1
|
Zhang J, Newman J, Wang Z, Qian Y, Feliciano-Ramos P, Guo W, Honda T, Chen ZS, Linghu C, Etienne-Cummings R, Fossum E, Boyden E, Wilson M. Pixel-wise programmability enables dynamic high-SNR cameras for high-speed microscopy. Nat Commun 2024; 15:4480. [PMID: 38802338 DOI: 10.1038/s41467-024-48765-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
High-speed wide-field fluorescence microscopy has the potential to capture biological processes with exceptional spatiotemporal resolution. However, conventional cameras suffer from low signal-to-noise ratio at high frame rates, limiting their ability to detect faint fluorescent events. Here, we introduce an image sensor where each pixel has individually programmable sampling speed and phase, so that pixels can be arranged to simultaneously sample at high speed with a high signal-to-noise ratio. In high-speed voltage imaging experiments, our image sensor significantly increases the output signal-to-noise ratio compared to a low-noise scientific CMOS camera (~2-3 folds). This signal-to-noise ratio gain enables the detection of weak neuronal action potentials and subthreshold activities missed by the standard scientific CMOS cameras. Our camera with flexible pixel exposure configurations offers versatile sampling strategies to improve signal quality in various experimental conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| | - Jonathan Newman
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zeguan Wang
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Yong Qian
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Pedro Feliciano-Ramos
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Wei Guo
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Takato Honda
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zhe Sage Chen
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Changyang Linghu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ralph Etienne-Cummings
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Eric Fossum
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Edward Boyden
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Matthew Wilson
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| |
Collapse
|
2
|
Zhang J, Newman J, Wang Z, Qian Y, Feliciano-Ramos P, Guo W, Honda T, Chen ZS, Linghu C, Etienne-Cummings R, Fossum E, Boyden E, Wilson M. Pixel-wise programmability enables dynamic high-SNR cameras for high-speed microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.27.546748. [PMID: 37425952 PMCID: PMC10327006 DOI: 10.1101/2023.06.27.546748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
High-speed wide-field fluorescence microscopy has the potential to capture biological processes with exceptional spatiotemporal resolution. However, conventional cameras suffer from low signal-to-noise ratio at high frame rates, limiting their ability to detect faint fluorescent events. Here, we introduce an image sensor where each pixel has individually programmable sampling speed and phase, so that pixels can be arranged to simultaneously sample at high speed with a high signal-to-noise ratio. In high-speed voltage imaging experiments, our image sensor significantly increases the output signal-to-noise ratio compared to a low-noise scientific CMOS camera (~2-3 folds). This signal-to-noise ratio gain enables the detection of weak neuronal action potentials and subthreshold activities missed by the standard scientific CMOS cameras. Our camera with flexible pixel exposure configurations offers versatile sampling strategies to improve signal quality in various experimental conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Jonathan Newman
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zeguan Wang
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Yong Qian
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Pedro Feliciano-Ramos
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Wei Guo
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Takato Honda
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Zhe Sage Chen
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Changyang Linghu
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ralph Etienne-Cummings
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Eric Fossum
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Edward Boyden
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Matthew Wilson
- Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| |
Collapse
|
3
|
Liu PW, Zhang H, Werley CA, Pichler M, Ryan SJ, Lewarch CL, Jacques J, Grooms J, Ferrante J, Li G, Zhang D, Bremmer N, Barnett A, Chantre R, Elder AE, Cohen AE, Williams LA, Dempsey GT, McManus OB. A phenotypic screening platform for chronic pain therapeutics using all-optical electrophysiology. Pain 2024; 165:922-940. [PMID: 37963235 PMCID: PMC10950549 DOI: 10.1097/j.pain.0000000000003090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
ABSTRACT Chronic pain associated with osteoarthritis (OA) remains an intractable problem with few effective treatment options. New approaches are needed to model the disease biology and to drive discovery of therapeutics. We present an in vitro model of OA pain, where dorsal root ganglion (DRG) sensory neurons were sensitized by a defined mixture of disease-relevant inflammatory mediators, here called Sensitizing PAin Reagent Composition or SPARC. Osteoarthritis-SPARC components showed synergistic or additive effects when applied in combination and induced pain phenotypes in vivo. To measure the effect of OA-SPARC on neural firing in a scalable format, we used a custom system for high throughput all-optical electrophysiology. This system enabled light-based membrane voltage recordings from hundreds of neurons in parallel with single cell and single action potential resolution and a throughput of up to 500,000 neurons per day. A computational framework was developed to construct a multiparameter OA-SPARC neuronal phenotype and to quantitatively assess phenotype reversal by candidate pharmacology. We screened ∼3000 approved drugs and mechanistically focused compounds, yielding data from over 1.2 million individual neurons with detailed assessment of functional OA-SPARC phenotype rescue and orthogonal "off-target" effects. Analysis of confirmed hits revealed diverse potential analgesic mechanisms including ion channel modulators and other mechanisms including MEK inhibitors and tyrosine kinase modulators. Our results suggest that the Raf-MEK-ERK axis in DRG neurons may integrate the inputs from multiple upstream inflammatory mediators found in osteoarthritis patient joints, and MAPK pathway activation in DRG neurons may contribute to chronic pain in patients with osteoarthritis.
