151
|
Todorova V, Stauffacher MF, Ravotto L, Nötzli S, Karademir D, Ebner LJA, Imsand C, Merolla L, Hauck SM, Samardzija M, Saab AS, Barros LF, Weber B, Grimm C. Deficits in mitochondrial TCA cycle and OXPHOS precede rod photoreceptor degeneration during chronic HIF activation. Mol Neurodegener 2023; 18:15. [PMID: 36882871 PMCID: PMC9990367 DOI: 10.1186/s13024-023-00602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/03/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Major retinal degenerative diseases, including age-related macular degeneration, diabetic retinopathy and retinal detachment, are associated with a local decrease in oxygen availability causing the formation of hypoxic areas affecting the photoreceptor (PR) cells. Here, we addressed the underlying pathological mechanisms of PR degeneration by focusing on energy metabolism during chronic activation of hypoxia-inducible factors (HIFs) in rod PR. METHODS We used two-photon laser scanning microscopy (TPLSM) of genetically encoded biosensors delivered by adeno-associated viruses (AAV) to determine lactate and glucose dynamics in PR and inner retinal cells. Retinal layer-specific proteomics, in situ enzymatic assays and immunofluorescence studies were used to analyse mitochondrial metabolism in rod PRs during chronic HIF activation. RESULTS PRs exhibited remarkably higher glycolytic flux through the hexokinases than neurons of the inner retina. Chronic HIF activation in rods did not cause overt change in glucose dynamics but an increase in lactate production nonetheless. Furthermore, dysregulation of the oxidative phosphorylation pathway (OXPHOS) and tricarboxylic acid (TCA) cycle in rods with an activated hypoxic response decelerated cellular anabolism causing shortening of rod photoreceptor outer segments (OS) before onset of cell degeneration. Interestingly, rods with deficient OXPHOS but an intact TCA cycle did not exhibit these early signs of anabolic dysregulation and showed a slower course of degeneration. CONCLUSION Together, these data indicate an exceeding high glycolytic flux in rods and highlight the importance of mitochondrial metabolism and especially of the TCA cycle for PR survival in conditions of increased HIF activity.
Collapse
Affiliation(s)
- Vyara Todorova
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Mia Fee Stauffacher
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - Sarah Nötzli
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Cornelia Imsand
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Luca Merolla
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Munich, Germany
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile.,Universidad San Sebastián, Valdivia, Chile
| | - Bruno Weber
- Institute of Pharmacology and Toxicology and Neuroscience Center Zurich, University and ETH Zurich, Winterthurerstr. 190, 8057, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952, Schlieren, Switzerland.
| |
Collapse
|
152
|
Hou SS, Yang J, Lee JH, Kwon Y, Calvo-Rodriguez M, Bao K, Ahn S, Kashiwagi S, Kumar ATN, Bacskai BJ, Choi HS. Near-infrared fluorescence lifetime imaging of amyloid-β aggregates and tau fibrils through the intact skull of mice. Nat Biomed Eng 2023; 7:270-280. [PMID: 36747008 PMCID: PMC10040441 DOI: 10.1038/s41551-023-01003-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/08/2023] [Indexed: 02/08/2023]
Abstract
Non-invasive methods for the in vivo detection of hallmarks of Alzheimer's disease can facilitate the study of the progression of the disease in mouse models and may enable its earlier diagnosis in humans. Here we show that the zwitterionic heptamethine fluorophore ZW800-1C, which has peak excitation and emission wavelengths in the near-infrared optical window, binds in vivo and at high contrast to amyloid-β deposits and to neurofibrillary tangles, and allows for the microscopic imaging of amyloid-β and tau aggregates through the intact skull of mice. In transgenic mouse models of Alzheimer's disease, we compare the performance of ZW800-1C with that of the two spectrally similar heptamethine fluorophores ZW800-1A and indocyanine green, and show that ZW800-1C undergoes a longer fluorescence-lifetime shift when bound to amyloid-β and tau aggregates than when circulating in blood vessels. ZW800-1C may prove advantageous for tracking the proteinic aggregates in rodent models of amyloid-β and tau pathologies.
