1
|
Leong LM, Storace DA. Imaging different cell populations in the mouse olfactory bulb using the genetically encoded voltage indicator ArcLight. NEUROPHOTONICS 2024; 11:033402. [PMID: 38288247 PMCID: PMC10823906 DOI: 10.1117/1.nph.11.3.033402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/31/2024]
Abstract
Genetically encoded voltage indicators (GEVIs) are protein-based optical sensors that allow for measurements from genetically defined populations of neurons. Although in vivo imaging in the mammalian brain with early generation GEVIs was difficult due to poor membrane expression and low signal-to-noise ratio, newer and more sensitive GEVIs have begun to make them useful for answering fundamental questions in neuroscience. We discuss principles of imaging using GEVIs and genetically encoded calcium indicators, both useful tools for in vivo imaging of neuronal activity, and review some of the recent mechanistic advances that have led to GEVI improvements. We provide an overview of the mouse olfactory bulb (OB) and discuss recent studies using the GEVI ArcLight to study different cell types within the bulb using both widefield and two-photon microscopy. Specific emphasis is placed on using GEVIs to begin to study the principles of concentration coding in the OB, how to interpret the optical signals from population measurements in the in vivo brain, and future developments that will push the field forward.
Collapse
Affiliation(s)
- Lee Min Leong
- Florida State University, Department of Biological Science, Tallahassee, Florida, United States
| | - Douglas A. Storace
- Florida State University, Department of Biological Science, Tallahassee, Florida, United States
- Florida State University, Program in Neuroscience, Tallahassee, Florida, United States
- Florida State University, Institute of Molecular Biophysics, Tallahassee, Florida, United States
| |
Collapse
|
2
|
Bowman AJ, Huang C, Schnitzer MJ, Kasevich MA. Wide-field fluorescence lifetime imaging of neuron spiking and subthreshold activity in vivo. Science 2023; 380:1270-1275. [PMID: 37347862 PMCID: PMC10361454 DOI: 10.1126/science.adf9725] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/16/2023] [Indexed: 06/24/2023]
Abstract
The development of voltage-sensitive fluorescent probes suggests fluorescence lifetime as a promising readout for electrical activity in biological systems. Existing approaches fail to achieve the speed and sensitivity required for voltage imaging in neuroscience applications. We demonstrated that wide-field electro-optic fluorescence lifetime imaging microscopy (EO-FLIM) allows lifetime imaging at kilohertz frame-acquisition rates, spatially resolving action potential propagation and subthreshold neural activity in live adult Drosophila. Lifetime resolutions of <5 picoseconds at 1 kilohertz were achieved for single-cell voltage recordings. Lifetime readout is limited by photon shot noise, and the method provides strong rejection of motion artifacts and technical noise sources. Recordings revealed local transmembrane depolarizations, two types of spikes with distinct fluorescence lifetimes, and phase locking of spikes to an external mechanical stimulus.
Collapse
Affiliation(s)
- Adam J Bowman
- Physics Department, Stanford University, Stanford, CA 94305, USA
| | - Cheng Huang
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Mark J Schnitzer
- James H. Clark Center, Stanford University, Stanford, CA 94305, USA
- CNC Program, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Mark A Kasevich
- Physics Department, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Silic MR, Zhang G. Bioelectricity in Developmental Patterning and Size Control: Evidence and Genetically Encoded Tools in the Zebrafish Model. Cells 2023; 12:cells12081148. [PMID: 37190057 DOI: 10.3390/cells12081148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Developmental patterning is essential for regulating cellular events such as axial patterning, segmentation, tissue formation, and organ size determination during embryogenesis. Understanding the patterning mechanisms remains a central challenge and fundamental interest in developmental biology. Ion-channel-regulated bioelectric signals have emerged as a player of the patterning mechanism, which may interact with morphogens. Evidence from multiple model organisms reveals the roles of bioelectricity in embryonic development, regeneration, and cancers. The Zebrafish model is the second most used vertebrate model, next to the mouse model. The zebrafish model has great potential for elucidating the functions of bioelectricity due to many advantages such as external development, transparent early embryogenesis, and tractable genetics. Here, we review genetic evidence from zebrafish mutants with fin-size and pigment changes related to ion channels and bioelectricity. In addition, we review the cell membrane voltage reporting and chemogenetic tools that have already been used or have great potential to be implemented in zebrafish models. Finally, new perspectives and opportunities for bioelectricity research with zebrafish are discussed.
Collapse
Affiliation(s)
- Martin R Silic
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - GuangJun Zhang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
- Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, 625 Harrison Street, West Lafayette, IN 47907, USA
| |
Collapse
|
4
|
Day-Cooney J, Dalangin R, Zhong H, Mao T. Genetically encoded fluorescent sensors for imaging neuronal dynamics in vivo. J Neurochem 2023; 164:284-308. [PMID: 35285522 PMCID: PMC11322610 DOI: 10.1111/jnc.15608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
Abstract
The brain relies on many forms of dynamic activities in individual neurons, from synaptic transmission to electrical activity and intracellular signaling events. Monitoring these neuronal activities with high spatiotemporal resolution in the context of animal behavior is a necessary step to achieve a mechanistic understanding of brain function. With the rapid development and dissemination of highly optimized genetically encoded fluorescent sensors, a growing number of brain activities can now be visualized in vivo. To date, cellular calcium imaging, which has been largely used as a proxy for electrical activity, has become a mainstay in systems neuroscience. While challenges remain, voltage imaging of neural populations is now possible. In addition, it is becoming increasingly practical to image over half a dozen neurotransmitters, as well as certain intracellular signaling and metabolic activities. These new capabilities enable neuroscientists to test previously unattainable hypotheses and questions. This review summarizes recent progress in the development and delivery of genetically encoded fluorescent sensors, and highlights example applications in the context of in vivo imaging.
Collapse
Affiliation(s)
- Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Rochelin Dalangin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, California, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
5
|
Nikolaev DM, Mironov VN, Shtyrov AA, Kvashnin ID, Mereshchenko AS, Vasin AV, Panov MS, Ryazantsev MN. Fluorescence Imaging of Cell Membrane Potential: From Relative Changes to Absolute Values. Int J Mol Sci 2023; 24:2435. [PMID: 36768759 PMCID: PMC9916766 DOI: 10.3390/ijms24032435] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Membrane potential is a fundamental property of biological cells. Changes in membrane potential characterize a vast number of vital biological processes, such as the activity of neurons and cardiomyocytes, tumorogenesis, cell-cycle progression, etc. A common strategy to record membrane potential changes that occur in the process of interest is to utilize organic dyes or genetically-encoded voltage indicators with voltage-dependent fluorescence. Sensors are introduced into target cells, and alterations of fluorescence intensity are recorded with optical methods. Techniques that allow recording relative changes of membrane potential and do not take into account fluorescence alterations due to factors other than membrane voltage are already widely used in modern biological and biomedical studies. Such techniques have been reviewed previously in many works. However, in order to investigate a number of processes, especially long-term processes, the measured signal must be corrected to exclude the contribution from voltage-independent factors or even absolute values of cell membrane potential have to be evaluated. Techniques that enable such measurements are the subject of this review.