Collapse
Affiliation(s)
- Pin W. Liu
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | | | | | | | | | - Guangde Li
- Quiver Bioscience, Cambridge, MA, United States
| | - Dawei Zhang
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | | | | | | |
Collapse
|
4
|
Heinson YW, Han JL, Entcheva E. OptoDyCE-plate as an affordable high throughput imager for all optical cardiac electrophysiology. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 6:100054. [PMID: 38130942 PMCID: PMC10735237 DOI: 10.1016/j.jmccpl.2023.100054] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
We present a simple low-cost system for comprehensive functional characterization of cardiac function under spontaneous and paced conditions, in standard 96 and 384-well plates. This full-plate actuator/imager, OptoDyCE-plate, uses optogenetic stimulation and optical readouts of voltage and calcium (parallel recordings from up to 100 wells in 384-well plates are demonstrated). The system is validated with syncytia of human induced pluripotent stem cell derived cardiomyocytes, iPSC-CMs, grown as monolayers, or in quasi-3D isotropic and anisotropic constructs using electrospun matrices, in 96 and 384-well format. Genetic modifications, e.g. interference CRISPR (CRISPRi), and nine compounds of acute and chronic action were tested, including five histone deacetylase inhibitors (HDACis). Their effects on voltage and calcium were compared across growth conditions and pacing rates. We also demonstrated optogenetic point pacing via cell spheroids to study conduction in 96-well format, as well as temporal multiplexing to register voltage and calcium simultaneously on a single camera. Opto-DyCE-plate showed excellent performance even in the small samples in 384-well plates. Anisotropic structured constructs may provide some benefits in drug testing, although drug responses were consistent across tested configurations. Differential voltage vs. calcium responses were seen for some drugs, especially for non-traditional modulators of cardiac function, e.g. HDACi, and pacing rate was a powerful modulator of drug response, highlighting the need for comprehensive multiparametric assessment, as offered by OptoDyCE-plate. Increasing throughput and speed and reducing cost of screening can help stratify potential compounds early in the drug development process and accelerate the development of safer drugs.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20052, United States of America
| |
Collapse
|
5
|
Emerson JI, Ariel P, Shi W, Conlon FL. Sex Differences in Mouse Cardiac Electrophysiology Revealed by Simultaneous Imaging of Excitation-Contraction Coupling. J Cardiovasc Dev Dis 2023; 10:479. [PMID: 38132647 PMCID: PMC10743987 DOI: 10.3390/jcdd10120479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Males and females differ in the basic anatomy and physiology of the heart. Sex differences are evident in cardiac repolarization in humans; women have longer corrected QT and JT intervals. However, the molecular mechanisms that lead to these differences are incompletely understood. Here, we present that, like in humans, sex differences in QT and JT intervals exist in mouse models; female mice had longer corrected QT and JT intervals compared with age-matched males. To further understand the molecular underpinning of these sex differences, we developed a novel technology using fluorescent confocal microscopy that allows the simultaneous visualization of action potential, Ca2+ transients, and contractions in isolated cardiomyocytes at a high temporal resolution. From this approach, we uncovered that females at baseline have increased action potential duration, decreased Ca2+ release and reuptake rates, and decreased contraction and relaxation velocities compared with males. Additionally, males had a shorter overall time from action potential onset to peak contraction. In aggregate, our studies uncovered male and female differences in excitation-contraction coupling that account for differences observed in the EKG. Overall, a better understanding of sex differences in electrophysiology is essential for equitably treating cardiac disease.
Collapse
Affiliation(s)
- James I. Emerson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Pablo Ariel
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Wei Shi
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Integrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Junge S, Ricci Signorini ME, Al Masri M, Gülink J, Brüning H, Kasperek L, Szepes M, Bakar M, Gruh I, Heisterkamp A, Torres-Mapa ML. A micro-LED array based platform for spatio-temporal optogenetic control of various cardiac models. Sci Rep 2023; 13:19490. [PMID: 37945622 PMCID: PMC10636122 DOI: 10.1038/s41598-023-46149-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Optogenetics relies on dynamic spatial and temporal control of light to address emerging fundamental and therapeutic questions in cardiac research. In this work, a compact micro-LED array, consisting of 16 × 16 pixels, is incorporated in a widefield fluorescence microscope for controlled light stimulation. We describe the optical design of the system that allows the micro-LED array to fully cover the field of view regardless of the imaging objective used. Various multicellular cardiac models are used in the experiments such as channelrhodopsin-2 expressing aggregates of cardiomyocytes, termed cardiac bodies, and bioartificial cardiac tissues derived from human induced pluripotent stem cells. The pacing efficiencies of the cardiac bodies and bioartificial cardiac tissues were characterized as a function of illumination time, number of switched-on pixels and frequency of stimulation. To demonstrate dynamic stimulation, steering of calcium waves in HL-1 cell monolayer expressing channelrhodopsin-2 was performed by applying different configurations of patterned light. This work shows that micro-LED arrays are powerful light sources for optogenetic control of contraction and calcium waves in cardiac monolayers, multicellular bodies as well as three-dimensional artificial cardiac tissues.
Collapse
Affiliation(s)
- Sebastian Junge
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Maria Elena Ricci Signorini
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Masa Al Masri
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Jan Gülink
- QubeDot GmbH, Wilhelmsgarten 3, 38100, Brunswick, Germany
| | - Heiko Brüning
- QubeDot GmbH, Wilhelmsgarten 3, 38100, Brunswick, Germany
| | - Leon Kasperek
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Monika Szepes
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Mine Bakar
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Ina Gruh
- Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, 30625, Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany
| | - Maria Leilani Torres-Mapa
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz University, 30167, Hannover, Germany.
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), 30625, Hannover, Germany.
| |
Collapse
|
7
|
Gewiess J, Eglauf J, Soubrier A, Grad S, Alini M, Peroglio M, Ma J. The influence of intervertebral disc overloading on nociceptor calcium flickering. JOR Spine 2023; 6:e1267. [PMID: 37780827 PMCID: PMC10540821 DOI: 10.1002/jsp2.1267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/07/2023] [Accepted: 05/28/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Mechanical overloading can trigger a degenerative-like cascade in an organ culture of intervertebral disc (IVD). Whether the overloaded IVD can influence the activation of nociceptors (i.e., the damage sensing neurons) remains unknown. The study aims to investigate the influence of overloaded IVD conditioned medium (CM) on the activation of nociceptors. Methods In the static loading regime, force-controlled loading of 0.2 MPa for 20 h/day representing "long-term sitting and standing" was compared with a displacement-controlled loading maintaining original IVD height. In the dynamic loading regime, high-frequency-intensity loading representing degenerative "wear and tear" was compared with a lower-frequency-intensity loading. CM of differently loaded IVDs were collected to stimulate the primary bovine dorsal root ganglion (DRG) cultures. Calcium imaging (Fluo-4) and calcitonin gene-related peptide (CGRP) immunofluorescent labeling were jointly used to record the calcium flickering in CGRP(+) nociceptors. Results Force-controlled loading led to a higher IVD cell death compared to displacement-controlled loading. Both static and dynamic overloading (force-controlled and high-frequency-intensity loadings) elevated the frequency of calcium flickering in the subsurface space of CGRP(+) nociceptors compared to their mild loading counterparts. Conclusion In the organ culture system, IVD overloading mediated an altered IVD-nociceptor communication suggesting a biological mechanism associated with discogenic pain.