Collapse
Affiliation(s)
- Steven S Hou
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joyce Yang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeong Heon Lee
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yeseo Kwon
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Calvo-Rodriguez
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kai Bao
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sung Ahn
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand T N Kumar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian J Bacskai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
153
|
Zhang Y, Rózsa M, Liang Y, Bushey D, Wei Z, Zheng J, Reep D, Broussard GJ, Tsang A, Tsegaye G, Narayan S, Obara CJ, Lim JX, Patel R, Zhang R, Ahrens MB, Turner GC, Wang SSH, Korff WL, Schreiter ER, Svoboda K, Hasseman JP, Kolb I, Looger LL. Fast and sensitive GCaMP calcium indicators for imaging neural populations. Nature 2023; 615:884-891. [PMID: 36922596 PMCID: PMC10060165 DOI: 10.1038/s41586-023-05828-9] [Citation(s) in RCA: 313] [Impact Index Per Article: 156.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/10/2023] [Indexed: 03/17/2023]
Abstract
Calcium imaging with protein-based indicators1,2 is widely used to follow neural activity in intact nervous systems, but current protein sensors report neural activity at timescales much slower than electrical signalling and are limited by trade-offs between sensitivity and kinetics. Here we used large-scale screening and structure-guided mutagenesis to develop and optimize several fast and sensitive GCaMP-type indicators3-8. The resulting 'jGCaMP8' sensors, based on the calcium-binding protein calmodulin and a fragment of endothelial nitric oxide synthase, have ultra-fast kinetics (half-rise times of 2 ms) and the highest sensitivity for neural activity reported for a protein-based calcium sensor. jGCaMP8 sensors will allow tracking of large populations of neurons on timescales relevant to neural computation.
Collapse
Affiliation(s)
- Yan Zhang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Márton Rózsa
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Yajie Liang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel Bushey
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ziqiang Wei
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jihong Zheng
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Daniel Reep
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | | | - Arthur Tsang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Getahun Tsegaye
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Sujatha Narayan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Jing-Xuan Lim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Rongwei Zhang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Glenn C Turner
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Samuel S-H Wang
- Neuroscience Institute, Princeton University, Princeton, NJ, USA.
| | - Wyatt L Korff
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Eric R Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Karel Svoboda
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Allen Institute for Neural Dynamics, Seattle, WA, USA.
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Jeremy P Hasseman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Ilya Kolb
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Genetically Encoded Neural Indicator and Effector (GENIE) Project, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
154
|
Gilday OD, Mizrahi A. Learning-Induced Odor Modulation of Neuronal Activity in Auditory Cortex. J Neurosci 2023; 43:1375-1386. [PMID: 36650061 PMCID: PMC9987573 DOI: 10.1523/jneurosci.1398-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Sensory cortices, even of primary regions, are not purely unisensory. Rather, cortical neurons in sensory cortex show various forms of multisensory interactions. While some multisensory interactions naturally co-occur, the combination of others will co-occur through experience. In real life, learning and experience will result in conjunction with seemingly disparate sensory information that ultimately becomes behaviorally relevant, impacting perception, cognition, and action. Here we describe a novel auditory discrimination task in mice, designed to manipulate the expectation of upcoming trials using olfactory cues. We show that, after learning, female mice display a transient period of several days during which they exploit odor-mediated expectations for making correct decisions. Using two-photon calcium imaging of single neurons in auditory cortex (ACx) during behavior, we found that the behavioral effects of odor-mediated expectations are accompanied by an odor-induced modulation of neuronal activity. Further, we find that these effects are manifested differentially, based on the response preference of individual cells. A significant portion of effects, but not all, are consistent with a predictive coding framework. Our data show that learning novel odor-sound associations evoke changes in ACx. We suggest that behaviorally relevant multisensory environments mediate contextual effects as early as ACx.SIGNIFICANCE STATEMENT Natural environments are composed of multisensory objects. It remains unclear whether and how animals learn the regularities of congruent multisensory associations and how these may impact behavior and neural activity. We tested how learned odor-sound associations affected single-neuron responses in auditory cortex. We introduce a novel auditory discrimination task for mice in which odors set different contexts of expectation to upcoming trials. We show that, although the task can be solved purely by sounds, odor-mediated expectation impacts performance. We further show that odors cause a modulation of neuronal activity in auditory cortex, which is correlated with behavior. These results suggest that learning prompts an interaction of odor and sound information as early as sensory cortex.