Collapse
Affiliation(s)
- Dmitrii M. Nikolaev
- Institute of Biomedical Systems and Biotechnologies, Peter the Great Saint Petersburg Polytechnic University, 29 Polytechnicheskaya str., 195251 Saint Petersburg, Russia
- Nanotechnology Research and Education Centre RAS, Saint Petersburg Academic University, 8/3 Khlopina str., 194021 Saint Petersburg, Russia
| | - Vladimir N. Mironov
- Nanotechnology Research and Education Centre RAS, Saint Petersburg Academic University, 8/3 Khlopina str., 194021 Saint Petersburg, Russia
| | - Andrey A. Shtyrov
- Institute of Biomedical Systems and Biotechnologies, Peter the Great Saint Petersburg Polytechnic University, 29 Polytechnicheskaya str., 195251 Saint Petersburg, Russia
- Nanotechnology Research and Education Centre RAS, Saint Petersburg Academic University, 8/3 Khlopina str., 194021 Saint Petersburg, Russia
| | - Iaroslav D. Kvashnin
- Nanotechnology Research and Education Centre RAS, Saint Petersburg Academic University, 8/3 Khlopina str., 194021 Saint Petersburg, Russia
| | - Andrey S. Mereshchenko
- Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii pr, 198504 Saint Petersburg, Russia
| | - Andrey V. Vasin
- Institute of Biomedical Systems and Biotechnologies, Peter the Great Saint Petersburg Polytechnic University, 29 Polytechnicheskaya str., 195251 Saint Petersburg, Russia
| | - Maxim S. Panov
- Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii pr, 198504 Saint Petersburg, Russia
- Center for Biophysical Studies, Saint Petersburg State Chemical Pharmaceutical University, 14 Professor Popov str., lit. A, 197022 Saint Petersburg, Russia
| | - Mikhail N. Ryazantsev
- Nanotechnology Research and Education Centre RAS, Saint Petersburg Academic University, 8/3 Khlopina str., 194021 Saint Petersburg, Russia
- Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii pr, 198504 Saint Petersburg, Russia
| |
Collapse
|
6
|
McCullough CM, Ramirez-Gordillo D, Hall M, Futia GL, Moran AK, Gibson EA, Restrepo D. GRINtrode: a neural implant for simultaneous two-photon imaging and extracellular electrophysiology in freely moving animals. NEUROPHOTONICS 2022; 9:045009. [PMID: 36466189 PMCID: PMC9713693 DOI: 10.1117/1.nph.9.4.045009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/01/2022] [Indexed: 06/11/2023]
Abstract
Significance In vivo imaging and electrophysiology are powerful tools to explore neuronal function that each offer unique complementary information with advantages and limitations. Capturing both data types from the same neural population in the freely moving animal would allow researchers to take advantage of the capabilities of both modalities and further understand how they relate to each other. Aim Here, we present a head-mounted neural implant suitable for in vivo two-photon imaging of neuronal activity with simultaneous extracellular electrical recording in head-fixed or fiber-coupled freely moving animals. Approach A gradient refractive index (GRIN) lens-based head-mounted neural implant with extracellular electrical recording provided by tetrodes on the periphery of the GRIN lens was chronically implanted. The design of the neural implant allows for recording from head-fixed animals, as well as freely moving animals by coupling the imaging system to a coherent imaging fiber bundle. Results We demonstrate simultaneous two-photon imaging of GCaMP and extracellular electrophysiology of neural activity in awake head-fixed and freely moving mice. Using the collected information, we perform correlation analysis to reveal positive correlation between optical and local field potential recordings. Conclusion Simultaneously recording neural activity using both optical and electrical methods provides complementary information from each modality. Designs that can provide such bi-modal recording in freely moving animals allow for the investigation of neural activity underlying a broader range of behavioral paradigms.
Collapse
Affiliation(s)
- Connor M. McCullough
- University of Colorado Anschutz Medical Campus, Department of Bioengineering, Aurora, Colorado, United States
| | - Daniel Ramirez-Gordillo
- University of Colorado Anschutz Medical Campus, Department of Neurosurgery, Aurora, Colorado, United States
| | - Michael Hall
- University of Colorado Anschutz Medical Campus, Neuroscience Machine Shop, Aurora, Colorado, United States
| | - Gregory L. Futia
- University of Colorado Anschutz Medical Campus, Department of Bioengineering, Aurora, Colorado, United States
| | - Andrew K. Moran
- University of Colorado Anschutz Medical Campus, Department of Cell and Development Biology, Aurora, Colorado, United States
| | - Emily A. Gibson
- University of Colorado Anschutz Medical Campus, Department of Bioengineering, Aurora, Colorado, United States
| | - Diego Restrepo
- University of Colorado Anschutz Medical Campus, Department of Cell and Development Biology, Aurora, Colorado, United States
| |
Collapse
|
7
|
Howe CL, Quicke P, Song P, Verinaz-Jadan H, Dragotti PL, Foust AJ. Comparing synthetic refocusing to deconvolution for the extraction of neuronal calcium transients from light fields. NEUROPHOTONICS 2022; 9:041404. [PMID: 35445141 PMCID: PMC8922050 DOI: 10.1117/1.nph.9.4.041404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Significance: Light-field microscopy (LFM) enables fast, light-efficient, volumetric imaging of neuronal activity with calcium indicators. Calcium transients differ in temporal signal-to-noise ratio (tSNR) and spatial confinement when extracted from volumes reconstructed by different algorithms. Aim: We evaluated the capabilities and limitations of two light-field reconstruction algorithms for calcium fluorescence imaging. Approach: We acquired light-field image series from neurons either bulk-labeled or filled intracellularly with the red-emitting calcium dye CaSiR-1 in acute mouse brain slices. We compared the tSNR and spatial confinement of calcium signals extracted from volumes reconstructed with synthetic refocusing and Richardson-Lucy three-dimensional deconvolution with and without total variation regularization. Results: Both synthetic refocusing and Richardson-Lucy deconvolution resolved calcium signals from single cells and neuronal dendrites in three dimensions. Increasing deconvolution iteration number improved spatial confinement but reduced tSNR compared with synthetic refocusing. Volumetric light-field imaging did not decrease calcium signal tSNR compared with interleaved, widefield image series acquired in matched planes. Conclusions: LFM enables high-volume rate, volumetric imaging of calcium transients in single cell somata (bulk-labeled) and dendrites (intracellularly loaded). The trade-offs identified for tSNR, spatial confinement, and computational cost indicate which of synthetic refocusing or deconvolution can better realize the scientific requirements of future LFM calcium imaging applications.
