MR. A photoswitchable GPCR-based opsin for presynaptic inhibition. Neuron 2021; 109:1791-1809.e11. [PMID: 33979635 PMCID: PMC8194251 DOI: 10.1016/j.neuron.2021.04.026] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022]
Abstract
Optical manipulations of genetically defined cell types have generated significant insights into the dynamics of neural circuits. While optogenetic activation has been relatively straightforward, rapid and reversible synaptic inhibition has proven more elusive. Here, we leveraged the natural ability of inhibitory presynaptic GPCRs to suppress synaptic transmission and characterize parapinopsin (PPO) as a GPCR-based opsin for terminal inhibition. PPO is a photoswitchable opsin that couples to Gi/o signaling cascades and is rapidly activated by pulsed blue light, switched off with amber light, and effective for repeated, prolonged, and reversible inhibition. PPO rapidly and reversibly inhibits glutamate, GABA, and dopamine release at presynaptic terminals. Furthermore, PPO alters reward behaviors in a time-locked and reversible manner in vivo. These results demonstrate that PPO fills a significant gap in the neuroscience toolkit for rapid and reversible synaptic inhibition and has broad utility for spatiotemporal control of inhibitory GPCR signaling cascades.
Collapse
Affiliation(s)
- Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Raaj Gowrishankar
- Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA, USA
| | - Patrick R O'Neill
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA; Shirley and Stefan Hatos Center for Neuropharmacology, Semel Institute, Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA
| | - Jun-Nan Li
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kasey S Girven
- Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA, USA
| | - Judy J Yoo
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xenia Meshik
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle E Parker
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Skylar M Spangler
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abigail J Elerding
- Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA, USA
| | - Bobbie J Brown
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sofia E Shirley
- Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA, USA
| | - Kelly K L Ma
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexis M Vasquez
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - M Christine Stander
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sherri K Vogt
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vijay K Samineni
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tommaso Patriarchi
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Roger K Sunahara
- Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Robert W Gereau
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA; Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
Kramer MM, Lataster L, Weber W, Radziwill G. Optogenetic Approaches for the Spatiotemporal Control of Signal Transduction Pathways. Int J Mol Sci 2021; 22:5300. [PMID: 34069904 PMCID: PMC8157557 DOI: 10.3390/ijms22105300] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Biological signals are sensed by their respective receptors and are transduced and processed by a sophisticated intracellular signaling network leading to a signal-specific cellular response. Thereby, the response to the signal depends on the strength, the frequency, and the duration of the stimulus as well as on the subcellular signal progression. Optogenetic tools are based on genetically encoded light-sensing proteins facilitating the precise spatiotemporal control of signal transduction pathways and cell fate decisions in the absence of natural ligands. In this review, we provide an overview of optogenetic approaches connecting light-regulated protein-protein interaction or caging/uncaging events with steering the function of signaling proteins. We briefly discuss the most common optogenetic switches and their mode of action. The main part deals with the engineering and application of optogenetic tools for the control of transmembrane receptors including receptor tyrosine kinases, the T cell receptor and integrins, and their effector proteins. We also address the hallmarks of optogenetics, the spatial and temporal control of signaling events.
Collapse
Affiliation(s)
- Markus M. Kramer
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
- SGBM—Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Levin Lataster
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
| | - Wilfried Weber
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
- SGBM—Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Gerald Radziwill
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
| |
Collapse
|
35
|
Gutzeit VA, Acosta-Ruiz A, Munguba H, Häfner S, Landra-Willm A, Mathes B, Mony J, Yarotski D, Börjesson K, Liston C, Sandoz G, Levitz J, Broichhagen J. A fine-tuned azobenzene for enhanced photopharmacology in vivo. Cell Chem Biol 2021; 28:1648-1663.e16. [PMID: 33735619 DOI: 10.1016/j.chembiol.2021.02.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/23/2020] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Despite the power of photopharmacology for interrogating signaling proteins, many photopharmacological systems are limited by their efficiency, speed, or spectral properties. Here, we screen a library of azobenzene photoswitches and identify a urea-substituted "azobenzene-400" core that offers bistable switching between cis and trans with improved kinetics, light sensitivity, and a red-shift. We then focus on the metabotropic glutamate receptors (mGluRs), neuromodulatory receptors that are major pharmacological targets. Synthesis of "BGAG12,400," a photoswitchable orthogonal, remotely tethered ligand (PORTL), enables highly efficient, rapid optical agonism following conjugation to SNAP-tagged mGluR2 and permits robust optical control of mGluR1 and mGluR5 signaling. We then produce fluorophore-conjugated branched PORTLs to enable dual imaging and manipulation of mGluRs and highlight their power in ex vivo slice and in vivo behavioral experiments in the mouse prefrontal cortex. Finally, we demonstrate the generalizability of our strategy by developing an improved soluble, photoswitchable pore blocker for potassium channels.