Collapse
Affiliation(s)
- Omri David Gilday
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Adi Mizrahi
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel,
- Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
155
|
Zheng T, Liversage AR, Tehrani KF, Call JA, Kner PA, Mortensen LJ. Imaging mitochondria through bone in live mice using two-photon fluorescence microscopy with adaptive optics. FRONTIERS IN NEUROIMAGING 2023; 2:959601. [PMID: 37554651 PMCID: PMC10406258 DOI: 10.3389/fnimg.2023.959601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/09/2023] [Indexed: 08/10/2023]
Abstract
INTRODUCTION Mitochondria are extremely important organelles in the regulation of bone marrow and brain activity. However, live imaging of these subcellular features with high resolution in scattering tissues like brain or bone has proven challenging. METHODS In this study, we developed a two-photon fluorescence microscope with adaptive optics (TPFM-AO) for high-resolution imaging, which uses a home-built Shack-Hartmann wavefront sensor (SHWFS) to correct system aberrations and a sensorless approach for correcting low order tissue aberrations. RESULTS Using AO increases the fluorescence intensity of the point spread function (PSF) and achieves fast imaging of subcellular organelles with 400 nm resolution through 85 μm of highly scattering tissue. We achieved ~1.55×, ~3.58×, and ~1.77× intensity increases using AO, and a reduction of the PSF width by ~0.83×, ~0.74×, and ~0.9× at the depths of 0, 50 μm and 85 μm in living mouse bone marrow respectively, allowing us to characterize mitochondrial health and the survival of functioning cells with a field of view of 67.5× 67.5 μm. We also investigate the role of initial signal and background levels in sample correction quality by varying the laser power and camera exposure time and develop an intensity-based criteria for sample correction. DISCUSSION This study demonstrates a promising tool for imaging of mitochondria and other organelles in optically distorting biological environments, which could facilitate the study of a variety of diseases connected to mitochondrial morphology and activity in a range of biological tissues.
Collapse
Affiliation(s)
- Tianyi Zheng
- School of Electrical and Computer Engineering, University of Georgia, Athens, GA, United States
| | - Adrian R. Liversage
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA, United States
| | - Kayvan F. Tehrani
- Biophotonics Imaging Laboratory, The University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Jarrod A. Call
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Peter A. Kner
- School of Electrical and Computer Engineering, University of Georgia, Athens, GA, United States
| | - Luke J. Mortensen
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, GA, United States
- Regenerative Bioscience Center, Rhodes Center for ADS, University of Georgia, Athens, GA, United States
| |
Collapse
|
156
|
Clary RC, Jenkins BA, Lumpkin EA. Spatiotemporal dynamics of sensory neuron and Merkel-cell remodeling are decoupled during epidermal homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528558. [PMID: 36824872 PMCID: PMC9949164 DOI: 10.1101/2023.02.14.528558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
As the juncture between the body and environment, epithelia are both protective barriers and sensory interfaces that continually renew. To determine whether sensory neurons remodel to maintain homeostasis, we used in vivo two-photon imaging of somatosensory axons innervating Merkel cells in adult mouse skin. These touch receptors were highly plastic: 63% of Merkel cells and 89% of branches appeared, disappeared, grew, regressed and/or relocated over a month. Interestingly, Merkel-cell plasticity was synchronized across arbors during rapid epithelial turnover. When Merkel cells remodeled, the degree of plasticity between Merkel-cell clusters and their axons was well correlated. Moreover, branches were stabilized by Merkel-cell contacts. These findings highlight the role of epithelial-neural crosstalk in homeostatic remodeling. Conversely, axons were also dynamic when Merkel cells were stable, indicating that intrinsic neural mechanisms drive branch plasticity. Two terminal morphologies innervated Merkel cells: transient swellings called boutons, and stable cups termed kylikes. In Atoh1 knockout mice that lack Merkel cells, axons showed higher complexity than control mice, with exuberant branching and no kylikes. Thus, Merkel cells limit axonal branching and promote branch maturation. Together, these results reveal a previously unsuspected high degree of plasticity in somatosensory axons that is biased, but not solely dictated, by plasticity of target epithelial cells. This system provides a platform to identify intrinsic and extrinsic mechanisms that govern axonal patterning in epithelial homeostasis.
