1
|
Jiang Y, Shen L, Wang B. Non-electrophysiological techniques targeting transient receptor potential (TRP) gene of gastrointestinal tract. Int J Biol Macromol 2024; 262:129551. [PMID: 38367416 DOI: 10.1016/j.ijbiomac.2024.129551] [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: 10/25/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/19/2024]
Abstract
Transient receptor potential (TRP) channels are cation channels related to a wide range of physical and chemical stimuli, they are expressed all along the gastrointestinal system, and a myriad of diseases are often associated with aberrant expression or mutation of the TRP gene, suggesting that TRPs are promising targets for drug therapy. Therefore, a better understanding of the information of TRPs in health and disease could facilitate the development of effective drugs for the treatment of gastrointestinal diseases like IBD. But there are very few generalizations about the experimental techniques studied in this field. In view of the promise of TRP as a therapeutic target, we discuss experimental methods that can be used for TRPs including their distribution, function and interaction with other proteins, as well as some promising emerging technologies to provide experimental methods for future studies.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
2
|
Fordyce BA, Roth BL. Making Sense of Psychedelics in the CNS. Int J Neuropsychopharmacol 2024; 27:pyae007. [PMID: 38289825 PMCID: PMC10888522 DOI: 10.1093/ijnp/pyae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024] Open
Abstract
For centuries, ancient lineages have consumed psychedelic compounds from natural sources. In the modern era, scientists have since harnessed the power of computational tools, cellular assays, and behavioral metrics to study how these compounds instigate changes on molecular, cellular, circuit-wide, and system levels. Here, we provide a brief history of psychedelics and their use in science, medicine, and culture. We then outline current techniques for studying psychedelics from a pharmacological perspective. Finally, we address known gaps in the field and potential avenues of further research to broaden our collective understanding of physiological changes induced by psychedelics, the limits of their therapeutic capabilities, and how researchers can improve and inform treatments that are rapidly becoming accessible worldwide.
Collapse
Affiliation(s)
- Blake A Fordyce
- Department of Neuroscience, UNC Chapel Hill Medical School Chapel Hill, North Carolina, USA
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill Medical School Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Hayward RF, Brooks FP, Yang S, Gao S, Cohen AE. Diminishing neuronal acidification by channelrhodopsins with low proton conduction. eLife 2023; 12:RP86833. [PMID: 37801078 PMCID: PMC10558203 DOI: 10.7554/elife.86833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Abstract
Many channelrhodopsins are permeable to protons. We found that in neurons, activation of a high-current channelrhodopsin, CheRiff, led to significant acidification, with faster acidification in the dendrites than in the soma. Experiments with patterned optogenetic stimulation in monolayers of HEK cells established that the acidification was due to proton transport through the opsin, rather than through other voltage-dependent channels. We identified and characterized two opsins which showed large photocurrents, but small proton permeability, PsCatCh2.0 and ChR2-3M. PsCatCh2.0 showed excellent response kinetics and was also spectrally compatible with simultaneous voltage imaging with QuasAr6a. Stimulation-evoked acidification is a possible source of disruptions to cell health in scientific and prospective therapeutic applications of optogenetics. Channelrhodopsins with low proton permeability are a promising strategy for avoiding these problems.
Collapse
Affiliation(s)
- Rebecca Frank Hayward
- School of Engineering and Applied Sciences, Harvard UniversityCambridgeUnited States
| | - F Phil Brooks
- Department of Chemistry, Harvard UniversityCambridgeUnited States
| | - Shang Yang
- Department of Neurophysiology, University of WurzburgWurzburgGermany
| | - Shiqiang Gao
- Department of Neurophysiology, University of WurzburgWurzburgGermany
| | - Adam E Cohen
- Department of Chemistry, Harvard UniversityCambridgeUnited States
- Department of Physics, Harvard UniversityCambridgeUnited States
| |
Collapse
|
4
|
Hayward RF, Brooks FP, Yang S, Gao S, Cohen AE. Diminishing neuronal acidification by channelrhodopsins with low proton conduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527404. [PMID: 36798192 PMCID: PMC9934520 DOI: 10.1101/2023.02.07.527404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Many channelrhodopsins are permeable to protons. We found that in neurons, activation of a high-current channelrhodopsin, CheRiff, led to significant acidification, with faster acidification in the dendrites than in the soma. Experiments with patterned optogenetic stimulation in monolayers of HEK cells established that the acidification was due to proton transport through the opsin, rather than through other voltage-dependent channels. We identified and characterized two opsins which showed large photocurrents, but small proton permeability, PsCatCh2.0 and ChR2-3M. PsCatCh2.0 showed excellent response kinetics and was also spectrally compatible with simultaneous voltage imaging with QuasAr6a. Stimulation-evoked acidification is a possible source of disruptions to cell health in scientific and prospective therapeutic applications of optogenetics. Channelrhodopsins with low proton permeability are a promising strategy for avoiding these problems. Statement of Significance Acidification is an undesirable artifact of optogenetic stimulation. Low proton-permeability opsins minimize this artifact while still allowing robust optogenetic control.
Collapse
Affiliation(s)
| | - F. Phil Brooks
- Department of Chemistry, Harvard University, Cambridge, MA 02138
| | - Shang Yang
- Department of Neurophysiology, University of Wurzburg, Germany
| | - Shiqiang Gao
- Department of Neurophysiology, University of Wurzburg, Germany
| | - Adam E Cohen
- Department of Chemistry, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| |
Collapse
|
5
|
Tian H, Davis HC, Wong-Campos JD, Park P, Fan LZ, Gmeiner B, Begum S, Werley CA, Borja GB, Upadhyay H, Shah H, Jacques J, Qi Y, Parot V, Deisseroth K, Cohen AE. Video-based pooled screening yields improved far-red genetically encoded voltage indicators. Nat Methods 2023; 20:1082-1094. [PMID: 36624211 PMCID: PMC10329731 DOI: 10.1038/s41592-022-01743-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023]
Abstract
Video-based screening of pooled libraries is a powerful approach for directed evolution of biosensors because it enables selection along multiple dimensions simultaneously from large libraries. Here we develop a screening platform, Photopick, which achieves precise phenotype-activated photoselection over a large field of view (2.3 × 2.3 mm, containing >103 cells, per shot). We used the Photopick platform to evolve archaerhodopsin-derived genetically encoded voltage indicators (GEVIs) with improved signal-to-noise ratio (QuasAr6a) and kinetics (QuasAr6b). These GEVIs gave improved signals in cultured neurons and in live mouse brains. By combining targeted in vivo optogenetic stimulation with high-precision voltage imaging, we characterized inhibitory synaptic coupling between individual cortical NDNF (neuron-derived neurotrophic factor) interneurons, and excitatory electrical synapses between individual hippocampal parvalbumin neurons. The QuasAr6 GEVIs are powerful tools for all-optical electrophysiology and the Photopick approach could be adapted to evolve a broad range of biosensors.