Collapse
Affiliation(s)
- Carmel L. Howe
- Imperial College London, Department of Bioengineering, London, United Kingdom
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
| | - Peter Quicke
- Imperial College London, Department of Bioengineering, London, United Kingdom
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
| | - Pingfan Song
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
- Imperial College London, Department of Electrical and Electronic Engineering, London, United Kingdom
| | - Herman Verinaz-Jadan
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
- Imperial College London, Department of Electrical and Electronic Engineering, London, United Kingdom
| | - Pier Luigi Dragotti
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
- Imperial College London, Department of Electrical and Electronic Engineering, London, United Kingdom
| | - Amanda J. Foust
- Imperial College London, Department of Bioengineering, London, United Kingdom
- Imperial College London, Centre for Neurotechnology, London, United Kingdom
| |
Collapse
|
8
|
Emiliani V, Entcheva E, Hedrich R, Hegemann P, Konrad KR, Lüscher C, Mahn M, Pan ZH, Sims RR, Vierock J, Yizhar O. Optogenetics for light control of biological systems. NATURE REVIEWS. METHODS PRIMERS 2022; 2:55. [PMID: 37933248 PMCID: PMC10627578 DOI: 10.1038/s43586-022-00136-4] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 11/08/2023]
Abstract
Optogenetic techniques have been developed to allow control over the activity of selected cells within a highly heterogeneous tissue, using a combination of genetic engineering and light. Optogenetics employs natural and engineered photoreceptors, mostly of microbial origin, to be genetically introduced into the cells of interest. As a result, cells that are naturally light-insensitive can be made photosensitive and addressable by illumination and precisely controllable in time and space. The selectivity of expression and subcellular targeting in the host is enabled by applying control elements such as promoters, enhancers and specific targeting sequences to the employed photoreceptor-encoding DNA. This powerful approach allows precise characterization and manipulation of cellular functions and has motivated the development of advanced optical methods for patterned photostimulation. Optogenetics has revolutionized neuroscience during the past 15 years and is primed to have a similar impact in other fields, including cardiology, cell biology and plant sciences. In this Primer, we describe the principles of optogenetics, review the most commonly used optogenetic tools, illumination approaches and scientific applications and discuss the possibilities and limitations associated with optogenetic manipulations across a wide variety of optical techniques, cells, circuits and organisms.
Collapse
Affiliation(s)
- Valentina Emiliani
- Wavefront Engineering Microscopy Group, Photonics Department, Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Rainer Hedrich
- Julius-von-Sachs Institute for Biosciences, Molecular Plant Physiology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Peter Hegemann
- Institute for Biology, Experimental Biophysics, Humboldt-Universitaet zu Berlin, Berlin, Germany
| | - Kai R. Konrad
- Julius-von-Sachs Institute for Biosciences, Molecular Plant Physiology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Christian Lüscher
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - Mathias Mahn
- Department of Neurobiology, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Zhuo-Hua Pan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ruth R. Sims
- Wavefront Engineering Microscopy Group, Photonics Department, Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Johannes Vierock
- Institute for Biology, Experimental Biophysics, Humboldt-Universitaet zu Berlin, Berlin, Germany
- Neuroscience Research Center, Charité – Universitaetsmedizin Berlin, Berlin, Germany
| | - Ofer Yizhar
- Departments of Brain Sciences and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
Voltage imaging in the olfactory bulb using transgenic mouse lines expressing the genetically encoded voltage indicator ArcLight. Sci Rep 2022; 12:1875. [PMID: 35115567 PMCID: PMC8813909 DOI: 10.1038/s41598-021-04482-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/09/2021] [Indexed: 01/06/2023] Open
Abstract
Genetically encoded voltage indicators (GEVIs) allow optical recordings of membrane potential changes in defined cell populations. Transgenic reporter animals that facilitate precise and repeatable targeting with high expression levels would further the use of GEVIs in the in vivo mammalian brain. However, the literature on developing and applying transgenic mouse lines as vehicles for GEVI expression is limited. Here we report the first in vivo experiments using a transgenic reporter mouse for the GEVI ArcLight, which utilizes a Cre/tTA dependent expression system (TIGRE 1.0). We developed two mouse lines with ArcLight expression restricted to either olfactory receptor neurons, or a subpopulation of interneurons located in the granule and glomerular layers in the olfactory bulb. The ArcLight expression in these lines was sufficient for in vivo imaging of odorant responses in single trials using epifluorescence and 2-photon imaging. The voltage responses were odor-specific and concentration-dependent, which supported earlier studies about perceptual transformations carried out by the bulb that used calcium sensors of neural activity. This study demonstrates that the ArcLight transgenic line is a flexible genetic tool that can be used to record the neuronal electrical activity of different cell types with a signal-to-noise ratio that is comparable to previous reports using viral transduction.
Collapse
|
10
|
Genetically encoded photo-switchable molecular sensors for optoacoustic and super-resolution imaging. Nat Biotechnol 2022; 40:598-605. [PMID: 34845372 PMCID: PMC9005348 DOI: 10.1038/s41587-021-01100-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/17/2021] [Indexed: 02/07/2023]
Abstract
Reversibly photo-switchable proteins are essential for many super-resolution fluorescence microscopic and optoacoustic imaging methods. However, they have yet to be used as sensors that measure the distribution of specific analytes at the nanoscale or in the tissues of live animals. Here we constructed the prototype of a photo-switchable Ca2+ sensor based on GCaMP5G that can be switched with 405/488-nm light and describe its molecular mechanisms at the structural level, including the importance of the interaction of the core barrel structure of the fluorescent protein with the Ca2+ receptor moiety. We demonstrate super-resolution imaging of Ca2+ concentration in cultured cells and optoacoustic Ca2+ imaging in implanted tumor cells in mice under controlled Ca2+ conditions. Finally, we show the generalizability of the concept by constructing examples of photo-switching maltose and dopamine sensors based on periplasmatic binding protein and G-protein-coupled receptor-based sensors.
Collapse
|
11
|
Jang J, Zhu MH, Jogdand AH, Antic SD. Studying Synaptically Evoked Cortical Responses ex vivo With Combination of a Single Neuron Recording (Whole-Cell) and Population Voltage Imaging (Genetically Encoded Voltage Indicator). Front Neurosci 2021; 15:773883. [PMID: 34776858 PMCID: PMC8579014 DOI: 10.3389/fnins.2021.773883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 11/15/2022] Open
Abstract
In a typical electrophysiology experiment, synaptic stimulus is delivered in a cortical layer (1-6) and neuronal responses are recorded intracellularly in individual neurons. We recreated this standard electrophysiological paradigm in brain slices of mice expressing genetically encoded voltage indicators (GEVIs). This allowed us to monitor membrane voltages in the target pyramidal neurons (whole-cell), and population voltages in the surrounding neuropil (optical imaging), simultaneously. Pyramidal neurons have complex dendritic trees that span multiple cortical layers. GEVI imaging revealed areas of the brain slice that experienced the strongest depolarization on a specific synaptic stimulus (location and intensity), thus identifying cortical layers that contribute the most afferent activity to the recorded somatic voltage waveform. By combining whole-cell with GEVI imaging, we obtained a crude distribution of activated synaptic afferents in respect to the dendritic tree of a pyramidal cell. Synaptically evoked voltage waves propagating through the cortical neuropil (dendrites and axons) were not static but rather they changed on a millisecond scale. Voltage imaging can identify areas of brain slices in which the neuropil was in a sustained depolarization (plateau), long after the stimulus onset. Upon a barrage of synaptic inputs, a cortical pyramidal neuron experiences: (a) weak temporal summation of evoked voltage transients (EPSPs); and (b) afterhyperpolarization (intracellular recording), which are not represented in the GEVI population imaging signal (optical signal). To explain these findings [(a) and (b)], we used four voltage indicators (ArcLightD, chi-VSFP, Archon1, and di-4-ANEPPS) with different optical sensitivity, optical response speed, labeling strategy, and a target neuron type. All four imaging methods were used in an identical experimental paradigm: layer 1 (L1) synaptic stimulation, to allow direct comparisons. The population voltage signal showed paired-pulse facilitation, caused in part by additional recruitment of new neurons and dendrites. "Synaptic stimulation" delivered in L1 depolarizes almost an entire cortical column to some degree.