Collapse
Affiliation(s)
- Jan Gewiess
- AO Research Institute, AO FoundationDavosSwitzerland
- Department of Orthopaedic Surgery and TraumatologyInselspital, Bern University Hospital, University of BernBernSwitzerland
| | - Janick Eglauf
- AO Research Institute, AO FoundationDavosSwitzerland
| | | | - Sibylle Grad
- AO Research Institute, AO FoundationDavosSwitzerland
| | - Mauro Alini
- AO Research Institute, AO FoundationDavosSwitzerland
| | | | - Junxuan Ma
- AO Research Institute, AO FoundationDavosSwitzerland
| |
Collapse
|
8
|
Heinson YW, Han JL, Entcheva E. OptoDyCE-plate as an affordable high throughput imager for all optical cardiac electrophysiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555447. [PMID: 37693544 PMCID: PMC10491208 DOI: 10.1101/2023.08.29.555447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
We present a simple low-cost system for comprehensive functional characterization of cardiac function under spontaneous and paced conditions, in standard 96 and 384-well plates. This full-plate actuator/imager, OptoDyCE-plate, uses optogenetic stimulation and optical readouts of voltage and calcium from all wells in parallel. The system is validated with syncytia of human induced pluripotent stem cell derived cardiomyocytes, iPSC-CMs, grown as monolayers, or in quasi-3D isotropic and anisotropic constructs using electrospun matrices, in 96 and 394-well format. Genetic modifications, e.g. interference CRISPR (CRISPRi), and nine compounds of acute and chronic action were tested, including five histone deacetylase inhibitors (HDACis). Their effects on voltage and calcium were compared across growth conditions and pacing rates. We also demonstrated deployment of optogenetic cell spheroids for point pacing to study conduction in 96-well format, and the use of temporal multiplexing to register voltage and calcium simultaneously on a single camera in this stand-alone platform. Opto-DyCE-plate showed excellent performance even in the small samples in 384-well plates, in the various configurations. Anisotropic structured constructs may provide some benefits in drug testing, although drug responses were consistent across tested configurations. Differential voltage vs. calcium responses were seen for some drugs, especially for non-traditional modulators of cardiac function, e.g. HDACi, and pacing rate was a powerful modulator of drug response, highlighting the need for comprehensive multiparametric assessment, as offered by OptoDyCE-plate. Increasing throughput and speed and reducing cost of screening can help stratify potential compounds early in the drug development process and accelerate the development of safer drugs.
Collapse
Affiliation(s)
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington, DC 20037
| |
Collapse
|
9
|
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
|
10
|
Formozov A, Dieter A, Wiegert JS. A flexible and versatile system for multi-color fiber photometry and optogenetic manipulation. CELL REPORTS METHODS 2023; 3:100418. [PMID: 37056369 PMCID: PMC10088095 DOI: 10.1016/j.crmeth.2023.100418] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/09/2023]
Abstract
Here, we present simultaneous fiber photometry recordings and optogenetic stimulation based on a multimode fused fiber coupler for both light delivery and collection without the need for dichroic beam splitters. In combination with a multi-color light source and appropriate optical filters, our approach offers remarkable flexibility in experimental design and facilitates the exploration of new molecular tools in vivo at minimal cost. We demonstrate straightforward re-configuration of the setup to operate with green, red, and near-infrared calcium indicators with or without simultaneous optogenetic stimulation and further explore the multi-color photometry capabilities of the system. The ease of assembly, operation, characterization, and customization of this platform holds the potential to foster the development of experimental strategies for multi-color fused fiber photometry combined with optogenetics far beyond its current state.
Collapse
Affiliation(s)
- Andrey Formozov
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Alexander Dieter
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - J. Simon Wiegert
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Neurophysiology, MCTN, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
11
|
Li Z, Zheng Y, Diao X, Li R, Sun N, Xu Y, Li X, Duan S, Gong W, Si K. Robust and adjustable dynamic scattering compensation for high-precision deep tissue optogenetics. Commun Biol 2023; 6:128. [PMID: 36721006 PMCID: PMC9889738 DOI: 10.1038/s42003-023-04487-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
The development of high-precision optogenetics in deep tissue is limited due to the strong optical scattering induced by biological tissue. Although various wavefront shaping techniques have been developed to compensate the scattering, it is still a challenge to non-invasively characterize the dynamic scattered optical wavefront inside the living tissue. Here, we present a non-invasive scattering compensation system with fast multidither coherent optical adaptive technique (fCOAT), which allows the rapid wavefront correction and stable focusing in dynamic scattering medium. We achieve subcellular-resolution focusing through 500-μm-thickness brain slices, or even three pieces overlapped mouse skulls after just one iteration with a 589 nm CW laser. Further, focusing through dynamic scattering medium such as live rat ear is also successfully achieved. The formed focus can maintain longer than 60 s, which satisfies the requirements of stable optogenetics manipulation. Moreover, the focus size is adjustable from subcellular level to tens of microns to freely match the various manipulation targets. With the specially designed fCOAT system, we successfully achieve single-cellular optogenetic manipulation through the brain tissue, with a stimulation efficiency enhancement up to 300% compared with that of the speckle.
Collapse
Affiliation(s)
- Zhenghan Li
- grid.13402.340000 0004 1759 700XState Key Laboratory of Modern Optical Instrumentation, Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XCollege of Optical Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yameng Zheng
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Xintong Diao
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Rongrong Li
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Ning Sun
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Yongxian Xu
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Xiaoming Li
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Shumin Duan
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Wei Gong
- grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
| | - Ke Si
- grid.13402.340000 0004 1759 700XState Key Laboratory of Modern Optical Instrumentation, Department of Psychiatry of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XCollege of Optical Science and Engineering, Zhejiang University, Hangzhou, China ,grid.13402.340000 0004 1759 700XLiangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China ,grid.13402.340000 0004 1759 700XIntelligent Optics & Photonics Research Center, Jiaxing Research Institute, Zhejiang University, Jiaxing, Zhejiang China
| |
Collapse
|
12
|
Kierzek M, Deal PE, Miller EW, Mukherjee S, Wachten D, Baumann A, Kaupp UB, Strünker T, Brenker C. Simultaneous recording of multiple cellular signaling events by frequency- and spectrally-tuned multiplexing of fluorescent probes. eLife 2021; 10:e63129. [PMID: 34859780 PMCID: PMC8700268 DOI: 10.7554/elife.63129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/01/2021] [Indexed: 12/21/2022] Open
Abstract
Fluorescent probes that change their spectral properties upon binding to small biomolecules, ions, or changes in the membrane potential (Vm) are invaluable tools to study cellular signaling pathways. Here, we introduce a novel technique for simultaneous recording of multiple probes at millisecond time resolution: frequency- and spectrally-tuned multiplexing (FASTM). Different from present multiplexing approaches, FASTM uses phase-sensitive signal detection, which renders various combinations of common probes for Vm and ions accessible for multiplexing. Using kinetic stopped-flow fluorimetry, we show that FASTM allows simultaneous recording of rapid changes in Ca2+, pH, Na+, and Vm with high sensitivity and minimal crosstalk. FASTM is also suited for multiplexing using single-cell microscopy and genetically encoded FRET biosensors. Moreover, FASTM is compatible with optochemical tools to study signaling using light. Finally, we show that the exceptional time resolution of FASTM also allows resolving rapid chemical reactions. Altogether, FASTM opens new opportunities for interrogating cellular signaling.