Collapse
Affiliation(s)
- He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Hunter C Davis
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - J David Wong-Campos
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Pojeong Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Linlin Z Fan
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Benjamin Gmeiner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Shahinoor Begum
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | | | | | | | | | | | - Yitong Qi
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Vicente Parot
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Institute for Biological and Medical Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karl Deisseroth
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Chevy Chase, MA, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Physics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
6
|
Marchese-Rojas M, Islas ÁA, Mancilla-Simbro C, Millan-PerezPeña L, León JS, Salinas-Stefanon EM. Inhibition of the Human Neuronal Sodium Channel Na v1.9 by Arachidonyl-2-Chloroethylamide, An Analogue of Anandamide in a hNa v1.9/rNa v1.4 Chimera, An Experimental and in Silico Study. Neuroscience 2023; 511:39-52. [PMID: 36156289 DOI: 10.1016/j.neuroscience.2022.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/12/2022] [Accepted: 09/16/2022] [Indexed: 01/23/2023]
Abstract
Cannabinoids regulate analgesia, which has aroused much interest in identifying new pharmacological therapies in the management of refractory pain. Voltage-gated Na+ channels (Navs) play an important role in inflammatory and neuropathic pain. In particular, Nav1.9 is involved in nociception and the understanding of its pharmacology has lagged behind because it is difficult to express in heterologous systems. Here, we utilized the chimeric channel hNav1.9_C4, that comprises the extracellular and transmembrane domains of hNav1.9, co-expressed with the ß1 subunit on CHO-K1 cells to characterize the electrophysiological effects of ACEA, a synthetic surrogate of the endogenous cannabinoid anandamide. ACEA induced a tonic block, decelerated the fast inactivation, markedly shifted steady-state inactivation in the hyperpolarized direction, decreasing the window current and showed use-dependent block, with a high affinity for the inactivated state (ki = 0.84 µM). Thus, we argue that ACEA possess a local anaesthetic-like profile. To provide a mechanistic understanding of its mode of action at the molecular level, we combined induced fit docking with Monte Carlo simulations and electrostatic complementarity. In agreement with the experimental evidence, our computer simulations revealed that ACEA binds Tyr1599 of the local anaesthetics binding site of the hNav1.9, contacting residues that bind cannabinol (CBD) in the NavMs channel. ACEA adopted a conformation remarkably similar to the crystallographic conformation of anandamide on a non-homologous protein, obstructing the Na+ permeation pathway below the selectivity filter to occupy a highly conserved binding pocket at the intracellular side. These results describe a mechanism of action, possibly involved in cannabinoid analgesia.
Collapse
Affiliation(s)
- Mario Marchese-Rojas
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Ángel A Islas
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla, Mexico; Laboratory of Computational Molecular Simulations, Department of Pharmacy, Benemérita Universidad Autónoma de Puebla, Mexico.
| | - Claudia Mancilla-Simbro
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | | | - Jorge S León
- Laboratory of Biophysics, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Mexico
| | | |
Collapse
|
7
|
Kannan M, Vasan G, Haziza S, Huang C, Chrapkiewicz R, Luo J, Cardin JA, Schnitzer MJ, Pieribone VA. Dual-polarity voltage imaging of the concurrent dynamics of multiple neuron types. Science 2022; 378:eabm8797. [PMID: 36378956 PMCID: PMC9703638 DOI: 10.1126/science.abm8797] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Genetically encoded fluorescent voltage indicators are ideally suited to reveal the millisecond-scale interactions among and between targeted cell populations. However, current indicators lack the requisite sensitivity for in vivo multipopulation imaging. We describe next-generation green and red voltage sensors, Ace-mNeon2 and VARNAM2, and their reverse response-polarity variants pAce and pAceR. Our indicators enable 0.4- to 1-kilohertz voltage recordings from >50 spiking neurons per field of view in awake mice and ~30-minute continuous imaging in flies. Using dual-polarity multiplexed imaging, we uncovered brain state–dependent antagonism between neocortical somatostatin-expressing (SST
+
) and vasoactive intestinal peptide–expressing (VIP
+
) interneurons and contributions to hippocampal field potentials from cell ensembles with distinct axonal projections. By combining three mutually compatible indicators, we performed simultaneous triple-population imaging. These approaches will empower investigations of the dynamic interplay between neuronal subclasses at single-spike resolution.
Collapse
Affiliation(s)
- Madhuvanthi Kannan
- The John B. Pierce Laboratory, New Haven, CT 06519, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Ganesh Vasan
- The John B. Pierce Laboratory, New Haven, CT 06519, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
| | - Simon Haziza
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Cheng Huang
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Radosław Chrapkiewicz
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
| | - Junjie Luo
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Jessica A. Cardin
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
- Kavli Institute of Neuroscience, Yale University, New Haven, CT 06520, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06520, USA
| | - Mark J. Schnitzer
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Vincent A. Pieribone
- The John B. Pierce Laboratory, New Haven, CT 06519, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
8
|
Luo Q, Zhang J, Huang M, Lin G, Tanaka M, Lepler S, Guan J, Siemann D, Tang X. Automatic Multi-functional Integration Program (AMFIP) towards all-optical mechano-electrophysiology interrogation. PLoS One 2022; 17:e0266098. [PMID: 35901062 PMCID: PMC9333221 DOI: 10.1371/journal.pone.0266098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Automatic operations of multi-functional and time-lapse live-cell imaging are necessary for the biomedical science community to study active, multi-faceted, and long-term biological phenomena. To achieve automatic control, most existing solutions often require the purchase of extra software programs and hardware that rely on the manufacturers’ own specifications. However, these software programs are usually non-user-programmable and unaffordable for many laboratories. To address this unmet need, we have developed a novel open-source software program, titled Automatic Multi-functional Integration Program (AMFIP), as a new Java-based and hardware-independent system that provides proven advantages over existing alternatives to the scientific community. Without extra hardware, AMFIP enables the functional synchronization of the μManager software platform, the Nikon NIS-Elements platform, and other 3rd party software to achieve automatic operations of most commercially available microscopy systems, including but not limited to those from Nikon. AMFIP provides a user-friendly and programmable graphical user interface (GUI), opening the door to expanding the customizability for myriad hardware and software systems according to user-specific experimental requirements and environments. To validate the intended purposes of developing AMFIP, we applied it to elucidate the question whether single cells, prior to their full spreading, can sense and respond to a soft solid substrate, and if so, how does the interaction depend on the cell spreading time and the stiffness of the substrate. Using a CRISPR/Cas9-engineered human epithelial Beas2B (B2B) cell line that expresses mNeonGreen2-tagged mechanosensitive Yes-associated protein (YAP), we show that single B2B cells develop distinct substrate-stiffness-dependent YAP expressions within 10 hours at most on the substrate, suggesting that cells are able to sense, distinguish, and respond to mechanical cues prior to the establishment of full cell spreading. In summary, AMFIP provides a reliable, open-source, and cost-free solution that has the validated long-term utility to satisfy the need of automatic imaging operations in the scientific community.
Collapse
Affiliation(s)
- Qin Luo
- Department of Electrical and Computer Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Justin Zhang
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, California, United States of America
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, UF, Gainesville, Florida, United States of America
| | - Gaoming Lin
- Department of Electrical and Computer Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, United States of America
| | - Mai Tanaka
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- UF Health Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Sharon Lepler
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- UF Health Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Juan Guan
- UF Health Cancer Center, University of Florida, Gainesville, Florida, United States of America
- Department of Physics, College of Liberal Arts and Sciences, University of Florida, Gainesville, Florida, United States of America
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Dietmar Siemann
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- UF Health Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, UF, Gainesville, Florida, United States of America
- UF Health Cancer Center, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
9
|
Borja GB, Zhang H, Harwood BN, Jacques J, Grooms J, Chantre RO, Zhang D, Barnett A, Werley CA, Lu Y, Nagle SF, McManus OB, Dempsey GT. Highly Parallelized, Multicolor Optogenetic Recordings of Cellular Activity for Therapeutic Discovery Applications in Ion Channels and Disease-Associated Excitable Cells. Front Mol Neurosci 2022; 15:896320. [PMID: 35860501 PMCID: PMC9289666 DOI: 10.3389/fnmol.2022.896320] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Optogenetic assays provide a flexible, scalable, and information rich approach to probe compound effects for ion channel drug targets in both heterologous expression systems and associated disease relevant cell types. Despite the potential utility and growing adoption of optogenetics, there remains a critical need for compatible platform technologies with the speed, sensitivity, and throughput to enable their application to broader drug screening applications. To address this challenge, we developed the SwarmTM, a custom designed optical instrument for highly parallelized, multicolor measurements in excitable cells, simultaneously recording changes in voltage and calcium activities at high temporal resolution under optical stimulation. The compact design featuring high power LEDs, large numerical aperture optics, and fast photodiode detection enables all-optical individual well readout of 24-wells simultaneously from multi-well plates while maintaining sufficient temporal resolution to probe millisecond response dynamics. The Swarm delivers variable intensity blue-light optogenetic stimulation to enable membrane depolarization and red or lime-light excitation to enable fluorescence detection of the resulting changes in membrane potential or calcium levels, respectively. The Swarm can screen ~10,000 wells/day in 384-well format, probing complex pharmacological interactions via a wide array of stimulation protocols. To evaluate the Swarm screening system, we optimized a series of heterologous optogenetic spiking HEK293 cell assays for several voltage-gated sodium channel subtypes including Nav1.2, Nav1.5, and Nav1.7. The Swarm was able to record pseudo-action potentials stably across all 24 objectives and provided pharmacological characterization of diverse sodium channel blockers. We performed a Nav1.7 screen of 200,000 small molecules in a 384-well plate format with all 560 plates reaching a Z' > 0.5. As a demonstration of the versatility of the Swarm, we also developed an assay measuring cardiac action potential and calcium waveform properties simultaneously under paced conditions using human induced pluripotent stem (iPS) cell-derived cardiomyocytes as an additional counter screen for cardiac toxicity. In summary, the Swarm is a novel high-throughput all-optical system capable of collecting information-dense data from optogenetic assays in both heterologous and iPS cell-derived models, which can be leveraged to drive diverse therapeutic discovery programs for nervous system disorders and other disease areas involving excitable cells.