Collapse
|
157
|
Real-time denoising enables high-sensitivity fluorescence time-lapse imaging beyond the shot-noise limit. Nat Biotechnol 2023; 41:282-292. [PMID: 36163547 PMCID: PMC9931589 DOI: 10.1038/s41587-022-01450-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/29/2022] [Indexed: 11/09/2022]
Abstract
A fundamental challenge in fluorescence microscopy is the photon shot noise arising from the inevitable stochasticity of photon detection. Noise increases measurement uncertainty and limits imaging resolution, speed and sensitivity. To achieve high-sensitivity fluorescence imaging beyond the shot-noise limit, we present DeepCAD-RT, a self-supervised deep learning method for real-time noise suppression. Based on our previous framework DeepCAD, we reduced the number of network parameters by 94%, memory consumption by 27-fold and processing time by a factor of 20, allowing real-time processing on a two-photon microscope. A high imaging signal-to-noise ratio can be acquired with tenfold fewer photons than in standard imaging approaches. We demonstrate the utility of DeepCAD-RT in a series of photon-limited experiments, including in vivo calcium imaging of mice, zebrafish larva and fruit flies, recording of three-dimensional (3D) migration of neutrophils after acute brain injury and imaging of 3D dynamics of cortical ATP release. DeepCAD-RT will facilitate the morphological and functional interrogation of biological dynamics with a minimal photon budget.
Collapse
|
158
|
Campbell EP, Abushawish AA, Valdez LA, Bell MK, Haryono M, Rangamani P, Bloodgood BL. Electrical signals in the ER are cell type and stimulus specific with extreme spatial compartmentalization in neurons. Cell Rep 2023; 42:111943. [PMID: 36640310 PMCID: PMC10033362 DOI: 10.1016/j.celrep.2022.111943] [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: 01/12/2022] [Revised: 10/04/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
The endoplasmic reticulum (ER) is a tortuous organelle that spans throughout a cell with a continuous membrane containing ion channels, pumps, and transporters. It is unclear if stimuli that gate ER ion channels trigger substantial membrane potential fluctuations and if those fluctuations spread beyond their site of origin. Here, we visualize ER membrane potential dynamics in HEK cells and cultured rat hippocampal neurons by targeting a genetically encoded voltage indicator specifically to the ER membrane. We report the existence of clear cell-type- and stimulus-specific ER membrane potential fluctuations. In neurons, direct stimulation of ER ryanodine receptors generates depolarizations that scale linearly with stimulus strength and reach tens of millivolts. However, ER potentials do not spread beyond the site of receptor activation, exhibiting steep attenuation that is exacerbated by intracellular large conductance K+ channels. Thus, segments of ER can generate large depolarizations that are actively restricted from impacting nearby, contiguous membrane.
Collapse
Affiliation(s)
- Evan P Campbell
- Neurobiology Department, School of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Ahmed A Abushawish
- Neurobiology Department, School of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Lauren A Valdez
- Neurobiology Department, School of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Miriam K Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Melita Haryono
- Neurobiology Department, School of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Brenda L Bloodgood
- Neurobiology Department, School of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
159
|
Komorowska-Müller JA, Gellner AK, Ravichandran KA, Bilkei-Gorzo A, Zimmer A, Stein V. Chronic low-dose Δ 9-tetrahydrocannabinol (THC) treatment stabilizes dendritic spines in 18-month-old mice. Sci Rep 2023; 13:1390. [PMID: 36697430 PMCID: PMC9877016 DOI: 10.1038/s41598-022-27146-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/27/2022] [Indexed: 01/26/2023] Open
Abstract
Cognitive functions decline during aging. This decline could be caused by changes in dendritic spine stability and altered spine dynamics. Previously, we have shown that a low dose chronic THC treatment improves learning abilities in old whereas impairs learning abilities in young mice. The mechanism underlying this age-dependent effect is not known. Dendritic spine stability is a key for memory formation, therefore we hypothesized that THC affects spine dynamics in an age-dependent manner. We applied longitudinal 2-photon in vivo imaging to 3- and 18-month-old mice treated with 3 mg/kg/day of THC for 28 days via an osmotic pump. We imaged the same dendritic segments before, during and after the treatment and assessed changes in spine density and stability. We now show that in old mice THC improved spine stability resulting in a long-lasting increase in spine density. In contrast, in young mice THC transiently increased spine turnover and destabilized the spines.