Collapse
Affiliation(s)
- Michelina Kierzek
- Centre of Reproductive Medicine and Andrology, University of MünsterMünsterGermany
- CiM-IMPRS Graduate School, University of MünsterMünsterGermany
| | - Parker E Deal
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
| | - Evan W Miller
- Department of Chemistry, University of California, BerkeleyBerkeleyUnited States
- Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Shatanik Mukherjee
- Molecular Sensory Systems, Center of Advanced European Studies and ResearchBonnGermany
| | - Dagmar Wachten
- Institute of Innate Immunity, Department of Biophysical Imaging, Medical Faculty, University of BonnBonnGermany
| | - Arnd Baumann
- Institute of Biological Information Processing (IBI-1), Research Center JülichJülichGermany
| | - U Benjamin Kaupp
- Life & Medical Sciences Institute (LIMES), University of BonnBonnGermany
| | - Timo Strünker
- Centre of Reproductive Medicine and Andrology, University of MünsterMünsterGermany
- Cells in Motion Interfaculty Centre, University of MünsterMünsterGermany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University of MünsterMünsterGermany
| |
Collapse
|
13
|
Penzkofer A, Silapetere A, Hegemann P. Photocycle dynamics of the Archaerhodopsin 3 based fluorescent voltage sensor Archon2. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2021; 225:112331. [PMID: 34688164 DOI: 10.1016/j.jphotobiol.2021.112331] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/21/2021] [Accepted: 10/05/2021] [Indexed: 11/28/2022]
Abstract
The retinal photocycle dynamics of the fluorescent voltage sensor Archon2 in pH 8 Tris buffer was studied. Archon2 is a mutant of Archaerhodopsin 3 (Arch) from Halorubrum sodomense obtained by a robotic multidimensional directed evolution approach (Archon2 = Arch T56P-P60S-T80P-D95H-T99S-T116I-F161V-T183I-L197I-A225C). The samples were photo-excited to the first absorption band of the protonated retinal Schiff base (PRSB) Ret_586 (absorption maximum at λmax = 586 nm, excitation wavelengths λexc = 590 nm and 632.8 nm). The photocycle dynamics were studied by recording absorption spectra during light exposure and after light exposure. Ret_586 photoisomerized to Ret_535 (main component) and Ret_485 (minor component). Ret_535 backward photoisomerized to Ret_586 in light-adapted state (named Ret_586la) and partly deprotonated to neutral retinal Schiff base (RSB) Ret_372 in light adapted state (named Ret_372la, same isomer form as Ret_535). After excitation light switch-off Ret_372la recovered to Ret_372 in dark-adapted state (Ret_372da) which slowly re-protonated to Ret_535, and Ret_535 slowly isomerized back to Ret_586 in dark-adapted state (Ret_586da). Photocycle schemes and reaction coordinate diagrams are developed and photocycle parameters are determined.
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
|
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
|
In Vitro Model to Investigate Communication between Dorsal Root Ganglion and Spinal Cord Glia. Int J Mol Sci 2021; 22:ijms22189725. [PMID: 34575886 PMCID: PMC8470479 DOI: 10.3390/ijms22189725] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/30/2022] Open
Abstract
Chronic discogenic back pain is associated with increased inflammatory cytokine levels that can influence the proximal peripheral nervous system, namely the dorsal root ganglion (DRG). However, transition to chronic pain is widely thought to involve glial activation in the spinal cord. In this study, an in vitro model was used to evaluate the communication between DRG and spinal cord glia. Primary neonatal rat DRG cells were treated with/without inflammatory cytokines (TNF-α, IL-1β, and IL-6). The conditioned media were collected at two time points (12 and 24 h) and applied to spinal cord mixed glial culture (MGC) for 24 h. Adult bovine DRG and spinal cord cell cultures were also tested, as an alternative large animal model, and results were compared with the neonatal rat findings. Compared with untreated DRG-conditioned medium, the second cytokine-treated DRG-conditioned medium (following medium change, thus containing solely DRG-derived molecules) elevated CD11b expression and calcium signal in neonatal rat microglia and enhanced Iba1 expression in adult bovine microglia. Cytokine treatment induced a DRG-mediated microgliosis. The described in vitro model allows the use of cells from large species and may represent an alternative to animal pain models (3R principles).
Collapse
|
16
|
Vila OF, Chavez M, Ma SP, Yeager K, Zholudeva LV, Colón-Mercado JM, Qu Y, Nash TR, Lai C, Feliciano CM, Carter M, Kamm RD, Judge LM, Conklin BR, Ward ME, McDevitt TC, Vunjak-Novakovic G. Bioengineered optogenetic model of human neuromuscular junction. Biomaterials 2021; 276:121033. [PMID: 34403849 DOI: 10.1016/j.biomaterials.2021.121033] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 12/28/2022]
Abstract
Functional human tissues engineered from patient-specific induced pluripotent stem cells (hiPSCs) hold great promise for investigating the progression, mechanisms, and treatment of musculoskeletal diseases in a controlled and systematic manner. For example, bioengineered models of innervated human skeletal muscle could be used to identify novel therapeutic targets and treatments for patients with complex central and peripheral nervous system disorders. There is a need to develop standardized and objective quantitative methods for engineering and using these complex tissues, in order increase their robustness, reproducibility, and predictiveness across users. Here we describe a standardized method for engineering an isogenic, patient specific human neuromuscular junction (NMJ) that allows for automated quantification of NMJ function to diagnose disease using a small sample of blood serum and evaluate new therapeutic modalities. By combining tissue engineering, optogenetics, microfabrication, optoelectronics and video processing, we created a novel platform for the precise investigation of the development and degeneration of human NMJ. We demonstrate the utility of this platform for the detection and diagnosis of myasthenia gravis, an antibody-mediated autoimmune disease that disrupts the NMJ function.
Collapse
Affiliation(s)
- Olaia F Vila
- Columbia University, 622 W 168th St, New York, NY, 10032, USA; Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA.
| | - Miguel Chavez
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Stephen P Ma
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Keith Yeager
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | | | | | - Yihuai Qu
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Trevor R Nash
- Columbia University, 622 W 168th St, New York, NY, 10032, USA
| | - Carmen Lai
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Carissa M Feliciano
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA; Department of Pediatrics, UCSF, 550 16th St, Floor 5, San Francisco, CA, 94143, USA
| | - Matthew Carter
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Roger D Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge MA, 02139, USA
| | - Luke M Judge
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA; Department of Pediatrics, UCSF, 550 16th St, Floor 5, San Francisco, CA, 94143, USA
| | - Bruce R Conklin
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Todd C McDevitt
- Gladstone Institutes, 1650 Owens St, San Francisco, CA, 94158, USA
| | | |
Collapse
|
17
|
Zhou Y, Liu E, Müller H, Cui B. Optical Electrophysiology: Toward the Goal of Label-Free Voltage Imaging. J Am Chem Soc 2021; 143:10482-10499. [PMID: 34191488 PMCID: PMC8514153 DOI: 10.1021/jacs.1c02960] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Measuring and monitoring the electrical signals transmitted between neurons is key to understanding the communication between neurons that underlies human perception, information processing, and decision-making. While electrode-based electrophysiology has been the gold standard, optical electrophysiology has opened up a new area in the past decade. Voltage-dependent fluorescent reporters enable voltage imaging with high spatial resolution and flexibility to choose recording locations. However, they exhibit photobleaching as well as phototoxicity and may perturb the physiology of the cell. Label-free optical electrophysiology seeks to overcome these hurdles by detecting electrical activities optically, without the incorporation of exogenous fluorophores in cells. For example, electrochromic optical recording detects neuroelectrical signals via a voltage-dependent color change of extracellular materials, and interferometric optical recording monitors membrane deformations that accompany electrical activities. Label-free optical electrophysiology, however, is in an early stage, and often has limited sensitivity and temporal resolution. In this Perspective, we review the recent progress to overcome these hurdles. We hope this Perspective will inspire developments of label-free optical electrophysiology techniques with high recording sensitivity and temporal resolution in the near future.