Collapse
|
10
|
Roy D, Shapira Z, Weiss S. Membrane Potential Sensing: Materials Design & Methods Development for Single Particle Optical Electrophysiology. J Chem Phys 2022; 156:084201. [DOI: 10.1063/5.0076522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Debjit Roy
- Department of Chemistry & Biochemistry, University of California Los Angeles, United States of America
| | - Zehavit Shapira
- Department of Physics, The institute of nanotechnology, Bar-Ilan University, Israel
| | - Shimon Weiss
- Department of Chemistry and Biochemistry, University of California Los Angeles Department of Chemistry and Biochemistry, United States of America
- Bar-Ilan University
| |
Collapse
|
11
|
Wan J, Zhou S, Mea HJ, Guo Y, Ku H, Urbina BM. Emerging Roles of Microfluidics in Brain Research: From Cerebral Fluids Manipulation to Brain-on-a-Chip and Neuroelectronic Devices Engineering. Chem Rev 2022; 122:7142-7181. [PMID: 35080375 DOI: 10.1021/acs.chemrev.1c00480] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Remarkable progress made in the past few decades in brain research enables the manipulation of neuronal activity in single neurons and neural circuits and thus allows the decipherment of relations between nervous systems and behavior. The discovery of glymphatic and lymphatic systems in the brain and the recently unveiled tight relations between the gastrointestinal (GI) tract and the central nervous system (CNS) further revolutionize our understanding of brain structures and functions. Fundamental questions about how neurons conduct two-way communications with the gut to establish the gut-brain axis (GBA) and interact with essential brain components such as glial cells and blood vessels to regulate cerebral blood flow (CBF) and cerebrospinal fluid (CSF) in health and disease, however, remain. Microfluidics with unparalleled advantages in the control of fluids at microscale has emerged recently as an effective approach to address these critical questions in brain research. The dynamics of cerebral fluids (i.e., blood and CSF) and novel in vitro brain-on-a-chip models and microfluidic-integrated multifunctional neuroelectronic devices, for example, have been investigated. This review starts with a critical discussion of the current understanding of several key topics in brain research such as neurovascular coupling (NVC), glymphatic pathway, and GBA and then interrogates a wide range of microfluidic-based approaches that have been developed or can be improved to advance our fundamental understanding of brain functions. Last, emerging technologies for structuring microfluidic devices and their implications and future directions in brain research are discussed.
Collapse
Affiliation(s)
- Jiandi Wan
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Sitong Zhou
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Hing Jii Mea
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Yaojun Guo
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Hansol Ku
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Brianna M Urbina
- Biochemistry, Molecular, Cellular and Developmental Biology Program, University of California, Davis, California 95616, United States
| |
Collapse
|
12
|
Abstract
Optogenetics has revolutionized not only neuroscience but also had an impact on muscle physiology and cell biology. Rhodopsin-based optogenetics started with the discovery of the light-gated cation channels, called channelrhodopsins. Together with the light-driven ion pumps, these channels allow light-mediated control of electrically excitable cells in culture tissue and living animals. They can be activated (depolarized) or silenced (hyperpolarized) by light with incomparably high spatiotemporal resolution. Optogenetics allows the light manipulation of cells under electrode-free conditions in a minimally invasive manner. Through modern genetic techniques, virus-induced transduction can be performed with extremely high cell specificity in tissue and living animals, allowing completely new approaches for analyzing neural networks, behavior studies, and investigations of neurodegenerative diseases. First clinical trials for the optogenetic recovery of vision are underway.This chapter provides a comprehensive description of the structure and function of the different light-gated channels and some new light-activated ion pumps. Some of them already play an essential role in optogenetics while others are supposed to become important tools for more specialized applications in the future.At the moment, a large number of publications are available concerning intrinsic mechanisms of microbial rhodopsins. Mostly they describe CrChR2 and its variants, as CrChR2 is still the most prominent optogenetic tool. Therefore, many biophysically and biochemically oriented groups contributed to the overwhelming mass of information on this unique ion channel's molecular mechanism. In this context, the function of new optogenetic tools is discussed, which is essential for rational optimization of the optogenetic approach for an eventual biomedical application. The comparison of the effectivity of ion pumps versus ion channels is discussed as well.Applications of rhodopsins-based optogenetic tools are also discussed in the chapter. Because of the enormous number of these applications in neuroscience, only exemplary studies on cell culture neural tissue, muscle physiology, and remote control of animal behavior are presented.
Collapse
Affiliation(s)
- Alexey Alekseev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Russia
| | - Valentin Gordeliy
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Ernst Bamberg
- Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Bloxham W, Brinks D, Kheifets S, Cohen AE. Linearly polarized excitation enhances signals from fluorescent voltage indicators. Biophys J 2021; 120:5333-5342. [PMID: 34710379 DOI: 10.1016/j.bpj.2021.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/10/2021] [Accepted: 10/20/2021] [Indexed: 10/20/2022] Open
Abstract
Voltage imaging in cells requires high-speed recording of small fluorescent signals, often leading to low signal/noise ratios. Because voltage indicators are membrane bound, their orientations are partially constrained by the plane of the membrane. We explored whether tuning the linear polarization of excitation light could enhance voltage indicator fluorescence. We tested a panel of dye- and protein-based voltage indicators in mammalian cells. The dye BeRST1 showed a 73% increase in brightness between the least and most favorable polarizations. The protein-based reporter ASAP1 showed a 22% increase in brightness, and QuasAr3 showed a 14% increase in brightness. In very thin neurites expressing QuasAr3, improvements were anomalously large, with a 170% increase in brightness between polarization parallel versus perpendicular to the dendrite. Signal/noise ratios of optically recorded action potentials were increased by up to 50% in neurites expressing QuasAr3. These results demonstrate that polarization control can be a facile means to enhance signals from fluorescent voltage indicators, particularly in thin neurites or in high-background environments.
Collapse
Affiliation(s)
- William Bloxham
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Physics, Harvard University, Cambridge, Massachusetts; Physics of Living Systems, Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Daan Brinks
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands; Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Simon Kheifets
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts; Department of Physics, Harvard University, Cambridge, Massachusetts.
| |
Collapse
|
14
|
Zhang XM, Yokoyama T, Sakamoto M. Imaging Voltage with Microbial Rhodopsins. Front Mol Biosci 2021; 8:738829. [PMID: 34513932 PMCID: PMC8423911 DOI: 10.3389/fmolb.2021.738829] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
Membrane potential is the critical parameter that reflects the excitability of a neuron, and it is usually measured by electrophysiological recordings with electrodes. However, this is an invasive approach that is constrained by the problems of lacking spatial resolution and genetic specificity. Recently, the development of a variety of fluorescent probes has made it possible to measure the activity of individual cells with high spatiotemporal resolution. The adaptation of this technique to image electrical activity in neurons has become an informative method to study neural circuits. Genetically encoded voltage indicators (GEVIs) can be used with superior performance to accurately target specific genetic populations and reveal neuronal dynamics on a millisecond scale. Microbial rhodopsins are commonly used as optogenetic actuators to manipulate neuronal activities and to explore the circuit mechanisms of brain function, but they also can be used as fluorescent voltage indicators. In this review, we summarize recent advances in the design and the application of rhodopsin-based GEVIs.