Collapse
Affiliation(s)
| | - Anne-Kathrin Gellner
- Medical Faculty, Institute of Physiology II, University of Bonn, Bonn, Germany.,Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Kishore Aravind Ravichandran
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany.,Medical Faculty, Institute of Physiology II, University of Bonn, Bonn, Germany
| | - Andras Bilkei-Gorzo
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | - Andreas Zimmer
- Medical Faculty, Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany.
| | - Valentin Stein
- Medical Faculty, Institute of Physiology II, University of Bonn, Bonn, Germany.
| |
Collapse
|
160
|
Rem PD, Sereikaite V, Fernández-Fernández D, Reinartz S, Ulrich D, Fritzius T, Trovo L, Roux S, Chen Z, Rondard P, Pin JP, Schwenk J, Fakler B, Gassmann M, Barkat TR, Strømgaard K, Bettler B. Soluble amyloid-β precursor peptide does not regulate GABA B receptor activity. eLife 2023; 12:82082. [PMID: 36688536 PMCID: PMC9917443 DOI: 10.7554/elife.82082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/22/2023] [Indexed: 01/24/2023] Open
Abstract
Amyloid-β precursor protein (APP) regulates neuronal activity through the release of secreted APP (sAPP) acting at cell surface receptors. APP and sAPP were reported to bind to the extracellular sushi domain 1 (SD1) of GABAB receptors (GBRs). A 17 amino acid peptide (APP17) derived from APP was sufficient for SD1 binding and shown to mimic the inhibitory effect of sAPP on neurotransmitter release and neuronal activity. The functional effects of APP17 and sAPP were similar to those of the GBR agonist baclofen and blocked by a GBR antagonist. These experiments led to the proposal that sAPP activates GBRs to exert its neuronal effects. However, whether APP17 and sAPP influence classical GBR signaling pathways in heterologous cells was not analyzed. Here, we confirm that APP17 binds to GBRs with nanomolar affinity. However, biochemical and electrophysiological experiments indicate that APP17 does not influence GBR activity in heterologous cells. Moreover, APP17 did not regulate synaptic GBR localization, GBR-activated K+ currents, neurotransmitter release, or neuronal activity in vitro or in vivo. Our results show that APP17 is not a functional GBR ligand and indicate that sAPP exerts its neuronal effects through receptors other than GBRs.
Collapse
Affiliation(s)
- Pascal Dominic Rem
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Vita Sereikaite
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | | | - Sebastian Reinartz
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Daniel Ulrich
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Thorsten Fritzius
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Luca Trovo
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | - Salomé Roux
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Ziyang Chen
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | - Philippe Rondard
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, Université de MontpellierMontpellierFrance
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of FreiburgFreiburgGermany
- CIBSS Center for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- Center for Basics in NeuroModulationFreiburgGermany
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| | | | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, UniversitetsparkenCopenhagenDenmark
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of BaselBaselSwitzerland
| |
Collapse
|
161
|
Rahmatullah N, Schmitt LM, De Stefano L, Post S, Robledo J, Chaudhari GR, Pedapati E, Erickson CA, Portera-Cailliau C, Goel A. Hypersensitivity to distractors in Fragile X syndrome from loss of modulation of cortical VIP interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522654. [PMID: 36711901 PMCID: PMC9881942 DOI: 10.1101/2023.01.03.522654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Attention deficit is one of the most prominent and disabling symptoms in Fragile X Syndrome (FXS). Hypersensitivity to sensory stimuli contributes to attention difficulties by overwhelming and/or distracting affected individuals, which disrupts activities of daily living at home and learning at school. We find that auditory or visual distractors selectively impair visual discrimination performance in both humans and mice with FXS, but not their typically developing controls. Vasoactive intestinal polypeptide (VIP) neurons were significantly modulated by incorrect responses in the post-stimulus period during early distractor trials in WT mice, consistent with their known role as 'error' signals. Strikingly, however, VIP cells from Fmr1-/- mice showed little modulation in error trials, and this correlated with their poor performance on the distractor task. Thus, VIP interneurons and their reduced modulatory influence on pyramidal cells, could be a potential therapeutic target for attentional difficulties in FXS.