Collapse
Affiliation(s)
- Yuecheng Zhou
- Department of Chemistry, Stanford University, S285 ChEM-H/Wu Tsai Neuroscience Research Complex, Stanford, California 94305, United States
| | - Erica Liu
- Department of Chemistry, Stanford University, S285 ChEM-H/Wu Tsai Neuroscience Research Complex, Stanford, California 94305, United States
| | - Holger Müller
- Department of Physics, University of California, 366 LeConte Hall, Berkeley, California 94720, United States
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, S285 ChEM-H/Wu Tsai Neuroscience Research Complex, Stanford, California 94305, United States
| |
Collapse
|
18
|
Puppo F, Pré D, Bang AG, Silva GA. Super-Selective Reconstruction of Causal and Direct Connectivity With Application to in vitro iPSC Neuronal Networks. Front Neurosci 2021; 15:647877. [PMID: 34335152 PMCID: PMC8323822 DOI: 10.3389/fnins.2021.647877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/31/2021] [Indexed: 12/22/2022] Open
Abstract
Despite advancements in the development of cell-based in-vitro neuronal network models, the lack of appropriate computational tools limits their analyses. Methods aimed at deciphering the effective connections between neurons from extracellular spike recordings would increase utility of in vitro local neural circuits, especially for studies of human neural development and disease based on induced pluripotent stem cells (hiPSC). Current techniques allow statistical inference of functional couplings in the network but are fundamentally unable to correctly identify indirect and apparent connections between neurons, generating redundant maps with limited ability to model the causal dynamics of the network. In this paper, we describe a novel mathematically rigorous, model-free method to map effective-direct and causal-connectivity of neuronal networks from multi-electrode array data. The inference algorithm uses a combination of statistical and deterministic indicators which, first, enables identification of all existing functional links in the network and then reconstructs the directed and causal connection diagram via a super-selective rule enabling highly accurate classification of direct, indirect, and apparent links. Our method can be generally applied to the functional characterization of any in vitro neuronal networks. Here, we show that, given its accuracy, it can offer important insights into the functional development of in vitro hiPSC-derived neuronal cultures.
Collapse
Affiliation(s)
- Francesca Puppo
- BioCircuits Institute and Center for Engineered Natural Intelligence, University of California, San Diego, La Jolla, CA, United States
| | - Deborah Pré
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Anne G. Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Gabriel A. Silva
- BioCircuits Institute, Center for Engineered Natural Intelligence, Department of Bioengineering, Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
19
|
Zeng Y, Ferdous ZI, Zhang W, Xu M, Yu A, Patel D, Post V, Guo X, Berdichevsky Y, Yan Z. Understanding the Impact of Neural Variations and Random Connections on Inference. Front Comput Neurosci 2021; 15:612937. [PMID: 34163343 PMCID: PMC8215547 DOI: 10.3389/fncom.2021.612937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/19/2021] [Indexed: 11/19/2022] Open
Abstract
Recent research suggests that in vitro neural networks created from dissociated neurons may be used for computing and performing machine learning tasks. To develop a better artificial intelligent system, a hybrid bio-silicon computer is worth exploring, but its performance is still inferior to that of a silicon-based computer. One reason may be that a living neural network has many intrinsic properties, such as random network connectivity, high network sparsity, and large neural and synaptic variability. These properties may lead to new design considerations, and existing algorithms need to be adjusted for living neural network implementation. This work investigates the impact of neural variations and random connections on inference with learning algorithms. A two-layer hybrid bio-silicon platform is constructed and a five-step design method is proposed for the fast development of living neural network algorithms. Neural variations and dynamics are verified by fitting model parameters with biological experimental results. Random connections are generated under different connection probabilities to vary network sparsity. A multi-layer perceptron algorithm is tested with biological constraints on the MNIST dataset. The results show that a reasonable inference accuracy can be achieved despite the presence of neural variations and random network connections. A new adaptive pre-processing technique is proposed to ensure good learning accuracy with different living neural network sparsity.
Collapse
Affiliation(s)
- Yuan Zeng
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Zubayer Ibne Ferdous
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Weixiang Zhang
- Electrical and Computer Engineering Department, Beihang University, Beijing, China
| | - Mufan Xu
- Electrical and Computer Engineering Department, Beihang University, Beijing, China
| | - Anlan Yu
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Drew Patel
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Valentin Post
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Xiaochen Guo
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| | - Yevgeny Berdichevsky
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States.,Bioengineering Department, Lehigh University, Bethlehem, PA, United States
| | - Zhiyuan Yan
- Electrical and Computer Engineering Department, Lehigh University, Bethlehem, PA, United States
| |
Collapse
|
20
|
Heeger P, Harre J, Warnecke A, Mueller D, Kalies S, Heisterkamp A. Probing interneuronal cell communication via optogenetic stimulation. TRANSLATIONAL BIOPHOTONICS 2021. [DOI: 10.1002/tbio.202100002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Patrick Heeger
- Leibniz University Hannover, Institute of Quantum Optics, AG Heisterkamp Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- Cluster of Excellence Hearing4all Hannover Germany
| | - Jennifer Harre
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- Cluster of Excellence Hearing4all Hannover Germany
- Department of Otorhinolaryngology, Head and Neck Surgery Hannover Medical School Hannover Germany
| | - Athanasia Warnecke
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- Cluster of Excellence Hearing4all Hannover Germany
- Department of Otorhinolaryngology, Head and Neck Surgery Hannover Medical School Hannover Germany
| | - Dominik Mueller
- Leibniz University Hannover, Institute of Quantum Optics, AG Heisterkamp Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- REBIRTH Research Center for Translational Regenerative Medicine Hannover Germany
| | - Stefan Kalies
- Leibniz University Hannover, Institute of Quantum Optics, AG Heisterkamp Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- REBIRTH Research Center for Translational Regenerative Medicine Hannover Germany
| | - Alexander Heisterkamp
- Leibniz University Hannover, Institute of Quantum Optics, AG Heisterkamp Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development Hannover Germany
- Cluster of Excellence Hearing4all Hannover Germany
- REBIRTH Research Center for Translational Regenerative Medicine Hannover Germany
| |
Collapse
|
21
|
Walker AS, Raliski BK, Karbasi K, Zhang P, Sanders K, Miller EW. Optical Spike Detection and Connectivity Analysis With a Far-Red Voltage-Sensitive Fluorophore Reveals Changes to Network Connectivity in Development and Disease. Front Neurosci 2021; 15:643859. [PMID: 34054405 PMCID: PMC8155641 DOI: 10.3389/fnins.2021.643859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to optically record dynamics of neuronal membrane potential promises to revolutionize our understanding of neurobiology. In this study, we show that the far-red voltage sensitive fluorophore, Berkeley Red Sensor of Transmembrane potential-1, or BeRST 1, can be used to monitor neuronal membrane potential changes across dozens of neurons at a sampling rate of 500 Hz. Notably, voltage imaging with BeRST 1 can be implemented with affordable, commercially available illumination sources, optics, and detectors. BeRST 1 is well-tolerated in cultures of rat hippocampal neurons and provides exceptional optical recording fidelity, as judged by dual fluorescence imaging and patch-clamp electrophysiology. We developed a semi-automated spike-picking program to reduce user bias when calling action potentials and used this in conjunction with BeRST 1 to develop an optical spike and connectivity analysis (OSCA) for high-throughput dissection of neuronal activity dynamics. The high temporal resolution of BeRST 1 enables dissection of firing rate changes in response to acute, pharmacological interventions with commonly used inhibitors like gabazine and picrotoxin. Over longer periods of time, BeRST 1 also tracks chronic perturbations to neurons exposed to amyloid beta 1-42 (Aβ 1-42), revealing modest changes to spiking frequency but profound changes to overall network connectivity. Finally, we use OSCA to track changes in neuronal connectivity during maturation in culture, providing a functional readout of network assembly. We envision that use of BeRST 1 and OSCA described here will be of use to the broad neuroscience community.