Collapse
Affiliation(s)
- Xiao Min Zhang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tatsushi Yokoyama
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Masayuki Sakamoto
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Kyoto, Japan
| |
Collapse
|
15
|
Wang H, Kelley FM, Milovanovic D, Schuster BS, Shi Z. Surface tension and viscosity of protein condensates quantified by micropipette aspiration. BIOPHYSICAL REPORTS 2021; 1:100011. [PMID: 36247368 PMCID: PMC9563586 DOI: 10.1016/j.bpr.2021.100011] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022]
Abstract
The material properties of biomolecular condensates have been suggested to play important biological and pathological roles. Despite the rapid increase in the number of biomolecules identified that undergo liquid-liquid phase separation, quantitative studies and direct measurements of the material properties of the resulting condensates have been severely lagging behind. Here, we develop a micropipette-based technique that uniquely, to our knowledge, allows quantifications of both the surface tension and viscosity of biomolecular condensates, independent of labeling and surface-wetting effects. We demonstrate the accuracy and versatility of this technique by measuring condensates of LAF-1 RGG domains and a polymer-based aqueous two-phase system. We further confirm our measurements using established condensate fusion and fluorescence recovery after photobleaching assays. We anticipate the micropipette-based technique will be widely applicable to biomolecular condensates and will resolve several limitations regarding current approaches.
Collapse
Affiliation(s)
- Huan Wang
- Department of Chemistry and Chemical Biology
| | - Fleurie M. Kelley
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases, Berlin, Germany
| | - Benjamin S. Schuster
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey
| | - Zheng Shi
- Department of Chemistry and Chemical Biology
| |
Collapse
|
16
|
DuBreuil DM, Chiang BM, Zhu K, Lai X, Flynn P, Sapir Y, Wainger BJ. A high-content platform for physiological profiling and unbiased classification of individual neurons. CELL REPORTS METHODS 2021; 1:100004. [PMID: 34318289 PMCID: PMC8312640 DOI: 10.1016/j.crmeth.2021.100004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/18/2021] [Accepted: 03/11/2021] [Indexed: 11/29/2022]
Abstract
High-throughput physiological assays lose single-cell resolution, precluding subtype-specific analyses of activation mechanism and drug effects. We demonstrate APPOINT (automated physiological phenotyping of individual neuronal types), a physiological assay platform combining calcium imaging, robotic liquid handling, and automated analysis to generate physiological activation profiles of single neurons at large scale. Using unbiased techniques, we quantify responses to sequential stimuli, enabling subgroup identification by physiology and probing of distinct mechanisms of neuronal activation within subgroups. Using APPOINT, we quantify primary sensory neuron activation by metabotropic receptor agonists and identify potential contributors to pain signaling. We expand the role of neuroimmune interactions by showing that human serum directly activates sensory neurons, elucidating a new potential pain mechanism. Finally, we apply APPOINT to develop a high-throughput, all-optical approach for quantification of activation threshold and pharmacologically validate contributions of ion channel families to optical activation.
Collapse
Affiliation(s)
- Daniel M. DuBreuil
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Brenda M. Chiang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kevin Zhu
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaofan Lai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Patrick Flynn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Yechiam Sapir
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Brian J. Wainger
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Anesthesiology, Critical Care, & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA
| |
Collapse
|
17
|
Chien MP, Brinks D, Testa-Silva G, Tian H, Phil Brooks F, Adam Y, Bloxham B, Gmeiner B, Kheifets S, Cohen AE. Photoactivated voltage imaging in tissue with an archaerhodopsin-derived reporter. SCIENCE ADVANCES 2021; 7:7/19/eabe3216. [PMID: 33952514 PMCID: PMC8099184 DOI: 10.1126/sciadv.abe3216] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 03/15/2021] [Indexed: 05/19/2023]
Abstract
Photoactivated genetically encoded voltage indicators (GEVIs) have the potential to enable optically sectioned voltage imaging at the intersection of a photoactivation beam and an imaging beam. We developed a pooled high-throughput screen to identify archaerhodopsin mutants with enhanced photoactivation. After screening ~105 cells, we identified a novel GEVI, NovArch, whose one-photon near-infrared fluorescence is reversibly enhanced by weak one-photon blue or two-photon near-infrared excitation. Because the photoactivation leads to fluorescent signals catalytically rather than stoichiometrically, high fluorescence signals, optical sectioning, and high time resolution are achieved simultaneously at modest blue or two-photon laser power. We demonstrate applications of the combined molecular and optical tools to optical mapping of membrane voltage in distal dendrites in acute mouse brain slices and in spontaneously active neurons in vivo.
Collapse
Affiliation(s)
- Miao-Ping Chien
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Daan Brinks
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Imaging Physics, Delft University of Technology, Delft, Netherlands
| | - Guilherme Testa-Silva
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Cambridge, MA 02138, USA
| | - He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - F Phil Brooks
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Yoav Adam
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Blox Bloxham
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Benjamin Gmeiner
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Simon Kheifets
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA.
- Howard Hughes Medical Institute, Cambridge, MA 02138, USA
| |
Collapse
|
18
|
Automated Functional Screening for Modulators of Optogenetically Activated Neural Responses in Living Organisms. Methods Mol Biol 2021. [PMID: 32865748 DOI: 10.1007/978-1-0716-0830-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
All-optical methods of probing in vivo brain function are advantageous for their compatibility with automated microscopy and fast spatial targeting of neural circuit excitation and response. Recent advances in optogenetic technologies allow simultaneous light activation of specific neurons and optical readout of neural activity via fluorescent calcium reporters, providing an attractive opportunity for high-throughput screening assays that directly assess dynamic neural function in vivo. Here we describe a method to automatically record optogenetically activated neural responses in living, hydrogel-embedded organisms over many hours in a multiwell plate format. This method is suitable for screening the neural effects of hundreds of chemical compounds and assessing the time course of bioactivity over 12 h or more. As examples, we show the suppression of neural responses over time with various concentrations of two voltage-gated calcium channel blockers and a full-plate screen of 320 chemicals with positive and negative controls in a single experiment.
Collapse
|
19
|
Bell DC, Dallas ML. Advancing Ion Channel Research with Automated Patch Clamp (APC) Electrophysiology Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:21-32. [DOI: 10.1007/978-981-16-4254-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Thomas M, Hughes TE. Optically activated, customizable, excitable cells. PLoS One 2020; 15:e0229051. [PMID: 33378334 PMCID: PMC7773186 DOI: 10.1371/journal.pone.0229051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 12/03/2020] [Indexed: 11/29/2022] Open
Abstract
Genetically encoded fluorescent biosensors are powerful tools for studying complex signaling in the nervous system, and now both Ca2+ and voltage sensors are available to study the signaling behavior of entire neural circuits. There is a pressing need for improved sensors, but improving them is challenging because testing them involves a low throughput, labor-intensive processes. Our goal was to create synthetic, excitable cells that can be activated with brief pulses of blue light and serve as a medium throughput platform for screening the next generation of sensors. In this live cell system, blue light activates an adenylyl cyclase enzyme (bPAC) that increases intracellular cAMP (Stierl M et al. 2011). In turn, the cAMP opens a cAMP-gated ion channel. This produces slow, whole-cell Ca2+ transients and voltage changes. To increase the speed of these transients, we add the inwardly rectifying potassium channel Kir2.1, the bacterial voltage-gated sodium channel NAVROSD, and Connexin-43. The result is a highly reproducible, medium-throughput, live cell system that can be used to screen voltage and Ca2+ sensors.