Collapse
Affiliation(s)
| | | | | | - Srdjan D. Antic
- Department of Neuroscience, Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
12
|
Ochmann SE, Joshi H, Büber E, Franquelim HG, Stegemann P, Saccà B, Keyser UF, Aksimentiev A, Tinnefeld P. DNA Origami Voltage Sensors for Transmembrane Potentials with Single-Molecule Sensitivity. NANO LETTERS 2021; 21:8634-8641. [PMID: 34662130 DOI: 10.1021/acs.nanolett.1c02584] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Signal transmission in neurons goes along with changes in the transmembrane potential. To report them, different approaches, including optical voltage-sensing dyes and genetically encoded voltage indicators, have evolved. Here, we present a DNA nanotechnology-based system and demonstrated its functionality on liposomes. Using DNA origami, we incorporated and optimized different properties such as membrane targeting and voltage sensing modularly. As a sensing unit, we used a hydrophobic red dye anchored to the membrane and an anionic green dye at the DNA to connect the nanostructure and the membrane dye anchor. Voltage-induced displacement of the anionic donor unit was read out by fluorescence resonance energy transfer (FRET) changes of single sensors attached to liposomes. A FRET change of ∼5% for ΔΨ = 100 mV was observed. The working mechanism of the sensor was rationalized by molecular dynamics simulations. Our approach holds potential for an application as nongenetically encoded membrane sensors.
Collapse
Affiliation(s)
- Sarah E Ochmann
- Department of Chemistry and Center for NanoScience, Ludwig-Maximilians-Universität München, 81377 München, Germany
| | - Himanshu Joshi
- Department of Physics and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois 61820, United States
| | - Ece Büber
- Department of Chemistry and Center for NanoScience, Ludwig-Maximilians-Universität München, 81377 München, Germany
| | | | - Pierre Stegemann
- Center of Medical Biotechnology (ZMB) and Center for Nano Integration Duisburg-Essen (CENIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Barbara Saccà
- Center of Medical Biotechnology (ZMB) and Center for Nano Integration Duisburg-Essen (CENIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Ulrich F Keyser
- Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, United Kingdom
| | - Aleksei Aksimentiev
- Department of Physics and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois 61820, United States
| | - Philip Tinnefeld
- Department of Chemistry and Center for NanoScience, Ludwig-Maximilians-Universität München, 81377 München, Germany
| |
Collapse
|
13
|
Stuyt G, Godenzini L, Palmer LM. Local and Global Dynamics of Dendritic Activity in the Pyramidal Neuron. Neuroscience 2021; 489:176-184. [PMID: 34280492 DOI: 10.1016/j.neuroscience.2021.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022]
Abstract
There has been increasing interest in the measurement and comparison of activity across compartments of the pyramidal neuron. Dendritic activity can occur both locally, on a single dendritic segment, or globally, involving multiple compartments of the single neuron. Little is known about how these dendritic dynamics shape and contribute to information processing and behavior. Although it has been difficult to characterize local and global activity in vivo due to the technical challenge of simultaneously recording from the entire dendritic arbor and soma, the rise of calcium imaging has driven the increased feasibility and interest of these experiments. However, the distinction between local and global activity made by calcium imaging requires careful consideration. In this review we describe local and global activity, discuss the difficulties and caveats of this distinction, and present the evidence of local and global activity in information processing and behavior.
Collapse
Affiliation(s)
- George Stuyt
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Luca Godenzini
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Lucy M Palmer
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
14
|
Abreu N, Levitz J. Optogenetic Techniques for Manipulating and Sensing G Protein-Coupled Receptor Signaling. Methods Mol Biol 2021; 2173:21-51. [PMID: 32651908 DOI: 10.1007/978-1-0716-0755-8_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptors (GPCRs) form the largest class of membrane receptors in the mammalian genome with nearly 800 human genes encoding for unique subtypes. Accordingly, GPCR signaling is implicated in nearly all physiological processes. However, GPCRs have been difficult to study due in part to the complexity of their function which can lead to a plethora of converging or diverging downstream effects over different time and length scales. Classic techniques such as pharmacological control, genetic knockout and biochemical assays often lack the precision required to probe the functions of specific GPCR subtypes. Here we describe the rapidly growing set of optogenetic tools, ranging from methods for optical control of the receptor itself to optical sensing and manipulation of downstream effectors. These tools permit the quantitative measurements of GPCRs and their downstream signaling with high specificity and spatiotemporal precision.
Collapse
Affiliation(s)
- Nohely Abreu
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Joshua Levitz
- Biochemistry, Cell and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Adam Y. All-optical electrophysiology in behaving animals. J Neurosci Methods 2021; 353:109101. [PMID: 33600851 DOI: 10.1016/j.jneumeth.2021.109101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/13/2021] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Technology for simultaneous control and readout of the membrane potential of multiple neurons in behaving animals at high spatio-temporal resolution will have a high impact on neuroscience research. Significant progress in the development of Genetically Encoded Voltage Indicators (GEVIs) now enables to optically record subthreshold and spiking activity from ensembles of cells in behaving animals. In some cases, the GEVIs were also combined with optogenetic actuators to enable 'all-optical' control and readout of membrane potential at cellular resolution. Here I describe the recent progress in GEVI development and discuss the various aspects necessary to perform a successful 'all-optical' electrophysiology experiment in behaving, head-fixed animals. These aspects include the voltage indicators, the optogenetic actuators, strategies for protein expression, optical hardware, and image processing software. Furthermore, I discuss various applications of the technology, highlighting its advantages over classic electrode-based techniques. I argue that GEVIs now transformed from a 'promising' technology to a practical tool that can be used to tackle fundamental questions in neuroscience.
Collapse
Affiliation(s)
- Yoav Adam
- Edmond and Lily Safra Center for Brain Science, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
16
|
A dark quencher genetically encodable voltage indicator (dqGEVI) exhibits high fidelity and speed. Proc Natl Acad Sci U S A 2021; 118:2020235118. [PMID: 33531364 PMCID: PMC8017929 DOI: 10.1073/pnas.2020235118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Voltage sensing with genetically expressed optical probes is highly desirable for large-scale recordings of neuronal activity and detection of localized voltage signals in single neurons. Here we describe a method for a two-component (hybrid) genetically encodable fluorescent voltage sensing in neurons. The approach uses a glycosylphosphatidylinositol-tagged fluorescent protein (enhanced green fluorescent protein) that ensures the fluorescence to be specifically confined to the outside of the plasma membrane and D3, a voltage-dependent quencher. Previous hybrid genetically encoded voltage sensing approaches relied on a single quenching molecule, dipycrilamine (DPA), which is toxic, increases membrane capacitance, interferes with neurotransmitters, and is explosive. Our method uses a nontoxic and nonexplosive compound that performs better than DPA in all aspects of fluorescent voltage sensing. Voltage sensing with genetically expressed optical probes is highly desirable for large-scale recordings of neuronal activity and detection of localized voltage signals in single neurons. Most genetically encodable voltage indicators (GEVI) have drawbacks including slow response, low fluorescence, or excessive bleaching. Here we present a dark quencher GEVI approach (dqGEVI) using a Förster resonance energy transfer pair between a fluorophore glycosylphosphatidylinositol–enhanced green fluorescent protein (GPI-eGFP) on the outer surface of the neuronal membrane and an azo-benzene dye quencher (D3) that rapidly moves in the membrane driven by voltage. In contrast to previous probes, the sensor has a single photon bleaching time constant of ∼40 min, has a high temporal resolution and fidelity for detecting action potential firing at 100 Hz, resolves membrane de- and hyperpolarizations of a few millivolts, and has negligible effects on passive membrane properties or synaptic events. The dqGEVI approach should be a valuable tool for optical recordings of subcellular or population membrane potential changes in nerve cells.