Collapse
Affiliation(s)
- Noorhan Rahmatullah
- Neuroscience Graduate Program, UC Riverside, CA
- Department of Psychology, UC Riverside, CA
| | - Lauren M. Schmitt
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, OH
| | - Lisa De Stefano
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, OH
| | - Sam Post
- Department of Psychology, UC Riverside, CA
| | | | | | - Ernest Pedapati
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, OH
- Department of Neurology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, OH
| | - Craig A. Erickson
- Department of Psychiatry, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, OH
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, CA
- Department of Neurobiology, David Geffen School of Medicine at UCLA, CA
| | - Anubhuti Goel
- Neuroscience Graduate Program, UC Riverside, CA
- Department of Psychology, UC Riverside, CA
| |
Collapse
|
162
|
Eleftheriou A, Ravotto L, Wyss MT, Warnock G, Siebert A, Zaiss M, Weber B. Simultaneous dynamic glucose-enhanced (DGE) MRI and fiber photometry measurements of glucose in the healthy mouse brain. Neuroimage 2023; 265:119762. [PMID: 36427752 DOI: 10.1016/j.neuroimage.2022.119762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Glucose is the main energy source in the brain and its regulated uptake and utilization are important biomarkers of pathological brain function. Glucose Chemical Exchange Saturation Transfer (GlucoCEST) and its time-resolved version Dynamic Glucose-Enhanced MRI (DGE) are promising approaches to monitor glucose and detect tumors, since they are radioactivity-free, do not require 13C labeling and are is easily translatable to the clinics. The main principle of DGE is clear. However, what remains to be established is to which extent the signal reflects vascular, extracellular or intracellular glucose. To elucidate the compartmental contributions to the DGE signal, we coupled it with FRET-based fiber photometry of genetically encoded sensors, a technique that combines quantitative glucose readout with cellular specificity. The glucose sensor FLIIP was used with fiber photometry to measure astrocytic and neuronal glucose changes upon injection of D-glucose, 3OMG and L-glucose, in the anaesthetized murine brain. By correlating the kinetic profiles of the techniques, we demonstrate the presence of a vascular contribution to the signal, especially at early time points after injection. Furthermore, we show that, in the case of the commonly used contrast agent 3OMG, the DGE signal actually anticorrelates with the glucose concentration in neurons and astrocytes.
Collapse
Affiliation(s)
- Afroditi Eleftheriou
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Matthias T Wyss
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Geoffrey Warnock
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Anita Siebert
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Moritz Zaiss
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nürnberg, Erlangen, Germany; High-field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
163
|
Murakoshi H, Ueda HH, Goto R, Hamada K, Nagasawa Y, Fuji T. In vivo three- and four-photon fluorescence microscopy using a 1.8 µm femtosecond fiber laser system. BIOMEDICAL OPTICS EXPRESS 2023; 14:326-334. [PMID: 36698657 PMCID: PMC9841992 DOI: 10.1364/boe.477322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/13/2022] [Accepted: 11/28/2022] [Indexed: 05/25/2023]
Abstract
Multiphoton microscopy has enabled us to image cellular dynamics in vivo. However, the excitation wavelength for imaging with commercially available lasers is mostly limited between 0.65-1.04 µm. Here we develop a femtosecond fiber laser system that produces ∼150 fs pulses at 1.8 µm. Our system starts from an erbium-doped silica fiber laser, and its wavelength is converted to 1.8 µm using a Raman shift fiber. The 1.8 µm pulses are amplified with a two-stage Tm:ZBLAN fiber amplifier. The final pulse energy is ∼1 µJ, sufficient for in vivo imaging. We successfully observe TurboFP635-expressing cortical neurons at a depth of 0.7 mm from the brain surface by three-photon excitation and Clover-expressing astrocytes at a depth of 0.15 mm by four-photon excitation.
Collapse
Affiliation(s)
- Hideji Murakoshi
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Kanagawa, 240-0193, Japan
- Contributed equally
| | - Hiromi H. Ueda
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Kanagawa, 240-0193, Japan
| | - Ryuichiro Goto
- FiberLabs Inc., KDDI Laboratories Building, 2-1-15 Ohara, Fujimino, Saitama 356-8502, Japan
| | - Kosuke Hamada
- Laser Science Laboratory, Toyota Technological Institute, 2-12-1 Hisakata, Tempaku-ku, Nagoya, 468-8511, Japan
| | - Yutaro Nagasawa
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8585, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Kanagawa, 240-0193, Japan
| | - Takao Fuji
- Laser Science Laboratory, Toyota Technological Institute, 2-12-1 Hisakata, Tempaku-ku, Nagoya, 468-8511, Japan
- Contributed equally
| |
Collapse
|
164
|
Johnson E, Clark M, Oncul M, Pantiru A, MacLean C, Deuchars J, Deuchars SA, Johnston J. Graded spikes differentially signal neurotransmitter input in cerebrospinal fluid contacting neurons of the mouse spinal cord. iScience 2022; 26:105914. [PMID: 36691620 PMCID: PMC9860393 DOI: 10.1016/j.isci.2022.105914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/06/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
The action potential and its all-or-none nature is fundamental to neural communication. Canonically, the action potential is initiated once voltage-activated Na+ channels are activated, and their rapid kinetics of activation and inactivation give rise to the action potential's all-or-none nature. Here we demonstrate that cerebrospinal fluid contacting neurons (CSFcNs) surrounding the central canal of the mouse spinal cord employ a different strategy. Rather than using voltage-activated Na+ channels to generate binary spikes, CSFcNs use two different types of voltage-activated Ca2+ channel, enabling spikes of different amplitude. T-type Ca2+ channels generate small amplitude spikes, whereas larger amplitude spikes require high voltage-activated Cd2+-sensitive Ca2+ channels. We demonstrate that these different amplitude spikes can signal input from different transmitter systems; purinergic inputs evoke smaller T-type dependent spikes whereas cholinergic inputs evoke larger spikes that do not rely on T-type channels. Different synaptic inputs to CSFcNs can therefore be signaled by the spike amplitude.