Collapse
Affiliation(s)
- Merrilee Thomas
- Department of Neuroscience and Cell Biology, Montana State University, Bozeman, Montana, United States of America
| | - Thomas E. Hughes
- Department of Neuroscience and Cell Biology, Montana State University, Bozeman, Montana, United States of America
- Montana Molecular, Bozeman, Montana, United States of America
| |
Collapse
|
21
|
Potekhina ES, Bass DY, Kelmanson IV, Fetisova ES, Ivanenko AV, Belousov VV, Bilan DS. Drug Screening with Genetically Encoded Fluorescent Sensors: Today and Tomorrow. Int J Mol Sci 2020; 22:E148. [PMID: 33375682 PMCID: PMC7794770 DOI: 10.3390/ijms22010148] [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: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Genetically-encoded fluorescent sensors have been actively developed over the last few decades and used in live imaging and drug screening. Real-time monitoring of drug action in a specific cellular compartment, organ, or tissue type; the ability to screen at the single-cell resolution; and the elimination of false-positive results caused by low drug bioavailability that is not detected by in vitro testing methods are a few of the obvious benefits of using genetically-encoded fluorescent sensors in drug screening. In combination with high-throughput screening (HTS), some genetically-encoded fluorescent sensors may provide high reproducibility and robustness to assays. We provide a brief overview of successful, perspective, and hopeful attempts at using genetically encoded fluorescent sensors in HTS of modulators of ion channels, Ca2+ homeostasis, GPCR activity, and for screening cytotoxic, anticancer, and anti-parasitic compounds. We discuss the advantages of sensors in whole organism drug screening models and the perspectives of the combination of human disease modeling by CRISPR techniques with genetically encoded fluorescent sensors for drug screening.
Collapse
Affiliation(s)
- Ekaterina S. Potekhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dina Y. Bass
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ilya V. Kelmanson
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Elena S. Fetisova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
| | - Alexander V. Ivanenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency, 117997 Moscow, Russia
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (D.Y.B.); (I.V.K.); (E.S.F.); (A.V.I.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
22
|
The Next 50 Years of Neuroscience. J Neurosci 2020; 40:101-106. [PMID: 31896564 DOI: 10.1523/jneurosci.0744-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023] Open
Abstract
On the 50th anniversary of the Society for Neuroscience, we reflect on the remarkable progress that the field has made in understanding the nervous system, and look forward to the contributions of the next 50 years. We predict a substantial acceleration of our understanding of the nervous system that will drive the development of new therapeutic strategies to treat diseases over the course of the next five decades. We also see neuroscience at the nexus of many societal topics beyond medicine, including education, consumerism, and the justice system. In combination, advances made by basic, translational, and clinical neuroscience research in the next 50 years have great potential for lasting improvements in human health, the economy, and society.
Collapse
|
23
|
Tian JJ, Tan CY, Chen QY, Zhou Y, Qu ZW, Zhang M, Ma KT, Shi WY, Li L, Si JQ. Upregulation of Nav1.7 by endogenous hydrogen sulfide contributes to maintenance of neuropathic pain. Int J Mol Med 2020; 46:782-794. [PMID: 32468069 PMCID: PMC7307826 DOI: 10.3892/ijmm.2020.4611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/06/2020] [Indexed: 01/26/2023] Open
Abstract
Nav1.7 is closely associated with neuropathic pain. Hydrogen sulfide (H2S) has recently been reported to be involved in numerous biological functions, and it has been shown that H2S can enhance the sodium current density, and inhibiting the endogenous production of H2S mediated by cystathionine β-synthetase (CBS) using O-(carboxymethyl) hydroxylamine hemihydrochloride (AOAA) can significantly reduce the expression of Nav1.7 and thus the sodium current density in rat dorsal root ganglion (DRG) neurons. In the present study, it was shown that the fluorescence intensity of H2S was increased in a spared nerve injury (SNI) model and AOAA inhibited this increase. Nav1.7 is expressed in DRG neurons, and the expression of CBS and Nav1.7 were increased in DRG neurons 7, 14 and 21 days post-operation. AOAA inhibited the increase in the expression of CBS, phosphorylated (p)-MEK1/2, p-ERK1/2 and Nav1.7 induced by SNI, and U0126 (a MEK blocker) was able to inhibit the increase in p-MEK1/2, p-ERK1/2 and Nav1.7 expression. However, PF-04856264 did not inhibit the increase in CBS, p-MEK1/2, p-ERK1/2 or Nav1.7 expression induced by SNI surgery. The current density of Nav1.7 was significantly increased in the SNI model and administration of AOAA and U0126 both significantly decreased the density. In addition, AOAA, U0126 and PF-04856264 inhibited the decrease in rheobase, and the increase in action potential induced by SNI in DRG neurons. There was no significant difference in thermal withdrawal latency among each group. However, the time the animals spent with their paw lifted increased significantly following SNI, and the time the animals spent with their paw lifted decreased significantly following the administration of AOAA, U0126 and PF-04856264. In conclusion, these data show that Nav1.7 expression in DRG neurons is upregulated by CBS-derived endogenous H2S in an SNI model, contributing to the maintenance of neuropathic pain.
Collapse
Affiliation(s)
- Jun-Jie Tian
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Chao-Yang Tan
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Qin-Yi Chen
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Ying Zhou
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Zu-Wei Qu
- Department of Pharmacology, Shihezi University Pharmaceutical College, Shihezi, Xinjiang 832002, P.R. China
| | - Meng Zhang
- First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Wen-Yan Shi
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Jiaxing University Medical College, Jiaxing, Zhejiang 314001, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Shihezi University Medical College, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
24
|
Zhang H, Moyer BD, Yu V, McGivern JG, Jarosh M, Werley CA, Hecht VC, Babcock RJ, Dong K, Dempsey GT, McManus OB, Hempel CM. Correlation of Optical and Automated Patch Clamp Electrophysiology for Identification of Na V1.7 Inhibitors. SLAS DISCOVERY 2020; 25:434-446. [PMID: 32292096 DOI: 10.1177/2472555220914532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The voltage-gated sodium channel Nav1.7 is a genetically validated target for pain; pharmacological blockers are promising as a new class of nonaddictive therapeutics. The search for Nav1.7 subtype selective inhibitors requires a reliable, scalable, and sensitive assay. Previously, we developed an all-optical electrophysiology (Optopatch) Spiking HEK platform to study activity-dependent modulation of Nav1.7 in a format compatible with high-throughput screening. In this study, we benchmarked the Optopatch Spiking HEK assay with an existing validated automated electrophysiology assay on the IonWorks Barracuda (IWB) platform. In a pilot screen of 3520 compounds, which included compound plates from a random library as well as compound plates enriched for Nav1.7 inhibitors, the Optopatch Spiking HEK assay identified 174 hits, of which 143 were confirmed by IWB. The Optopatch Spiking HEK assay maintained the high reliability afforded by traditional fluorescent assays and further demonstrated comparable sensitivity to IWB measurements. We speculate that the Optopatch assay could provide an affordable high-throughput screening platform to identify novel Nav1.7 subtype selective inhibitors with diverse mechanisms of action, if coupled with a multiwell parallel optogenetic recording instrument.
Collapse
Affiliation(s)
| | - Bryan D Moyer
- Neuroscience, Amgen Research, Thousand Oaks, CA, USA
| | - Violeta Yu
- Neuroscience, Amgen Research, Cambridge, MA, USA
| | - Joseph G McGivern
- Discovery Technologies, Amgen Research, South San Francisco, CA, USA
| | | | | | - Vivian C Hecht
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan J Babcock
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kevin Dong
- Q-State Biosciences, Cambridge, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Chris M Hempel
- Q-State Biosciences, Cambridge, MA, USA.,Expressive Neuroscience, Syracuse, NY, USA
| |
Collapse
|
25
|
Agus V, Janovjak H. All-Optical Miniaturized Co-culture Assay of Voltage-Gated Ca 2+ Channels. Methods Mol Biol 2020; 2173:247-260. [PMID: 32651923 DOI: 10.1007/978-1-0716-0755-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Light-activated proteins enable the reversible and spatiotemporal control of cellular events in optogenetics. Optogenetics is also rapidly expanding into the field of drug discovery where it provides cost-effective and noninvasive approaches for cell manipulation in high-throughput screens. Here, we present a prototypical cell-based assay that applies Channelrhodopsin2 (ChR2) to recapitulate physiological membrane potential changes and test for voltage-gated ion channel (VGIC) blockade. ChR2 and the voltage-gated Ca2+ channel 1.2 (CaV1.2) are expressed in individual HEK293 cell lines that are then co-cultured for formation of gap junctions and an electrical syncytium. This co-culture allows identification of blockers using parallel fluorescence plate readers in the 384-well plate format in an all-optical mode of operation. The assay is transferable to other VGICs by modularly combining new and existing cell lines and potentially also to other drug targets.