Collapse
|
17
|
Kuner R, Kuner T. Cellular Circuits in the Brain and Their Modulation in Acute and Chronic Pain. Physiol Rev 2020; 101:213-258. [PMID: 32525759 DOI: 10.1152/physrev.00040.2019] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic, pathological pain remains a global health problem and a challenge to basic and clinical sciences. A major obstacle to preventing, treating, or reverting chronic pain has been that the nature of neural circuits underlying the diverse components of the complex, multidimensional experience of pain is not well understood. Moreover, chronic pain involves diverse maladaptive plasticity processes, which have not been decoded mechanistically in terms of involvement of specific circuits and cause-effect relationships. This review aims to discuss recent advances in our understanding of circuit connectivity in the mammalian brain at the level of regional contributions and specific cell types in acute and chronic pain. A major focus is placed on functional dissection of sub-neocortical brain circuits using optogenetics, chemogenetics, and imaging technological tools in rodent models with a view towards decoding sensory, affective, and motivational-cognitive dimensions of pain. The review summarizes recent breakthroughs and insights on structure-function properties in nociceptive circuits and higher order sub-neocortical modulatory circuits involved in aversion, learning, reward, and mood and their modulation by endogenous GABAergic inhibition, noradrenergic, cholinergic, dopaminergic, serotonergic, and peptidergic pathways. The knowledge of neural circuits and their dynamic regulation via functional and structural plasticity will be beneficial towards designing and improving targeted therapies.
Collapse
Affiliation(s)
- Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
18
|
Hartung JE, Gold MS. GCaMP as an indirect measure of electrical activity in rat trigeminal ganglion neurons. Cell Calcium 2020; 89:102225. [PMID: 32505783 DOI: 10.1016/j.ceca.2020.102225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 11/19/2022]
Abstract
While debate continues over whether somatosensory information is transmitted via labeled line, population coding, frequency coding, or some combination therein, researchers have begun to address this question at the level of the primary afferent by using optical approaches that enable the assessment of neural activity in hundreds to even thousands of neurons simultaneously. However, with limited availability of tools to optically assess electrical activity in large populations of neurons, researchers have turned to genetically encoded Ca2+ indicators (GECIs) including GCaMP to enable the detection of increases in cytosolic Ca2+ concentrations as a correlate for neuronal activity. One of the most widely used GECIs is GCaMP6, which is available in three different versions tuned for sensitivity (GCaMP6s), speed (GCaMP6f), or a balance of the two (GCaMP6m). In order to determine if these issues were unique to GCaMP6 itself, or if they were inherent to more than one generation of GCaMP, we also characterized jGCaMP7. In the present study, we sought to determine the utility of the three GCaMP6 isoforms to detect changes in activity in primary afferents at frequencies ranging from 0.1-30 Hz. Given the heterogeneity of sensory neurons, we also compared the performance of each GCaMP6 isoform in subpopulations of neurons defined by properties used to identify putative nociceptive afferents: cell body size, isolectin B4 (IB4) binding, and capsaicin sensitivity. Finally, we compared results generated with GCaMP6 with that generated from neurons expressing the next generation of GCaMP, jGCaMP7s and jGCaMP7f. A viral approach, with AAV9-CAG-GCaMP6s/m/f, was used to drive GECI expression in acutely dissociated rat trigeminal ganglion (TG) neurons, and neural activity was driven by electrical field stimulation. Infection efficiency with the AAV serotype was high >95 %, and the impact of GCaMP6 expression in TG neurons over the period of study (<10 days) on the regulation of intracellular Ca2+, as assessed with fura-2, was minimal. Having confirmed that the field stimulation evoked Ca2+ transients were dependent on Ca2+ influx secondary to the activation of action potentials and voltage-gated Ca2+ channels, we also confirmed that the signal-to-noise ratio for each of the isoforms was excellent, enabling detection of a single spike in>90% of neurons. However, the utility of the GCaMP6 isoforms to enable an assessment of the firing frequency let alone changes in firing frequency of each neuron was relatively limited and isoform specific: GCaMP6s and 6m had the lowest resolution, enabling detection of spikes at 3 Hz in 15% and 32% of neurons respectively, but it was possible to resolve discrete single spikes up to 10 Hz in 36% of GCaMP6f neurons. Unfortunately, using other parameters of the Ca2+ transient, such as magnitude of the transient or the rate of rise, did not improve the range over which these indicators could be used to assess changes in spike number or firing frequency. Furthermore, in the presence of ongoing neural activity, it was even more difficult to detect a change in firing frequency. The frequency response relationship for the increase in Ca2+ was highly heterogeneous among sensory neurons and was influenced by both the GCaMP6 isoform used to assess it, the timing between the delivery of stimulation trains (inter-burst interval), and afferent subpopulation. Notably, the same deficiencies were observed with jGCaMP7s and 7f in resolving the degree of activity as were present for the GCaMP6 isoforms. Together, these data suggest that while both GCaMP6 and jGCaMP7 are potentially useful tools in sensory neurons to determine the presence or absence of neural activity, the ability to discriminate changes in firing frequency ≥ 3 Hz is extremely limited. As a result, GECIs should probably not be used in sensory neurons to assess changes in activity within or between subpopulations of neurons.
Collapse
Affiliation(s)
- Jane E Hartung
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, United States.
| | - Michael S Gold
- Department of Neurobiology and the Pittsburgh Center for Pain Research, University of Pittsburgh, United States.
| |
Collapse
|
19
|
Fiala T, Wang J, Dunn M, Šebej P, Choi SJ, Nwadibia EC, Fialova E, Martinez DM, Cheetham CE, Fogle KJ, Palladino MJ, Freyberg Z, Sulzer D, Sames D. Chemical Targeting of Voltage Sensitive Dyes to Specific Cells and Molecules in the Brain. J Am Chem Soc 2020; 142:9285-9301. [PMID: 32395989 DOI: 10.1021/jacs.0c00861] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Voltage sensitive fluorescent dyes (VSDs) are important tools for probing signal transduction in neurons and other excitable cells. The impact of these highly lipophilic sensors has, however, been limited due to the lack of cell-specific targeting methods in brain tissue or living animals. We address this key challenge by introducing a nongenetic molecular platform for cell- and molecule-specific targeting of synthetic VSDs in the brain. We employ a dextran polymer particle to overcome the inherent lipophilicity of VSDs by dynamic encapsulation and high-affinity ligands to target the construct to specific neuronal cells utilizing only native components of the neurotransmission machinery at physiological expression levels. Dichloropane, a monoamine transporter ligand, enables targeting of dense dopaminergic axons in the mouse striatum and sparse noradrenergic axons in the mouse cortex in acute brain slices. PFQX in conjunction with ligand-directed acyl imidazole chemistry enables covalent labeling of AMPA-type glutamate receptors in the same brain regions. Probe variants bearing either a classical electrochromic ANEP dye or state-of-the-art VoltageFluor-type dye respond to membrane potential changes in a similar manner to the parent dyes, as shown by whole-cell patch recording. We demonstrate the feasibility of optical voltage recording with our probes in brain tissue with one-photon and two-photon fluorescence microscopy and define the signal limits of optical voltage imaging with synthetic sensors under a low photon budget determined by the native expression levels of the target proteins. This work demonstrates the feasibility of a chemical targeting approach and expands the possibilities of cell-specific imaging and pharmacology.