Collapse
Affiliation(s)
- Vanessa A Gutzeit
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Amanda Acosta-Ruiz
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hermany Munguba
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stephanie Häfner
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Arnaud Landra-Willm
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Bettina Mathes
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Jürgen Mony
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Dzianis Yarotski
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Karl Börjesson
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41296 Gothenburg, Sweden
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Guillaume Sandoz
- Université Cote d'Azur, CNRS, INSERM, iBV, Nice, France; Laboratories of Excellence, Ion Channel Science and Therapeutics, Nice, France
| | - Joshua Levitz
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Johannes Broichhagen
- Department of Chemical Biology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany; Department of Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany.
| |
Collapse
|
36
|
Abstract
Allosteric regulation in proteins is fundamental to many important biological processes. Allostery has been employed to control protein functions by regulating protein activity. Engineered allosteric regulation allows controlling protein activity in subsecond time scale and has a broad range of applications, from dissecting spatiotemporal dynamics in biochemical cascades to applications in biotechnology and medicine. Here, we review the concept of allostery in proteins and various approaches to identify allosteric sites and pathways. We then provide an overview of strategies and tools used in allosteric protein regulation and their utility in biological applications. We highlight various classes of proteins, where regulation is achieved through allostery. Finally, we analyze the current problems, critical challenges, and future prospective in achieving allosteric regulation in proteins.
Collapse
Affiliation(s)
| | - Jiaxing Chen
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, United States
- Departments of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, United States
- Department of Chemistry, Penn State University, University Park, Pennsylvania 16802, United States
- Department of Biomedical Engineering, Penn State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
37
|
Optogenetic Modulation of Ion Channels by Photoreceptive Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:73-88. [PMID: 33398808 DOI: 10.1007/978-981-15-8763-4_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In these 15 years, researches to control cellular responses by light have flourished dramatically to establish "optogenetics" as a research field. In particular, light-dependent excitation/inhibition of neural cells using channelrhodopsins or other microbial rhodopsins is the most powerful and the most widely used optogenetic technique. New channelrhodopsin-based optogenetic tools having favorable characteristics have been identified from a wide variety of organisms or created through mutagenesis. Despite the great efforts, some neuronal activities are still hard to be manipulated by the channelrhodopsin-based tools, indicating that complementary approaches are needed to make optogenetics more comprehensive. One of the feasible and complementary approaches is optical control of ion channels using photoreceptive proteins other than channelrhodopsins. In particular, animal opsins can modulate various ion channels via light-dependent G protein activation. In this chapter, we summarize how such alternative optogenetic tools work and they will be improved.
Collapse
|
38
|
Gaur P, Galkin M, Hauke S, Redkin R, Barnes C, Shvadchak VV, Yushchenko DA. Reversible spatial and temporal control of lipid signaling. Chem Commun (Camb) 2020; 56:10646-10649. [PMID: 32857092 DOI: 10.1039/d0cc04146g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein, we introduce versatile molecular tools that enable specific delivery and visualization of photoswitchable lipids at cellular membranes, namely at the plasma membrane and internal membranes. These molecules were prepared by tethering ortho-nitrobenzyl-based fluorescent cages with a signaling lipid bearing an azobenzene photoswitch. They permit two sequential photocontrolled reactions, which are uncaging of a lipid analogue and then its repeated activation and deactivation. We used these molecules to activate GPR40 receptor transiently expressed in HeLa cells and demonstrated downstream modulation of intracellular Ca2+ levels.