Collapse
Affiliation(s)
- Viviana Agus
- Department of Cell Biology, AXXAM S.p.A, Milan, Italy.
| | - 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
| |
Collapse
|
26
|
Lazzari-Dean JR, Gest AM, Miller EW. Optical estimation of absolute membrane potential using fluorescence lifetime imaging. eLife 2019; 8:44522. [PMID: 31545164 PMCID: PMC6814365 DOI: 10.7554/elife.44522] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022] Open
Abstract
All cells maintain ionic gradients across their plasma membranes, producing transmembrane potentials (Vmem). Mounting evidence suggests a relationship between resting Vmem and the physiology of non-excitable cells with implications in diverse areas, including cancer, cellular differentiation, and body patterning. A lack of non-invasive methods to record absolute Vmem limits our understanding of this fundamental signal. To address this need, we developed a fluorescence lifetime-based approach (VF-FLIM) to visualize and optically quantify Vmem with single-cell resolution in mammalian cell culture. Using VF-FLIM, we report Vmem distributions over thousands of cells, a 100-fold improvement relative to electrophysiological approaches. In human carcinoma cells, we visualize the voltage response to growth factor stimulation, stably recording a 10-15 mV hyperpolarization over minutes. Using pharmacological inhibitors, we identify the source of the hyperpolarization as the Ca2+-activated K+ channel KCa3.1. The ability to optically quantify absolute Vmem with cellular resolution will allow a re-examination of its signaling roles.
Collapse
Affiliation(s)
- Julia R Lazzari-Dean
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Anneliese Mm Gest
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Evan W Miller
- Department of Chemistry, University of California, Berkeley, Berkeley, United States.,Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, United States.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
27
|
Penzkofer A, Silapetere A, Hegemann P. Absorption and Emission Spectroscopic Investigation of the Thermal Dynamics of the Archaerhodopsin 3 Based Fluorescent Voltage Sensor QuasAr1. Int J Mol Sci 2019; 20:E4086. [PMID: 31438573 PMCID: PMC6747118 DOI: 10.3390/ijms20174086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022] Open
Abstract
QuasAr1 is a fluorescent voltage sensor derived from Archaerhodopsin 3 (Arch) of Halorubrum sodomense by directed evolution. Here we report absorption and emission spectroscopic studies of QuasAr1 in Tris buffer at pH 8. Absorption cross-section spectra, fluorescence quantum distributions, fluorescence quantum yields, and fluorescence excitation spectra were determined. The thermal stability of QuasAr1 was studied by long-time attenuation coefficient measurements at room temperature (23 ± 2 °C) and at 2.5 ± 0.5 °C. The apparent melting temperature was determined by stepwise sample heating up and cooling down (obtained apparent melting temperature: 65 ± 3 °C). In the protein melting process the originally present protonated retinal Schiff base (PRSB) with absorption maximum at 580 nm converted to de-protonated retinal Schiff base (RSB) with absorption maximum at 380 nm. Long-time storage of QuasAr1 at temperatures around 2.5 °C and around 23 °C caused gradual protonated retinal Schiff base isomer changes to other isomer conformations, de-protonation to retinal Schiff base isomers, and apoprotein structure changes showing up in ultraviolet absorption increase. Reaction coordinate schemes are presented for the thermal protonated retinal Schiff base isomerizations and deprotonations in parallel with the dynamic apoprotein restructurings.
Collapse
Affiliation(s)
- Alfons Penzkofer
- Fakultät für Physik, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Arita Silapetere
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany
| | - Peter Hegemann
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany
| |
Collapse
|
28
|
Nguyen HX, Bursac N. Ion channel engineering for modulation and de novo generation of electrical excitability. Curr Opin Biotechnol 2019; 58:100-107. [PMID: 30776744 DOI: 10.1016/j.copbio.2019.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023]
Abstract
Ion channels play essential roles in regulating electrical properties of excitable tissues. By leveraging various ion channel gating mechanisms, scientists have developed a versatile set of genetically encoded tools to modulate intrinsic tissue excitability under different experimental settings. In this article, we will review how ion channels activated by voltage, light, small chemicals, stretch, and temperature have been customized to enable control of tissue excitability both in vitro and in vivo. Advantages and limitations of each of these ion channel-engineering platforms will be discussed and notable applications will be highlighted. Furthermore, we will describe recent progress on de novo generation of excitable tissues via expression of appropriate sets of engineered voltage-gated ion channels and discuss potential therapeutic implications.
Collapse
Affiliation(s)
- Hung X Nguyen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
29
|
McNamara HM, Dodson S, Huang YL, Miller EW, Sandstede B, Cohen AE. Geometry-Dependent Arrhythmias in Electrically Excitable Tissues. Cell Syst 2018; 7:359-370.e6. [PMID: 30292705 PMCID: PMC6204347 DOI: 10.1016/j.cels.2018.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/14/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022]
Abstract
Little is known about how individual cells sense the macroscopic geometry of their tissue environment. Here, we explore whether long-range electrical signaling can convey information on tissue geometry to individual cells. First, we studied an engineered electrically excitable cell line. Cells grown in patterned islands of different shapes showed remarkably diverse firing patterns under otherwise identical conditions, including regular spiking, period-doubling alternans, and arrhythmic firing. A Hodgkin-Huxley numerical model quantitatively reproduced these effects, showing how the macroscopic geometry affected the single-cell electrophysiology via the influence of gap junction-mediated electrical coupling. Qualitatively similar geometry-dependent dynamics were observed in human induced pluripotent stem cell (iPSC)-derived cardiomyocytes. The cardiac results urge caution in translating observations of arrhythmia in vitro to predictions in vivo, where the tissue geometry is very different. We study how to extrapolate electrophysiological measurements between tissues with different geometries and different gap junction couplings.
Collapse
Affiliation(s)
- Harold M McNamara
- Department of Physics, Harvard University, Cambridge, MA 02138, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02138, USA
| | - Stephanie Dodson
- Department of Applied Mathematics, Brown University, Providence, RI 02912, USA
| | - Yi-Lin Huang
- Departments of Chemistry, Molecular and Cell Biology, and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Evan W Miller
- Departments of Chemistry, Molecular and Cell Biology, and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Björn Sandstede
- Department of Applied Mathematics, Brown University, Providence, RI 02912, USA
| | - Adam E Cohen
- Department of Physics, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
30
|
Colley CS, England E, Linley JE, Wilkinson TCI. Screening Strategies for the Discovery of Ion Channel Monoclonal Antibodies. ACTA ACUST UNITED AC 2018; 82:e44. [DOI: 10.1002/cpph.44] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Caroline S. Colley
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - Elizabeth England
- Antibody Discovery and Protein Engineering, MedImmune; Cambridge United Kingdom
| | - John E. Linley
- Neuroscience, IMED Biotech Unit, AstraZeneca; Cambridge United Kingdom
| | | |
Collapse
|
31
|
Verstraelen P, Van Dyck M, Verschuuren M, Kashikar ND, Nuydens R, Timmermans JP, De Vos WH. Image-Based Profiling of Synaptic Connectivity in Primary Neuronal Cell Culture. Front Neurosci 2018; 12:389. [PMID: 29997468 PMCID: PMC6028601 DOI: 10.3389/fnins.2018.00389] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/22/2018] [Indexed: 12/04/2022] Open
Abstract
Neurological disorders display a broad spectrum of clinical manifestations. Yet, at the cellular level, virtually all these diseases converge into a common phenotype of dysregulated synaptic connectivity. In dementia, synapse dysfunction precedes neurodegeneration and cognitive impairment by several years, making the synapse a crucial entry point for the development of diagnostic and therapeutic strategies. Whereas high-resolution imaging and biochemical fractionations yield detailed insight into the molecular composition of the synapse, standardized assays are required to quickly gauge synaptic connectivity across large populations of cells under a variety of experimental conditions. Such screening capabilities have now become widely accessible with the advent of high-throughput, high-content microscopy. In this review, we discuss how microscopy-based approaches can be used to extract quantitative information about synaptic connectivity in primary neurons with deep coverage. We elaborate on microscopic readouts that may serve as a proxy for morphofunctional connectivity and we critically analyze their merits and limitations. Finally, we allude to the potential of alternative culture paradigms and integrative approaches to enable comprehensive profiling of synaptic connectivity.