Collapse
Affiliation(s)
- Tomas Fiala
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Jihang Wang
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Matthew Dunn
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Peter Šebej
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Se Joon Choi
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10027, United States
| | - Ekeoma C Nwadibia
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Eva Fialova
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Diana M Martinez
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10027, United States
| | - Claire E Cheetham
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Keri J Fogle
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States.,Pittsburgh Institute of Neurodegenerative Diseases (PIND), University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Michael J Palladino
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States.,Pittsburgh Institute of Neurodegenerative Diseases (PIND), University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10027, United States.,Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10027, United States.,Department of Pharmacology, Columbia University Irving Medical Center, New York, New York 10027, United States.,Department of Molecular Therapeutics, New York Psychiatric Institute, New York, New York 10032, United States
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, New York 10027, United States.,NeuroTechnology Center at Columbia University, New York, New York 10027, United States
| |
Collapse
|
20
|
Chen G, Cao Y, Tang Y, Yang X, Liu Y, Huang D, Zhang Y, Li C, Wang Q. Advanced Near-Infrared Light for Monitoring and Modulating the Spatiotemporal Dynamics of Cell Functions in Living Systems. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903783. [PMID: 32328436 PMCID: PMC7175256 DOI: 10.1002/advs.201903783] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/06/2020] [Indexed: 05/07/2023]
Abstract
Light-based technique, including optical imaging and photoregulation, has become one of the most important tools for both fundamental research and clinical practice, such as cell signal sensing, cancer diagnosis, tissue engineering, drug delivery, visual regulation, neuromodulation, and disease treatment. In particular, low energy near-infrared (NIR, 700-1700 nm) light possesses lower phototoxicity and higher tissue penetration depth in living systems as compared with ultraviolet/visible light, making it a promising tool for in vivo applications. Currently, the NIR light-based imaging and photoregulation strategies have offered a possibility to real-time sense and/or modulate specific cellular events in deep tissues with subcellular accuracy. Herein, the recent progress with respect to NIR light for monitoring and modulating the spatiotemporal dynamics of cell functions in living systems are summarized. In particular, the applications of NIR light-based techniques in cancer theranostics, regenerative medicine, and neuroscience research are systematically introduced and discussed. In addition, the challenges and prospects for NIR light-based cell sensing and regulating techniques are comprehensively discussed.
Collapse
Affiliation(s)
- Guangcun Chen
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yuheng Cao
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Yanxing Tang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
| | - Xue Yang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yongyang Liu
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Dehua Huang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Yejun Zhang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Chunyan Li
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
| | - Qiangbin Wang
- CAS Key Laboratory of Nano‐Bio InterfaceDivision of Nanobiomedicine and i‐LabCAS Center for Excellence in Brain ScienceSuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- Suzhou Key Laboratory of Functional Molecular Imaging TechnologySuzhou Institute of Nano‐Tech and Nano‐BionicsChinese Academy of SciencesSuzhou215123China
- School of Nano‐Tech and Nano‐BionicsUniversity of Science and Technology of ChinaHefei230026China
- College of Materials Sciences and Opto‐Electronic TechnologyUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
21
|
Guitchounts G, Cox D. 64-Channel Carbon Fiber Electrode Arrays for Chronic Electrophysiology. Sci Rep 2020; 10:3830. [PMID: 32123283 PMCID: PMC7052209 DOI: 10.1038/s41598-020-60873-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 02/17/2020] [Indexed: 12/11/2022] Open
Abstract
A chief goal in neuroscience is to understand how neuronal activity relates to behavior, perception, and cognition. However, monitoring neuronal activity over long periods of time is technically challenging, and limited, in part, by the invasive nature of recording tools. While electrodes allow for recording in freely-behaving animals, they tend to be bulky and stiff, causing damage to the tissue they are implanted in. One solution to this invasiveness problem may be probes that are small enough to fly under the immune system's radar. Carbon fiber (CF) electrodes are thinner and more flexible than typical metal or silicon electrodes, but the arrays described in previous reports had low channel counts and required time-consuming manual assembly. Here we report the design of an expanded-channel-count carbon fiber electrode array (CFEA) as well as a method for fast preparation of the recording sites using acid etching and electroplating with PEDOT-TFB, and demonstrate the ability of the 64-channel CFEA to record from rat visual cortex. We include designs for interfacing the system with micro-drives or flex-PCB cables for recording from multiple brain regions, as well as a facilitated method for coating CFs with the insulator Parylene-C. High-channel-count CFEAs may thus be an alternative to traditional microwire-based electrodes and a practical tool for exploring the neural code.
Collapse
Affiliation(s)
- Grigori Guitchounts
- Center for Brain Science, Harvard University, Cambridge, Massachusetts, 02138, USA.
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA.
- Program in Neuroscience, Harvard University, Cambridge, Massachusetts, 02138, USA.
| | - David Cox
- Center for Brain Science, Harvard University, Cambridge, Massachusetts, 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
22
|
Singh S, Sharma MP, Ahmad A. Construction and characterization of protein-based cysteine nanosensor for the real time measurement of cysteine level in living cells. Int J Biol Macromol 2019; 143:273-284. [PMID: 31830444 DOI: 10.1016/j.ijbiomac.2019.12.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/25/2019] [Accepted: 12/03/2019] [Indexed: 11/26/2022]
Abstract
Cysteine plays a critical role in maintaining normal human metabolism, redox homeostasis, and immune regulation. Despite its functional versatility, cysteine metabolism in the human body is not well understood because of the lack of a robust tool for real-time measurement of cysteine at the cellular and sub-cellular level. In the present study, a genetically encoded nanosensor was developed using Cj0982 protein of Campylobacter jejuni, Enhanced Cyan Fluorescent Protein (ECFP) and Venus. The Cj0982 was sandwiched between ECFP and Venus for the construction of the nanosensor, named as Cys-FS (Cysteine-Fluorescent-Sensor). The Cys-FS is pH stable, specific to cysteine and has an affinity of 1.2 × 10-5 M. A range of affinity mutants were also developed with a cumulative cysteine detection range from 800 nM to 3.5 mM. The Cys-FS nanosensor was expressed in bacterial, yeast and mammalian cells, and the dynamics of cysteine level was measured in living cells using the confocal microscopy. The results showed that the Cys-FS nanosensor successfully monitored the dynamics of cysteine in both prokaryotic and eukaryotic systems without disrupting the cell. Thus, this study presents a novel nanosensor that can measure cysteine in living cells. This nanosensor is minimally invasive and non-toxic.