Collapse
Affiliation(s)
- Pankaj Gaur
- Laboratory of Chemical Biology, The Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague 6, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
39
|
Mortensen M, Huckvale R, Pandurangan AP, Baker JR, Smart TG. Optopharmacology reveals a differential contribution of native GABA A receptors to dendritic and somatic inhibition using azogabazine. Neuropharmacology 2020; 176:108135. [PMID: 32445639 PMCID: PMC7482436 DOI: 10.1016/j.neuropharm.2020.108135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/20/2020] [Accepted: 05/09/2020] [Indexed: 12/13/2022]
Abstract
γ-aminobutyric acid type-A receptors (GABAARs) are inhibitory ligand-gated ion channels in the brain that are crucial for controlling neuronal excitation. To explore their physiological roles in cellular and neural network activity, it is important to understand why specific GABAAR isoforms are distributed not only to various brain regions and cell types, but also to specific areas of the membrane in individual neurons. To address this aim we have developed a novel photosensitive compound, azogabazine, that targets and reversibly inhibits GABAARs. The receptor selectivity of the compound is based on the competitive antagonist, gabazine, and photosensitivity is conferred by a photoisomerisable azobenzene group. Azogabazine can exist in either cis or trans conformations that are controlled by UV and blue light respectively, to affect receptor inhibition. We report that the trans-isomer preferentially binds and inhibits GABAAR function, whilst promotion of the cis-isomer caused unbinding of azogabazine from GABAARs. Using cultured cerebellar granule cells, azogabazine in conjunction with UV light applied to defined membrane domains, revealed higher densities of GABAARs at somatic inhibitory synapses compared to those populating proximal dendritic zones, even though the latter displayed a higher number of synapses per unit area of membrane. Azogabazine also revealed more pronounced GABA-mediated inhibition of action potential firing in proximal dendrites compared to the soma. Overall, azogabazine is a valuable addition to the photochemical toolkit that can be used to interrogate GABAAR function and inhibition.
Collapse
Affiliation(s)
- Martin Mortensen
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Rosemary Huckvale
- The Institute of Cancer Research, 123 Old Brompton Road, London, SW7 3RP, UK
| | - Arun P Pandurangan
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - James R Baker
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK
| | - Trevor G Smart
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
40
|
Kamimura HAS, Conti A, Toschi N, Konofagou EE. Ultrasound neuromodulation: mechanisms and the potential of multimodal stimulation for neuronal function assessment. FRONTIERS IN PHYSICS 2020; 8:150. [PMID: 32509757 PMCID: PMC7274478 DOI: 10.3389/fphy.2020.00150] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Focused ultrasound (FUS) neuromodulation has shown that mechanical waves can interact with cell membranes and mechanosensitive ion channels, causing changes in neuronal activity. However, the thorough understanding of the mechanisms involved in these interactions are hindered by different experimental conditions for a variety of animal scales and models. While the lack of complete understanding of FUS neuromodulation mechanisms does not impede benefiting from the current known advantages and potential of this technique, a precise characterization of its mechanisms of action and their dependence on experimental setup (e.g., tuning acoustic parameters and characterizing safety ranges) has the potential to exponentially improve its efficacy as well as spatial and functional selectivity. This could potentially reach the cell type specificity typical of other, more invasive techniques e.g., opto- and chemogenetics or at least orientation-specific selectivity afforded by transcranial magnetic stimulation. Here, the mechanisms and their potential overlap are reviewed along with discussions on the potential insights into mechanisms that magnetic resonance imaging sequences along with a multimodal stimulation approach involving electrical, magnetic, chemical, light, and mechanical stimuli can provide.