Collapse
Affiliation(s)
- Peter Verstraelen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Michiel Van Dyck
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Rony Nuydens
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Cell Systems and Imaging, Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
32
|
Nguyen HX, Kirkton RD, Bursac N. Generation and customization of biosynthetic excitable tissues for electrophysiological studies and cell-based therapies. Nat Protoc 2018; 13:927-945. [PMID: 29622805 PMCID: PMC6050172 DOI: 10.1038/nprot.2018.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We describe a two-stage protocol to generate electrically excitable and actively conducting cell networks with stable and customizable electrophysiological phenotypes. Using this method, we have engineered monoclonally derived excitable tissues as a robust and reproducible platform to investigate how specific ion channels and mutations affect action potential (AP) shape and conduction. In the first stage of the protocol, we combine computational modeling, site-directed mutagenesis, and electrophysiological techniques to derive optimal sets of mammalian and/or prokaryotic ion channels that produce specific AP shape and conduction characteristics. In the second stage of the protocol, selected ion channels are stably expressed in unexcitable human cells by means of viral or nonviral delivery, followed by flow cytometry or antibiotic selection to purify the desired phenotype. This protocol can be used with traditional heterologous expression systems or primary excitable cells, and application of this method to primary fibroblasts may enable an alternative approach to cardiac cell therapy. Compared with existing methods, this protocol generates a well-defined, relatively homogeneous electrophysiological phenotype of excitable cells that facilitates experimental and computational studies of AP conduction and can decrease arrhythmogenic risk upon cell transplantation. Although basic cell culture and molecular biology techniques are sufficient to generate excitable tissues using the described protocol, experience with patch-clamp techniques is required to characterize and optimize derived cell populations.
Collapse
Affiliation(s)
- Hung X Nguyen
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA. Correspondence should be addressed to N.B. ()
| | - Robert D Kirkton
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA. Correspondence should be addressed to N.B. ()
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA. Correspondence should be addressed to N.B. ()
| |
Collapse
|
33
|
Zheng YM, Wang WF, Li YF, Yu Y, Gao ZB. Enhancing inactivation rather than reducing activation of Nav1.7 channels by a clinically effective analgesic CNV1014802. Acta Pharmacol Sin 2018; 39:587-596. [PMID: 29094728 PMCID: PMC5888685 DOI: 10.1038/aps.2017.151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/03/2017] [Indexed: 12/16/2022] Open
Abstract
The Nav1.7 channel represents a promising target for pain relief. In the recent decades, a number of Nav1.7 channel inhibitors have been developed. According to the effects on channel kinetics, these inhibitors could be divided into two major classes: reducing activation or enhancing inactivation. To date, however, only several inhibitors have moved forward into phase 2 clinical trials and most of them display a less than ideal analgesic efficacy, thus intensifying the controversy regarding if an ideal candidate should preferentially affect the activation or inactivation state. In the present study, we investigated the action mechanisms of a recently clinically confirmed inhibitor CNV1014802 using both electrophysiology and site-directed mutagenesis. We found that CNV1014802 inhibited Nav1.7 channels through stabilizing a nonconductive inactivated state. When the cells expressing Nav1.7 channels were hold at 70 mV or 120 mV, the half maximal inhibitory concentration (IC50) values (with 95% confidence limits) were 1.77 (1.20-2.33) and 71.66 (46.85-96.48) μmol/L, respectively. This drug caused dramatic hyperpolarizing shift of channel inactivation but did not affect activation. Moreover, CNV1014802 accelerated the onset of inactivation and delayed the recovery from inactivation. Notably, application of CNV1014802 (30 μmol/L) could rescue the Nav1.7 mutations expressed in CHO cells that cause paroxysmal extreme pain disorder (PEPD), thereby restoring the impaired inactivation to those of the wild-type channel. Our study demonstrates that CNV1014802 enhances the inactivation but does not reduce the activation of Nav1.7 channels, suggesting that identifying inhibitors that preferentially affect inactivation is a promising approach for developing drugs targeting Nav1.7.
Collapse
Affiliation(s)
- Yue-ming Zheng
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wan-fu Wang
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yan-fen Li
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yong Yu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhao-bing Gao
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
34
|
Streit J, Kleinlogel S. Dynamic all-optical drug screening on cardiac voltage-gated ion channels. Sci Rep 2018; 8:1153. [PMID: 29348631 PMCID: PMC5773578 DOI: 10.1038/s41598-018-19412-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/27/2017] [Indexed: 11/09/2022] Open
Abstract
Voltage-gated ion channels (VGCs) are prime targets for the pharmaceutical industry, but drug profiling on VGCs is challenging, since drug interactions are confined to specific conformational channel states mediated by changes in transmembrane potential. Here we combined various optogenetic tools to develop dynamic, high-throughput drug profiling assays with defined light-step protocols to interrogate VGC states on a millisecond timescale. We show that such light-induced electrophysiology (LiEp) yields high-quality pharmacological data with exceptional screening windows for drugs acting on the major cardiac VGCs, including hNav1.5, hKv1.5 and hERG. LiEp-based screening remained robust when using a variety of optogenetic actuators (ChR2, ChR2(H134R), CatCh, ChR2-EYFP-βArchT) and different types of organic (RH421, Di-4-ANBDQPQ, BeRST1) or genetic voltage sensors (QuasAr1). The tractability of LiEp allows a versatile and precise alternative to state-of-the-art VGC drug screening platforms such as automated electrophysiology or FLIPR readers.
Collapse
Affiliation(s)
- Jonas Streit
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland
| | - Sonja Kleinlogel
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland.
| |
Collapse
|
35
|
|
36
|
Xu Y, Zou P, Cohen AE. Voltage imaging with genetically encoded indicators. Curr Opin Chem Biol 2017; 39:1-10. [PMID: 28460291 PMCID: PMC5581692 DOI: 10.1016/j.cbpa.2017.04.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 02/06/2023]
Abstract
Membrane voltages are ubiquitous throughout cell biology. Voltage is most commonly associated with excitable cells such as neurons and cardiomyocytes, although many other cell types and organelles also support electrical signaling. Voltage imaging in vivo would offer unique capabilities in reporting the spatial pattern and temporal dynamics of electrical signaling at the cellular and circuit levels. Voltage is not directly visible, and so a longstanding challenge has been to develop genetically encoded fluorescent voltage indicator proteins. Recent advances have led to a profusion of new voltage indicators, based on different scaffolds and with different tradeoffs between voltage sensitivity, speed, brightness, and spectrum. In this review, we describe recent advances in design and applications of genetically-encoded voltage indicators (GEVIs). We also highlight the protein engineering strategies employed to improve the dynamic range and kinetics of GEVIs and opportunities for future advances.