Collapse
Affiliation(s)
- Alison S. Walker
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | - Benjamin K. Raliski
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
| | - Kaveh Karbasi
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
| | - Patrick Zhang
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
| | - Kate Sanders
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
| | - Evan W. Miller
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, United States
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
22
|
Probing Synaptic Signaling with Optogenetic Stimulation and Genetically Encoded Calcium Reporters. Methods Mol Biol 2021. [PMID: 32865742 DOI: 10.1007/978-1-0716-0830-2_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Optogenetics provides a powerful approach for investigating neuronal electrophysiology at the scale required for drug discovery applications. Probing synaptic function with high throughput using optogenetics requires robust tools that enable both precise stimulation of and facile readout of synaptic activity. Here we describe two functional assays to achieve this end: (1) a pre-synaptic calcium assay that utilizes the channelrhodopsin, CheRiff, patterned optogenetic stimulus, and the pre-synaptically targeted calcium reporter jRGECO1a to monitor pre-synaptic changes in calcium influx and (2) a synaptic transmission assay in which CheRiff and cytosolic jRGECO1a are expressed in non-overlapping sets of neurons, enabling pre-synaptic stimulation and post-synaptic readout of activity. This chapter describes the methodology and practical considerations for implementation of these two assays.
Collapse
|
23
|
Population imaging discrepancies between a genetically-encoded calcium indicator (GECI) versus a genetically-encoded voltage indicator (GEVI). Sci Rep 2021; 11:5295. [PMID: 33674659 PMCID: PMC7935943 DOI: 10.1038/s41598-021-84651-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/11/2021] [Indexed: 11/25/2022] Open
Abstract
Genetically-encoded calcium indicators (GECIs) are essential for studying brain function, while voltage indicators (GEVIs) are slowly permeating neuroscience. Fundamentally, GECI and GEVI measure different things, but both are advertised as reporters of “neuronal activity”. We quantified the similarities and differences between calcium and voltage imaging modalities, in the context of population activity (without single-cell resolution) in brain slices. GECI optical signals showed 8–20 times better SNR than GEVI signals, but GECI signals attenuated more with distance from the stimulation site. We show the exact temporal discrepancy between calcium and voltage imaging modalities, and discuss the misleading aspects of GECI imaging. For example, population voltage signals already repolarized to the baseline (~ disappeared), while the GECI signals were still near maximum. The region-to-region propagation latencies, easily captured by GEVI imaging, are blurred in GECI imaging. Temporal summation of GECI signals is highly exaggerated, causing uniform voltage events produced by neuronal populations to appear with highly variable amplitudes in GECI population traces. Relative signal amplitudes in GECI recordings are thus misleading. In simultaneous recordings from multiple sites, the compound EPSP signals in cortical neuropil (population signals) are less distorted by GEVIs than by GECIs.
Collapse
|
24
|
Roberts R, Authier S, Mellon RD, Morton M, Suzuki I, Tjalkens RB, Valentin JP, Pierson JB. Can We Panelize Seizure? Toxicol Sci 2021; 179:3-13. [PMID: 33165543 DOI: 10.1093/toxsci/kfaa167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Seizure liability remains a significant cause of attrition in drug discovery and development, leading to loss of competitiveness, delays, and increased costs. Current detection methods rely on observations made in in vivo studies intended to support clinical trials, such as tremors or other abnormal movements. These signs could be missed or misinterpreted; thus, definitive confirmation of drug-induced seizure requires a follow-up electroencephalogram study. There has been progress in in vivo detection of seizure using automated video systems that record and analyze animal movements. Nonetheless, it would be preferable to have earlier prediction of seizurogenic risk that could be used to eliminate liabilities early in discovery while there are options for medicinal chemists making potential new drugs. Attrition due to cardiac adverse events has benefited from routine early screening; could we reduce attrition due to seizure using a similar approach? Specifically, microelectrode arrays could be used to detect potential seizurogenic signals in stem-cell-derived neurons. In addition, there is clear evidence implicating neuronal voltage-gated and ligand-gated ion channels, GPCRs and transporters in seizure. Interactions with surrounding glial cells during states of stress or inflammation can also modulate ion channel function in neurons, adding to the challenge of seizure prediction. It is timely to evaluate the opportunity to develop an in vitro assessment of seizure linked to a panel of ion channel assays that predict seizure, with the aim of influencing structure-activity relationship at the design stage and eliminating compounds predicted to be associated with pro-seizurogenic state.