Collapse
Affiliation(s)
- Hermes A. S. Kamimura
- Ultrasound Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New Yor, NY, USA
| | - Allegra Conti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Charlestown, MA, USA
| | - Elisa E. Konofagou
- Ultrasound Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New Yor, NY, USA
| |
Collapse
|
41
|
Lesca E. Light-Sensitive Membrane Proteins as Tools to Generate Precision Treatments. J Membr Biol 2020; 253:81-86. [PMID: 32248246 DOI: 10.1007/s00232-020-00115-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION BY ANA-NICOLETA BONDAR, BIOPHYSICS SECTION HEAD EDITOR: This issue of the Journal of Membrane Biology inaugurates Up-and-Coming Scientist, in which investigators at early career stages are invited to present recent research in the broad context of their discipline. We inaugurate Up-and-Coming Scientist with the essay by Dr. Elena Lesca of the ETH Zürich and the Paul Scherrer Institut, Switzerland. Dr. Lesca has completed her doctoral degree at the Technical University München, Germany, in 2014, and pursued postdoctoral research at the ETH Zürich and Paul Scherrer Institut, where she is Senior Assistant since 2019. Two recent papers by Dr. Lesca et al. (references 33 and 39) have used X-ray crystallography and experimental biophysics approaches to shed light on the mechanism of action of a membrane receptor from the G Protein-Coupled Receptor (GPCR) family, Jumping Spider Rhodopsin-1 (JSR-1). JSR-1 is a visual rhodopsin activated upon absorption of light by its covalently bound retinal chromophore. Unlike the better-understood bovine rhodopsin GPCR, which is monostable, JSR-1 is bistable (i.e., in JSR-1 the Schiff base that binds retinal to the protein stays protonated throughout the reaction cycle), and absorption of a second photon resets the retinal ligand to the resting state configuration. In her essay, Dr. Lesca discusses the implications of her work on JSR-1 and, more broadly, GPCR research, for state-of-the-art applications in optogenetics and drug design.
Collapse
Affiliation(s)
- Elena Lesca
- Department of Biology, ETH Zürich, 8093, Zurich, Switzerland. .,Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, 5232, Villigen, Switzerland.
| |
Collapse
|
42
|
Paoletti P, Ellis-Davies GCR, Mourot A. Optical control of neuronal ion channels and receptors. Nat Rev Neurosci 2020; 20:514-532. [PMID: 31289380 DOI: 10.1038/s41583-019-0197-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Light-controllable tools provide powerful means to manipulate and interrogate brain function with relatively low invasiveness and high spatiotemporal precision. Although optogenetic approaches permit neuronal excitation or inhibition at the network level, other technologies, such as optopharmacology (also known as photopharmacology) have emerged that provide molecular-level control by endowing light sensitivity to endogenous biomolecules. In this Review, we discuss the challenges and opportunities of photocontrolling native neuronal signalling pathways, focusing on ion channels and neurotransmitter receptors. We describe existing strategies for rendering receptors and channels light sensitive and provide an overview of the neuroscientific insights gained from such approaches. At the crossroads of chemistry, protein engineering and neuroscience, optopharmacology offers great potential for understanding the molecular basis of brain function and behaviour, with promises for future therapeutics.
Collapse
Affiliation(s)
- Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France.
| | | | - Alexandre Mourot
- Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS), CNRS, INSERM, Sorbonne Université, Paris, France.
| |
Collapse
|
43
|
Lutzu S, Castillo PE. Modulation of NMDA Receptors by G-protein-coupled receptors: Role in Synaptic Transmission, Plasticity and Beyond. Neuroscience 2020; 456:27-42. [PMID: 32105741 DOI: 10.1016/j.neuroscience.2020.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 01/11/2023]
Abstract
NMDA receptors (NMDARs) play a critical role in excitatory synaptic transmission, plasticity and in several forms of learning and memory. In addition, NMDAR dysfunction is believed to underlie a number of neuropsychiatric conditions. Growing evidence has demonstrated that NMDARs are tightly regulated by several G-protein-coupled receptors (GPCRs). Ligands that bind to GPCRs, such as neurotransmitters and neuromodulators, activate intracellular pathways that modulate NMDAR expression, subcellular localization and/or functional properties in a short- or a long-term manner across many synapses throughout the central nervous system. In this review article we summarize current knowledge on the molecular and cellular mechanisms underlying NMDAR modulation by GPCRs, and we discuss the implications of this modulation spanning from synaptic transmission and plasticity to circuit function and brain disease.