Collapse
Affiliation(s)
- Yongxian Xu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Peng Zou
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Adam E Cohen
- Departments of Chemistry and Chemical Biology and of Physics, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute.
| |
Collapse
|
37
|
Zhang H, Cohen AE. Optogenetic Approaches to Drug Discovery in Neuroscience and Beyond. Trends Biotechnol 2017; 35:625-639. [PMID: 28552428 PMCID: PMC5495001 DOI: 10.1016/j.tibtech.2017.04.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/08/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
Recent advances in optogenetics have opened new routes to drug discovery, particularly in neuroscience. Physiological cellular assays probe functional phenotypes that connect genomic data to patient health. Optogenetic tools, in particular tools for all-optical electrophysiology, now provide a means to probe cellular disease models with unprecedented throughput and information content. These techniques promise to identify functional phenotypes associated with disease states and to identify compounds that improve cellular function regardless of whether the compound acts directly on a target or through a bypass mechanism. This review discusses opportunities and unresolved challenges in applying optogenetic techniques throughout the discovery pipeline - from target identification and validation, to target-based and phenotypic screens, to clinical trials.
Collapse
Affiliation(s)
- Hongkang Zhang
- Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
38
|
Genetic evidence for role of integration of fast and slow neurotransmission in schizophrenia. Mol Psychiatry 2017; 22:792-801. [PMID: 28348379 PMCID: PMC5495879 DOI: 10.1038/mp.2017.33] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/12/2022]
Abstract
The most recent genome-wide association studies (GWAS) of schizophrenia (SCZ) identified hundreds of risk variants potentially implicated in the disease. Further, novel statistical methodology designed for polygenic architecture revealed more potential risk variants. This can provide a link between individual genetic factors and the mechanistic underpinnings of SCZ. Intriguingly, a large number of genes coding for ionotropic and metabotropic receptors for various neurotransmitters-glutamate, γ-aminobutyric acid (GABA), dopamine, serotonin, acetylcholine and opioids-and numerous ion channels were associated with SCZ. Here, we review these findings from the standpoint of classical neurobiological knowledge of neuronal synaptic transmission and regulation of electrical excitability. We show that a substantial proportion of the identified genes are involved in intracellular cascades known to integrate 'slow' (G-protein-coupled receptors) and 'fast' (ionotropic receptors) neurotransmission converging on the protein DARPP-32. Inspection of the Human Brain Transcriptome Project database confirms that that these genes are indeed expressed in the brain, with the expression profile following specific developmental trajectories, underscoring their relevance to brain organization and function. These findings extend the existing pathophysiology hypothesis by suggesting a unifying role of dysregulation in neuronal excitability and synaptic integration in SCZ. This emergent model supports the concept of SCZ as an 'associative' disorder-a breakdown in the communication across different slow and fast neurotransmitter systems through intracellular signaling pathways-and may unify a number of currently competing hypotheses of SCZ pathophysiology.
Collapse
|
39
|
Hempel CM, Werley CA, Dempsey GT, Gerber DJ. Targeting neuronal function for CNS drug discovery. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017. [PMID: 28647082 DOI: 10.1016/j.ddtec.2017.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is a pressing need for new and more effective treatments for central nervous system (CNS) disorders. A large body of evidence now suggests that alterations in synaptic transmission and neuronal excitability represent underlying factors for many neurological and psychiatric diseases. However, it has been challenging to target these complex functional domains for therapeutic discovery using traditional neuronal assay methods. Here we review advances in neuronal screening technologies and cellular model systems that enable phenotypic screening of neuronal function as a basis for novel CNS drug discovery approaches.
Collapse
Affiliation(s)
- Chris M Hempel
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | | | - Graham T Dempsey
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - David J Gerber
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA.
| |
Collapse
|
40
|
Noebels J. Precision physiology and rescue of brain ion channel disorders. J Gen Physiol 2017; 149:533-546. [PMID: 28428202 PMCID: PMC5412535 DOI: 10.1085/jgp.201711759] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/24/2017] [Indexed: 11/20/2022] Open
Abstract
Ion channel genes, originally implicated in inherited excitability disorders of muscle and heart, have captured a major role in the molecular diagnosis of central nervous system disease. Their arrival is heralded by neurologists confounded by a broad phenotypic spectrum of early-onset epilepsy, autism, and cognitive impairment with few effective treatments. As detection of rare structural variants in channel subunit proteins becomes routine, it is apparent that primary sequence alone cannot reliably predict clinical severity or pinpoint a therapeutic solution. Future gains in the clinical utility of variants as biomarkers integral to clinical decision making and drug discovery depend on our ability to unravel complex developmental relationships bridging single ion channel structure and human physiology.
Collapse
Affiliation(s)
- Jeffrey Noebels
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030 .,Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
41
|
Agus V, Janovjak H. Optogenetic methods in drug screening: technologies and applications. Curr Opin Biotechnol 2017; 48:8-14. [PMID: 28273648 DOI: 10.1016/j.copbio.2017.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/13/2017] [Accepted: 02/15/2017] [Indexed: 12/19/2022]
Abstract
The optogenetic revolution enabled spatially-precise and temporally-precise control over protein function, signaling pathway activation, and animal behavior with tremendous success in the dissection of signaling networks and neural circuits. Very recently, optogenetic methods have been paired with optical reporters in novel drug screening platforms. In these all-optical platforms, light remotely activated ion channels and kinases thereby obviating the use of electrophysiology or reagents. Consequences were remarkable operational simplicity, throughput, and cost-effectiveness that culminated in the identification of new drug candidates. These blueprints for all-optical assays also revealed potential pitfalls and inspire all-optical variants of other screens, such as those that aim at better understanding dynamic drug action or orphan protein function.
Collapse
Affiliation(s)
- Viviana Agus
- AXXAM SpA, via Meucci 3, 20091 Bresso, Milan, Italy
| | - Harald Janovjak
- Institute of Science and Technology (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
42
|
Molokanova E, Mercola M, Savchenko A. Bringing new dimensions to drug discovery screening: impact of cellular stimulation technologies. Drug Discov Today 2017; 22:1045-1055. [PMID: 28179145 DOI: 10.1016/j.drudis.2017.01.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/09/2016] [Accepted: 01/27/2017] [Indexed: 01/08/2023]
Abstract
The current mandate for the drug discovery industry is to develop more efficient drugs faster while reducing the costs associated with their development. Incorporation of cell stimulation technologies during screening assays is expected to revolutionize the discovery of novel drugs as well as safety pharmacology. In this review, we highlight 'classical' and emerging cell stimulation technologies that provide the ability to evaluate the effects of drug candidates on cells in different functional states to assess clinically relevant phenotypes.
Collapse
Affiliation(s)
- Elena Molokanova
- Nanotools Bioscience, Encinitas, CA 92024, USA; Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mark Mercola
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA; Department of Medicine and Cardiovascular Institute, Stanford University, Palo Alto, CA 94304, USA
| | - Alex Savchenko
- Department of Medicine and Cardiovascular Institute, Stanford University, Palo Alto, CA 94304, USA; Department of Pediatrics, University of California-San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
43
|
Genetically Encoded Voltage Indicators: Opportunities and Challenges. J Neurosci 2016; 36:9977-89. [PMID: 27683896 DOI: 10.1523/jneurosci.1095-16.2016] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/25/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED A longstanding goal in neuroscience is to understand how spatiotemporal patterns of neuronal electrical activity underlie brain function, from sensory representations to decision making. An emerging technology for monitoring electrical dynamics, voltage imaging using genetically encoded voltage indicators (GEVIs), couples the power of genetics with the advantages of light. Here, we review the properties that determine indicator performance and applicability, discussing both recent progress and technical limitations. We then consider GEVI applications, highlighting studies that have already deployed GEVIs for biological discovery. We also examine which classes of biological questions GEVIs are primed to address and which ones are beyond their current capabilities. As GEVIs are further developed, we anticipate that they will become more broadly used by the neuroscience community to eavesdrop on brain activity with unprecedented spatiotemporal resolution. SIGNIFICANCE STATEMENT Genetically encoded voltage indicators are engineered light-emitting protein sensors that typically report neuronal voltage dynamics as changes in brightness. In this review, we systematically discuss the current state of this emerging method, considering both its advantages and limitations for imaging neural activity. We also present recent applications of this technology and discuss what is feasible now and what we anticipate will become possible with future indicator development. This review will inform neuroscientists of recent progress in the field and help potential users critically evaluate the suitability of genetically encoded voltage indicator imaging to answer their specific biological questions.
Collapse
|