Collapse
Affiliation(s)
- Shruti Singh
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110019, India
| | - M P Sharma
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110019, India
| | - Altaf Ahmad
- Department of Botany, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
23
|
Optical voltage imaging in neurons: moving from technology development to practical tool. Nat Rev Neurosci 2019; 20:719-727. [PMID: 31705060 DOI: 10.1038/s41583-019-0231-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/13/2022]
Abstract
A central goal in neuroscience is to determine how the brain's neuronal circuits generate perception, cognition and emotions and how these lead to appropriate behavioural actions. A methodological platform based on genetically encoded voltage indicators (GEVIs) that enables the monitoring of large-scale circuit dynamics has brought us closer to this ambitious goal. This Review provides an update on the current state of the art and the prospects of emerging optical GEVI imaging technologies.
Collapse
|
24
|
Sun YH, Kao HKJ, Chang CW, Merleev A, Overton JL, Pretto D, Yechikov S, Maverakis E, Chiamvimonvat N, Chan JW, Lieu DK. Human induced pluripotent stem cell line with genetically encoded fluorescent voltage indicator generated via CRISPR for action potential assessment post-cardiogenesis. Stem Cells 2019; 38:90-101. [PMID: 31566285 DOI: 10.1002/stem.3085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 08/08/2019] [Indexed: 12/22/2022]
Abstract
Genetically encoded fluorescent voltage indicators, such as ArcLight, have been used to report action potentials (APs) in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). However, the ArcLight expression, in all cases, relied on a high number of lentiviral vector-mediated random genome integrations (8-12 copy/cell), raising concerns such as gene disruption and alteration of global and local gene expression, as well as loss or silencing of reporter genes after differentiation. Here, we report the use of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 nuclease technique to develop a hiPSC line stably expressing ArcLight from the AAVS1 safe harbor locus. The hiPSC line retained proliferative ability with a growth rate similar to its parental strain. Optical recording with conventional epifluorescence microscopy allowed the detection of APs as early as 21 days postdifferentiation, and could be repeatedly monitored for at least 5 months. Moreover, quantification and analysis of the APs of ArcLight-CMs identified two distinctive subtypes: a group with high frequency of spontaneous APs of small amplitudes that were pacemaker-like CMs and a group with low frequency of automaticity and large amplitudes that resembled the working CMs. Compared with FluoVolt voltage-sensitive dye, although dimmer, the ArcLight reporter exhibited better optical performance in terms of phototoxicity and photostability with comparable sensitivities and signal-to-noise ratios. The hiPSC line with targeted ArcLight engineering design represents a useful tool for studying cardiac development or hiPSC-derived cardiac disease models and drug testing.
Collapse
Affiliation(s)
- Yao-Hui Sun
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California
| | - Hillary K J Kao
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California
| | - Che-Wei Chang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California
| | - Alexander Merleev
- Department of Dermatology, University of California, Davis, Davis, California
| | - James L Overton
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California.,Bridges to Stem Cell Research Program, California State University, Sacramento, Sacramento, California
| | - Dalyir Pretto
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California
| | - Sergey Yechikov
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Davis, California
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Department of Veterans Affairs, Northern California Health Care System, Mather, California
| | - James W Chan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, California
| | - Deborah K Lieu
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, California.,Institute for Regenerative Cures and Stem Cell Program, University of California Davis Health Systems, Sacramento, California
| |
Collapse
|
25
|
Kleinfeld D, Luan L, Mitra PP, Robinson JT, Sarpeshkar R, Shepard K, Xie C, Harris TD. Can One Concurrently Record Electrical Spikes from Every Neuron in a Mammalian Brain? Neuron 2019; 103:1005-1015. [PMID: 31495645 PMCID: PMC6763354 DOI: 10.1016/j.neuron.2019.08.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/30/2019] [Accepted: 08/03/2019] [Indexed: 12/26/2022]
Abstract
The classic approach to measure the spiking response of neurons involves the use of metal electrodes to record extracellular potentials. Starting over 60 years ago with a single recording site, this technology now extends to ever larger numbers and densities of sites. We argue, based on the mechanical and electrical properties of existing materials, estimates of signal-to-noise ratios, assumptions regarding extracellular space in the brain, and estimates of heat generation by the electronic interface, that it should be possible to fabricate rigid electrodes to concurrently record from essentially every neuron in the cortical mantle. This will involve fabrication with existing yet nontraditional materials and procedures. We further emphasize the need to advance materials for improved flexible electrodes as an essential advance to record from neurons in brainstem and spinal cord in moving animals.
Collapse
Affiliation(s)
- David Kleinfeld
- Section of Neurobiology, University of California, San Diego, CA, USA; Department of Physics, University of California, San Diego, CA, USA.
| | - Lan Luan
- Department of Biomedical Engineering, University of Texas, Austin, TX, USA
| | - Partha P Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jacob T Robinson
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Rahul Sarpeshkar
- Department of Engineering, Dartmouth, Hanover, NH, USA; Department of Microbiology and Immunology, Dartmouth, Hanover, NH, USA; Department of Molecular and Systems Biology, Dartmouth, Hanover, NH, USA; Department of Physics, Dartmouth, Hanover, NH, USA
| | - Kenneth Shepard
- Department of Electrical Engineering, Columbia University, New York, NY, USA
| | - Chong Xie
- Department of Biomedical Engineering, University of Texas, Austin, TX, USA
| | - Timothy D Harris
- Howard Hughes Medical Institutes, Janelia Research Campus, Ashburn, VA, USA; Department of Bioengineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
26
|
Snyder AZ, Bauer AQ. Mapping Structure-Function Relationships in the Brain. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2019; 4:510-521. [PMID: 30528965 PMCID: PMC6488459 DOI: 10.1016/j.bpsc.2018.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 01/06/2023]
Abstract
Mapping the structural and functional connectivity of the brain is a major focus of systems neuroscience research and will help to identify causally important changes in neural circuitry responsible for behavioral dysfunction. Several methods for examining brain activity in humans have been extended to rodent and monkey models in which molecular and genetic manipulations exist for linking to human disease. In this review, which is part of a special issue focused on bridging brain connectivity information across species and spatiotemporal scales, we address mapping brain activity and neural connectivity in rodents using optogenetics in conjunction with either functional magnetic resonance imaging or optical intrinsic signal imaging. We chose to focus on these techniques because they are capable of reporting spontaneous or evoked hemodynamic activity most closely linked to human neuroimaging studies. We discuss the capabilities and limitations of blood-based imaging methods, usage of optogenetic techniques to map neural systems in rodent models, and other powerful mapping techniques for examining neural connectivity over different spatial and temporal scales. We also discuss implementing strategies for mapping brain connectivity in humans with both basic and clinical applications, and conclude with how cross-species mapping studies can be utilized to influence preclinical imaging studies and clinical practices alike.