Collapse
Affiliation(s)
- Stefano Lutzu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
44
|
Delaney SL, Gendreau JL, D'Souza M, Feng AY, Ho AL. Optogenetic Modulation for the Treatment of Traumatic Brain Injury. Stem Cells Dev 2020; 29:187-197. [DOI: 10.1089/scd.2019.0187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
| | | | | | - Austin Y. Feng
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, Georgia
| | - Allen L. Ho
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, Georgia
| |
Collapse
|
45
|
Yu X, Nagai J, Khakh BS. Improved tools to study astrocytes. Nat Rev Neurosci 2020; 21:121-138. [DOI: 10.1038/s41583-020-0264-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
46
|
Guo J, Otis JM, Suciu SK, Catalano C, Xing L, Constable S, Wachten D, Gupton S, Lee J, Lee A, Blackley KH, Ptacek T, Simon JM, Schurmans S, Stuber GD, Caspary T, Anton ES. Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2019; 51:759-774.e5. [PMID: 31846650 PMCID: PMC6953258 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA.
| | - James M Otis
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah K Suciu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Catalano
- Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA
| | - Lei Xing
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stephanie Gupton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Janice Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Amelia Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Katherine H Blackley
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis Ptacek
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Stephane Schurmans
- Laboratory of Functional Genetics, GIGA Research Center, University of Liège, Liège, Belgium
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain and Emotion, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
47
|
Grupi A, Ashur I, Degani-Katzav N, Yudovich S, Shapira Z, Marzouq A, Morgenstein L, Mandel Y, Weiss S. Interfacing the Cell with "Biomimetic Membrane Proteins". SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903006. [PMID: 31765076 DOI: 10.1002/smll.201903006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/30/2019] [Indexed: 06/10/2023]
Abstract
Integral membrane proteins mediate a myriad of cellular processes and are the target of many therapeutic drugs. Enhancement and extension of the functional scope of membrane proteins can be realized by membrane incorporation of engineered nanoparticles designed for specific diagnostic and therapeutic applications. In contrast to hydrophobic insertion of small amphiphilic molecules, delivery and membrane incorporation of particles on the nanometric scale poses a crucial barrier for technological development. In this perspective, the transformative potential of biomimetic membrane proteins (BMPs), current state of the art, and the barriers that need to be overcome in order to advance the field are discussed.
Collapse
Affiliation(s)
- Asaf Grupi
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Idan Ashur
- Agricultural Research Organization, The Volcani Center, Institute of Agricultural Engineering, Rishon LeZion, 7505101, Israel
| | - Nurit Degani-Katzav
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shimon Yudovich
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Zehavit Shapira
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Adan Marzouq
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Lion Morgenstein
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Yossi Mandel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- School of Optometry and Vision Science, Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shimon Weiss
- Department of Physics, Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
48
|
Branched Photoswitchable Tethered Ligands Enable Ultra-efficient Optical Control and Detection of G Protein-Coupled Receptors In Vivo. Neuron 2019; 105:446-463.e13. [PMID: 31784287 DOI: 10.1016/j.neuron.2019.10.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/26/2019] [Accepted: 10/27/2019] [Indexed: 01/01/2023]
Abstract
The limitations of classical drugs have spurred the development of covalently tethered photoswitchable ligands to control neuromodulatory receptors. However, a major shortcoming of tethered photopharmacology is the inability to obtain optical control with an efficacy comparable with that of the native ligand. To overcome this, we developed a family of branched photoswitchable compounds to target metabotropic glutamate receptors (mGluRs). These compounds permit photo-agonism of Gi/o-coupled group II mGluRs with near-complete efficiency relative to glutamate when attached to receptors via a range of orthogonal, multiplexable modalities. Through a chimeric approach, branched ligands also allow efficient optical control of Gq-coupled mGluR5, which we use to probe the spatiotemporal properties of receptor-induced calcium oscillations. In addition, we report branched, photoswitch-fluorophore compounds for simultaneous receptor imaging and manipulation. Finally, we demonstrate this approach in vivo in mice, where photoactivation of SNAP-mGluR2 in the medial prefrontal cortex reversibly modulates working memory in normal and disease-associated states.