Collapse
Affiliation(s)
- Ruth Roberts
- ApconiX, Alderley Park, SK10 4TG, UK.,University of Birmingham, B15 2SD, UK
| | | | - R Daniel Mellon
- US Food and Drug Administration, Silver Spring, Maryland 20993
| | | | - Ikuro Suzuki
- Tohoku Institute of Technology, Sendai, 980-8577, Japan
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Early Solutions, Development Science, Investigative Toxicology, Chemin du Foriest, B-1420, Braine-l'Alleud, Belgium
| | - Jennifer B Pierson
- Health and Environmental Sciences Institute, Washington, District of Columbia 20005
| |
Collapse
|
25
|
Abstract
Inherited cardiac arrhythmias contribute substantially to sudden cardiac death in the young. The underlying pathophysiology remains incompletely understood because of the lack of representative study models and the labour-intensive nature of electrophysiological patch clamp experiments. Whereas patch clamp is still considered the gold standard for investigating electrical properties in a cell, optical mapping of voltage and calcium transients has paved the way for high-throughput studies. Moreover, the development of human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) has enabled the study of patient specific cell lines capturing the full genomic background. Nevertheless, hiPSC-CMs do not fully address the complex interactions between various cell types in the heart. Studies using in vivo models, are therefore necessary. Given the analogies between the human and zebrafish cardiovascular system, zebrafish has emerged as a cost-efficient model for arrhythmogenic diseases. In this review, we describe how hiPSC-CM and zebrafish are employed as models to study primary electrical disorders. We provide an overview of the contemporary electrophysiological phenotyping tools and discuss in more depth the different strategies available for optical mapping. We consider the current advantages and disadvantages of both hiPSC-CM and zebrafish as a model and optical mapping as phenotyping tool and propose strategies for further improvement. Overall, the combination of experimental readouts at cellular (hiPSC-CM) and whole organ (zebrafish) level can raise our understanding of the complexity of inherited cardiac arrhythmia disorders to the next level.
Collapse
|
26
|
Abstract
The electromechanical function of the heart involves complex, coordinated activity over time and space. Life-threatening cardiac arrhythmias arise from asynchrony in these space-time events; therefore, therapies for prevention and treatment require fundamental understanding and the ability to visualize, perturb and control cardiac activity. Optogenetics combines optical and molecular biology (genetic) approaches for light-enabled sensing and actuation of electrical activity with unprecedented spatiotemporal resolution and parallelism. The year 2020 marks a decade of developments in cardiac optogenetics since this technology was adopted from neuroscience and applied to the heart. In this Review, we appraise a decade of advances that define near-term (immediate) translation based on all-optical electrophysiology, including high-throughput screening, cardiotoxicity testing and personalized medicine assays, and long-term (aspirational) prospects for clinical translation of cardiac optogenetics, including new optical therapies for rhythm control. The main translational opportunities and challenges for optogenetics to be fully embraced in cardiology are also discussed.
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 Archon2. Int J Mol Sci 2020; 21:ijms21186576. [PMID: 32911811 PMCID: PMC7555599 DOI: 10.3390/ijms21186576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 01/12/2023] Open
Abstract
Archon2 is a fluorescent voltage sensor derived from Archaerhodopsin 3 (Arch) of Halorubrum sodomense using robotic multidimensional directed evolution approach. Here we report absorption and emission spectroscopic studies of Archon2 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 Archon2 was studied by long-time attenuation coefficient measurements at room temperature (21 ± 1 °C) and at refrigerator temperature (3 ± 1 °C). The apparent melting temperature was determined by stepwise sample heating up and cooling down (obtained apparent melting temperature: 63 ± 3 °C). In the protein melting process protonated retinal Schiff base (PRSB) with absorption maximum at 586 nm converted to de-protonated retinal Schiff base (RSB) with absorption maximum at 380 nm. Storage of Archon2 at room temperature and refrigerator temperature caused absorption coefficient decrease because of partial protein clustering to aggregates at condensation nuclei and sedimentation. At room temperature an onset of light scattering was observed after two days because of the beginning of protein unfolding. During the period of observation (18 days at 21 °C, 22 days at 3 °C) no change of retinal isomer composition was observed indicating a high potential energy barrier of S0 ground-state isomerization.
Collapse
Affiliation(s)
- Alfons Penzkofer
- Fakultät für Physik, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-943-2107
| | - Arita Silapetere
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany; (A.S.); (P.H.)
| | - Peter Hegemann
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany; (A.S.); (P.H.)
| |
Collapse
|
28
|
Werley CA, Boccardo S, Rigamonti A, Hansson EM, Cohen AE. Multiplexed Optical Sensors in Arrayed Islands of Cells for multimodal recordings of cellular physiology. Nat Commun 2020; 11:3881. [PMID: 32753572 PMCID: PMC7403318 DOI: 10.1038/s41467-020-17607-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 07/07/2020] [Indexed: 11/29/2022] Open
Abstract
Cells typically respond to chemical or physical perturbations via complex signaling cascades which can simultaneously affect multiple physiological parameters, such as membrane voltage, calcium, pH, and redox potential. Protein-based fluorescent sensors can report many of these parameters, but spectral overlap prevents more than ~4 modalities from being recorded in parallel. Here we introduce the technique, MOSAIC, Multiplexed Optical Sensors in Arrayed Islands of Cells, where patterning of fluorescent sensor-encoding lentiviral vectors with a microarray printer enables parallel recording of multiple modalities. We demonstrate simultaneous recordings from 20 sensors in parallel in human embryonic kidney (HEK293) cells and in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), and we describe responses to metabolic and pharmacological perturbations. Together, these results show that MOSAIC can provide rich multi-modal data on complex physiological responses in multiple cell types.
Collapse
Affiliation(s)
- Christopher A Werley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Q-State Biosciences, Cambridge, MA, 02139, USA
| | - Stefano Boccardo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
- Nobel Biocare AG, Kloten, Switzerland
| | - Alessandra Rigamonti
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
| | - Emil M Hansson
- Integrated Cardio Metabolic Centre, Department of Medicine Huddinge, Karolinska Institute, Huddinge, Sweden
- Department of Stem Cell and Regenerative 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, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
29
|
Ma J, Stefanoska D, Grad S, Alini M, Peroglio M. Direct and Intervertebral Disc-Mediated Sensitization of Dorsal Root Ganglion Neurons by Hypoxia and Low pH. Neurospine 2020; 17:42-59. [PMID: 32252154 PMCID: PMC7136118 DOI: 10.14245/ns.2040052.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/28/2020] [Indexed: 12/21/2022] Open
Abstract
Objective Ischemia-related risk factors are consistently correlated with discogenic pain, but it remains unclear how the ischemia-associated hypoxia and acidosis influence the peripheral sensory nervous system, namely the dorsal root ganglion (DRG), either directly or indirectly via intervertebral disc (IVD) mediation.
Methods Bovine tail IVD organ cultures were preconditioned in different hypoxic and/or acidic conditions for 3 days to collect the conditioned medium (CM). The DRG-derived ND7/23 cells were either treated by the IVD CM or directly stimulated by hypoxic and/or acidic conditions. Neuronal sensitization was evaluated using calcium imaging (Fluo-4) after 3 days.