Collapse
Affiliation(s)
- Abraham Z Snyder
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Adam Q Bauer
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
27
|
Emmenegger V, Obien MEJ, Franke F, Hierlemann A. Technologies to Study Action Potential Propagation With a Focus on HD-MEAs. Front Cell Neurosci 2019; 13:159. [PMID: 31118887 PMCID: PMC6504789 DOI: 10.3389/fncel.2019.00159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/08/2019] [Indexed: 12/26/2022] Open
Abstract
Axons convey information in neuronal circuits via reliable conduction of action potentials (APs) from the axon initial segment (AIS) to the presynaptic terminals. Recent experimental findings increasingly evidence that the axonal function is not limited to the simple transmission of APs. Advances in subcellular-resolution recording techniques have shown that axons display activity-dependent modulation in spike shape and conduction velocity, which influence synaptic strength and latency. We briefly review here, how recent methodological developments facilitate the understanding of the axon physiology. We included the three most common methods, i.e., genetically encoded voltage imaging (GEVI), subcellular patch-clamp and high-density microelectrode arrays (HD-MEAs). We then describe the potential of using HD-MEAs in studying axonal physiology in more detail. Due to their robustness, amenability to high-throughput and high spatiotemporal resolution, HD-MEAs can provide a direct functional electrical readout of single cells and cellular ensembles at subcellular resolution. HD-MEAs can, therefore, be employed in investigating axonal pathologies, the effects of large-scale genomic interventions (e.g., with RNAi or CRISPR) or in compound screenings. A combination of extracellular microelectrode arrays (MEAs), intracellular microelectrodes and optical imaging may potentially reveal yet unexplored repertoires of axonal functions.
Collapse
Affiliation(s)
- Vishalini Emmenegger
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Marie Engelene J. Obien
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- MaxWell Biosystems AG, Basel, Switzerland
| | - Felix Franke
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
28
|
|
29
|
Sigl-Glöckner J, Seibt J. Peeking into the sleeping brain: Using in vivo imaging in rodents to understand the relationship between sleep and cognition. J Neurosci Methods 2019; 316:71-82. [PMID: 30208306 PMCID: PMC6390172 DOI: 10.1016/j.jneumeth.2018.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022]
Abstract
Sleep is well known to benefit cognitive function. In particular, sleep has been shown to enhance learning and memory in both humans and animals. While the underlying mechanisms are not fully understood, it has been suggested that brain activity during sleep modulates neuronal communication through synaptic plasticity. These insights were mostly gained using electrophysiology to monitor ongoing large scale and single cell activity. While these efforts were instrumental in the characterisation of important network and cellular activity during sleep, several aspects underlying cognition are beyond the reach of this technology. Neuronal circuit activity is dynamically regulated via the precise interaction of different neuronal and non-neuronal cell types and relies on subtle modifications of individual synapses. In contrast to established electrophysiological approaches, recent advances in imaging techniques, mainly applied in rodents, provide unprecedented access to these aspects of neuronal function in vivo. In this review, we describe various techniques currently available for in vivo brain imaging, from single synapse to large scale network activity. We discuss the advantages and limitations of these approaches in the context of sleep research and describe which particular aspects related to cognition lend themselves to this kind of investigation. Finally, we review the few studies that used in vivo imaging in rodents to investigate the sleeping brain and discuss how the results have already significantly contributed to a better understanding on the complex relation between sleep and plasticity across development and adulthood.
Collapse
Affiliation(s)
- Johanna Sigl-Glöckner
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, D-10115, Berlin, Germany
| | - Julie Seibt
- Surrey Sleep Research Centre, University of Surrey, GU2 7XP, Guildford, UK.
| |
Collapse
|
30
|
Padamsey Z, Tong R, Emptage N. Optical Quantal Analysis Using Ca 2+ Indicators: A Robust Method for Assessing Transmitter Release Probability at Excitatory Synapses by Imaging Single Glutamate Release Events. Front Synaptic Neurosci 2019; 11:5. [PMID: 30886576 PMCID: PMC6409341 DOI: 10.3389/fnsyn.2019.00005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
Despite evidence that presynaptic efficacy and plasticity influence circuit function and behavior in vivo, studies of presynaptic function remain challenging owing to the difficulty of assessing transmitter release in intact tissue. Electrophysiological analyses of transmitter release are indirect and cannot readily resolve basic presynaptic parameters, most notably transmitter release probability (p r), at single synapses. These issues can be circumvented by optical quantal analysis, which uses the all-or-none optical detection of transmitter release in order to calculate p r. Over the past two decades, we and others have successfully demonstrated that Ca2+ indicators can be strategically implemented to perform optical quantal analysis at single glutamatergic synapses in ex vivo and in vitro preparations. We have found that high affinity Ca2+ indicators can reliably detect spine Ca2+ influx generated by single quanta of glutamate, thereby enabling precise calculation of pr at single synapses. Importantly, we have shown this method to be robust to changes in postsynaptic efficacy, and to be sensitive to activity-dependent presynaptic changes at central synapses following the induction of long-term potentiation (LTP) and long-term depression (LTD). In this report, we describe how to use Ca2+-sensitive dyes to perform optical quantal analysis at single synapses in hippocampal slice preparations. The general technique we describe here can be applied to other glutamatergic synapses and can be used with other reporters of glutamate release, including recently improved genetically encoded Ca2+ and glutamate sensors. With ongoing developments in imaging techniques and genetically encoded probes, optical quantal analysis is a promising strategy for assessing presynaptic function and plasticity in vivo.
Collapse
Affiliation(s)
- Zahid Padamsey
- Center for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Rudi Tong
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Nigel Emptage
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
31
|
Melzer W. No voltage change at skeletal muscle SR membrane during Ca 2+ release-just Mermaids on acid. J Gen Physiol 2018; 150:1055-1058. [PMID: 29970411 PMCID: PMC6080887 DOI: 10.1085/jgp.201812084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Melzer highlights new work confirming that the sarcoplasmic reticulum transmembrane voltage changes little during Ca2+ release Calcium ions control multiple physiological functions by binding to extracellular and intracellular targets. One of the best-studied Ca2+-dependent functions is contraction of smooth and striated muscle tissue, which results from Ca2+ ligation to calmodulin and troponin C, respectively. Ca2+ signaling typically involves flux of the ion across membranes via specifically gated channel proteins. Because calcium ions are charged, they possess the ability to generate changes in the respective transmembrane voltage. Ca2+-dependent voltage alterations of the surface membrane are easily measured using microelectrodes. A well-known example is the characteristic plateau phase of the action potential in cardiac ventricular cells that results from the opening of voltage-gated L-type Ca2+ channels. Ca2+ ions are also released from intracellular storage compartments in many cells, but these membranes are not accessible to direct voltage recording with microelectrodes. In muscle, for example, release of Ca2+ from the sarcoplasmic reticulum (SR) to the myoplasm constitutes a flux that is considerably larger than the entry flux from the extracellular space. Whether this flux is accompanied by a voltage change across the SR membrane is an obvious question of mechanistic importance and has been the subject of many investigations. Because the tiny spaces enclosed by the SR membrane are inaccessible to microelectrodes, alternative methods have to be applied. In a study by Sanchez et al. (2018. J. Gen. Physiol.https://doi.org/10.1085/jgp.201812035) in this issue, modern confocal light microscopy and genetically encoded voltage probes targeted to the SR were applied in a new approach to search for changes in the membrane potential of the SR during Ca2+ release.
Collapse
Affiliation(s)
- Werner Melzer
- Institut für Angewandte Physiologie, Universität Ulm, Ulm, Germany
| |
Collapse
|