Collapse
|
49
|
Smith SJ, Sümbül U, Graybuck LT, Collman F, Seshamani S, Gala R, Gliko O, Elabbady L, Miller JA, Bakken TE, Rossier J, Yao Z, Lein E, Zeng H, Tasic B, Hawrylycz M. Single-cell transcriptomic evidence for dense intracortical neuropeptide networks. eLife 2019; 8:47889. [PMID: 31710287 PMCID: PMC6881117 DOI: 10.7554/elife.47889] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022] Open
Abstract
Seeking new insights into the homeostasis, modulation and plasticity of cortical synaptic networks, we have analyzed results from a single-cell RNA-seq study of 22,439 mouse neocortical neurons. Our analysis exposes transcriptomic evidence for dozens of molecularly distinct neuropeptidergic modulatory networks that directly interconnect all cortical neurons. This evidence begins with a discovery that transcripts of one or more neuropeptide precursor (NPP) and one or more neuropeptide-selective G-protein-coupled receptor (NP-GPCR) genes are highly abundant in all, or very nearly all, cortical neurons. Individual neurons express diverse subsets of NP signaling genes from palettes encoding 18 NPPs and 29 NP-GPCRs. These 47 genes comprise 37 cognate NPP/NP-GPCR pairs, implying the likelihood of local neuropeptide signaling. Here, we use neuron-type-specific patterns of NP gene expression to offer specific, testable predictions regarding 37 peptidergic neuromodulatory networks that may play prominent roles in cortical homeostasis and plasticity.
Collapse
Affiliation(s)
| | - Uygar Sümbül
- Allen Institute for Brain Science, Seattle, United States
| | | | | | | | - Rohan Gala
- Allen Institute for Brain Science, Seattle, United States
| | - Olga Gliko
- Allen Institute for Brain Science, Seattle, United States
| | - Leila Elabbady
- Allen Institute for Brain Science, Seattle, United States
| | | | | | - Jean Rossier
- Neuroscience Paris Seine, Sorbonne Université, Paris, France
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, United States
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, United States
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, United States
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, United States
| | | |
Collapse
|
50
|
Seo DO, Motard LE, Bruchas MR. Contemporary strategies for dissecting the neuronal basis of neurodevelopmental disorders. Neurobiol Learn Mem 2019; 165:106835. [PMID: 29550367 PMCID: PMC6138573 DOI: 10.1016/j.nlm.2018.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/22/2018] [Accepted: 03/13/2018] [Indexed: 01/07/2023]
Abstract
Great efforts in clinical and basic research have shown progress in understanding the neurobiological mechanisms of neurodevelopmental disorders, such as autism, schizophrenia, and attention-deficit hyperactive disorders. Literature on this field have suggested that these disorders are affected by the complex interaction of genetic, biological, psychosocial and environmental risk factors. However, this complexity of interplaying risk factors during neurodevelopment has prevented a complete understanding of the causes of those neuropsychiatric symptoms. Recently, with advances in modern high-resolution neuroscience methods, the neural circuitry analysis approach has provided new solutions for understanding the causal relationship between dysfunction of a neural circuit and behavioral alteration in neurodevelopmental disorders. In this review we will discuss recent progress in developing novel optogenetic and chemogenetic strategies to investigate neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dong-Oh Seo
- Departmentof Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Laura E Motard
- Departmentof Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Michael R Bruchas
- Departmentof Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, United States; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States; Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|