Results We found that direct exposure of DRG cell line to hypoxia and acidosis increased both spontaneous and bradykinin-stimulated calcium response compared to normoxia-neutral pH cultures. Hypoxia and low pH in combination showed stronger effect than either parameter on its own. Indirect exposure of DRG to hypoxia-acidosis-stressed IVD CM also increased spontaneous and bradykinin-stimulated response, but to a lower extent than direct exposure. The impact of direct hypoxia and acidosis on DRG was validated in a primary sheep DRG cell culture, showing the same trend.
Conclusion Our data suggest that targeting hypoxia and acidosis stresses both in IVD and DRG could be a relevant objective in discogenic pain treatment.
Collapse
Affiliation(s)
- Junxuan Ma
- AO Research Institute Davos, Davos, Switzerland
| | | | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
30
|
Xu X, Mee T, Jia X. New era of optogenetics: from the central to peripheral nervous system. Crit Rev Biochem Mol Biol 2020; 55:1-16. [PMID: 32070147 DOI: 10.1080/10409238.2020.1726279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Optogenetics has recently gained recognition as a biological technique to control the activity of cells using light stimulation. Many studies have applied optogenetics to cell lines in the central nervous system because it has the potential to elucidate neural circuits, treat neurological diseases and promote nerve regeneration. There have been fewer studies on the application of optogenetics in the peripheral nervous system. This review introduces the basic principles and approaches of optogenetics and summarizes the physiology and mechanism of opsins and how the technology enables bidirectional control of unique cell lines with superior spatial and temporal accuracy. Further, this review explores and discusses the therapeutic potential for the development of optogenetics and its capacity to revolutionize treatment for refractory epilepsy, depression, pain, and other nervous system disorders, with a focus on neural regeneration, especially in the peripheral nervous system. Additionally, this review synthesizes the latest preclinical research on optogenetic stimulation, including studies on non-human primates, summarizes the challenges, and highlights future perspectives. The potential of optogenetic stimulation to optimize therapy for peripheral nerve injuries (PNIs) is also highlighted. Optogenetic technology has already generated exciting, preliminary evidence, supporting its role in applications to several neurological diseases, including PNIs.
Collapse
Affiliation(s)
- Xiang Xu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas Mee
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
31
|
Penzkofer A, Silapetere A, Hegemann P. Photocycle Dynamics of the Archaerhodopsin 3 Based Fluorescent Voltage Sensor QuasAr1. Int J Mol Sci 2019; 21:ijms21010160. [PMID: 31881701 PMCID: PMC6982170 DOI: 10.3390/ijms21010160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 11/16/2022] Open
Abstract
The retinal photocycle dynamics of the fluorescent voltage sensor QuasAr1 (Archaerhodopsin 3 P60S-T80S-D95H-D106H-F161V mutant from Halorubrum sodomense) in pH 8 Tris buffer was studied. The samples were photoexcited to the first absorption band of the protonated retinal Schiff base (PRSB) Ret_580 (absorption maximum at λmax ≈ 580 nm), and the retinal Schiff base photoisomerization and protonation state changes were followed by absorption spectra recordings during light exposure and after light exposure. Ret_580 turned out to be composed of two protonated retinal Schiff base isomers, namely Ret_580I and Ret_580II. Photoexcitation of Ret_580I resulted in barrier-involved isomerization to Ret_540 (quantum yield ≈ 0.056) and subsequent retinal proton release leading to Ret_410 deprotonated retinal Schiff base (RSB). In the dark, Ret_410 partially recovered to Ret_580I and partially stabilized to irreversible Ret_400 due to apoprotein restructuring (Ret_410 lifetime ≈ 2 h). Photoexcitation of Ret_580II resulted in barrier-involved isomerization to Ret_640 (quantum yield ≈ 0.00135) and subsequent deprotonation to Ret_370 (RSB). In the dark, Ret_370 partially recovered to Ret_580II and partially stabilized to irreversible Ret_350 due to apoprotein restructuring (Ret_370 lifetime ≈ 10 h). Photocycle schemes and reaction coordinate diagrams for Ret_580I and Ret_580II were developed and photocyle parameters were determined.
Collapse
Affiliation(s)
- Alfons Penzkofer
- Fakultät für Physik, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-943-2107
| | - Arita Silapetere
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany; (A.S.); (P.H.)
| | - Peter Hegemann
- Experimentelle Biophysik, Institut für Biologie, Humboldt Universität zu Berlin, Invalidenstraße 42, D-10115 Berlin, Germany; (A.S.); (P.H.)
| |
Collapse
|
32
|
Coussens NP, Sittampalam GS, Jonson SG, Hall MD, Gorby HE, Tamiz AP, McManus OB, Felder CC, Rasmussen K. The Opioid Crisis and the Future of Addiction and Pain Therapeutics. J Pharmacol Exp Ther 2019; 371:396-408. [PMID: 31481516 DOI: 10.1124/jpet.119.259408] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/29/2019] [Indexed: 12/26/2022] Open
Abstract
Opioid misuse and addiction are a public health crisis resulting in debilitation, deaths, and significant social and economic impact. Curbing this crisis requires collaboration among academic, government, and industrial partners toward the development of effective nonaddictive pain medications, interventions for opioid overdose, and addiction treatments. A 2-day meeting, The Opioid Crisis and the Future of Addiction and Pain Therapeutics: Opportunities, Tools, and Technologies Symposium, was held at the National Institutes of Health (NIH) to address these concerns and to chart a collaborative path forward. The meeting was supported by the NIH Helping to End Addiction Long-TermSM (HEAL) Initiative, an aggressive, trans-agency effort to speed scientific solutions to stem the national opioid crisis. The event was unique in bringing together two research disciplines, addiction and pain, in order to create a forum for crosscommunication and collaboration. The output from the symposium will be considered by the HEAL Initiative; this article summarizes the scientific presentations and key takeaways. Improved understanding of the etiology of acute and chronic pain will enable the discovery of novel targets and regulatable pain circuits for safe and effective therapeutics, as well as relevant biomarkers to ensure adequate testing in clinical trials. Applications of improved technologies including reagents, assays, model systems, and validated probe compounds will likely increase the delivery of testable hypotheses and therapeutics to enable better health outcomes for patients. The symposium goals were achieved by increasing interdisciplinary collaboration to accelerate solutions for this pressing public health challenge and provide a framework for focused efforts within the research community. SIGNIFICANCE STATEMENT: This article summarizes key messages and discussions resulting from a 2-day symposium focused on challenges and opportunities in developing addiction- and pain-related medications. Speakers and attendees came from 40 states in the United States and 15 countries, bringing perspectives from academia, industry, government, and healthcare by researchers, clinicians, regulatory experts, and patient advocates.
Collapse
Affiliation(s)
- Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - G Sitta Sittampalam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Samantha G Jonson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Heather E Gorby
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Amir P Tamiz
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Owen B McManus
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Christian C Felder
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Kurt Rasmussen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| |
Collapse
|
33
|
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
|