Collapse
Affiliation(s)
- Emily Johnson
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Marilyn Clark
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Merve Oncul
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Andreea Pantiru
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Claudia MacLean
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jim Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Susan A. Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jamie Johnston
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK,Corresponding author
| |
Collapse
|
165
|
Institoris A, Vandal M, Peringod G, Catalano C, Tran CH, Yu X, Visser F, Breiteneder C, Molina L, Khakh BS, Nguyen MD, Thompson RJ, Gordon GR. Astrocytes amplify neurovascular coupling to sustained activation of neocortex in awake mice. Nat Commun 2022; 13:7872. [PMID: 36550102 PMCID: PMC9780254 DOI: 10.1038/s41467-022-35383-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Functional hyperemia occurs when enhanced neuronal activity signals to increase local cerebral blood flow (CBF) to satisfy regional energy demand. Ca2+ elevation in astrocytes can drive arteriole dilation to increase CBF, yet affirmative evidence for the necessity of astrocytes in functional hyperemia in vivo is lacking. In awake mice, we discovered that functional hyperemia is bimodal with a distinct early and late component whereby arteriole dilation progresses as sensory stimulation is sustained. Clamping astrocyte Ca2+ signaling in vivo by expressing a plasma membrane Ca2+ ATPase (CalEx) reduces sustained but not brief sensory-evoked arteriole dilation. Elevating astrocyte free Ca2+ using chemogenetics selectively augments sustained hyperemia. Antagonizing NMDA-receptors or epoxyeicosatrienoic acid production reduces only the late component of functional hyperemia, leaving brief increases in CBF to sensory stimulation intact. We propose that a fundamental role of astrocyte Ca2+ is to amplify functional hyperemia when neuronal activation is prolonged.
Collapse
Affiliation(s)
- Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Milène Vandal
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Govind Peringod
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Christy Catalano
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Cam Ha Tran
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557-352, USA
| | - Xinzhu Yu
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Molecular and Integrative Physiology, Beckman Institute, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Frank Visser
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Cheryl Breiteneder
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Leonardo Molina
- Hotchkiss Brain Institute, Department of Clinical Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095-1751, USA
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Department of Clinical Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
166
|
Bulovaite E, Qiu Z, Kratschke M, Zgraj A, Fricker DG, Tuck EJ, Gokhale R, Koniaris B, Jami SA, Merino-Serrais P, Husi E, Mendive-Tapia L, Vendrell M, O'Dell TJ, DeFelipe J, Komiyama NH, Holtmaat A, Fransén E, Grant SGN. A brain atlas of synapse protein lifetime across the mouse lifespan. Neuron 2022; 110:4057-4073.e8. [PMID: 36202095 PMCID: PMC9789179 DOI: 10.1016/j.neuron.2022.09.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022]
Abstract
The lifetime of proteins in synapses is important for their signaling, maintenance, and remodeling, and for memory duration. We quantified the lifetime of endogenous PSD95, an abundant postsynaptic protein in excitatory synapses, at single-synapse resolution across the mouse brain and lifespan, generating the Protein Lifetime Synaptome Atlas. Excitatory synapses have a wide range of PSD95 lifetimes extending from hours to several months, with distinct spatial distributions in dendrites, neurons, and brain regions. Synapses with short protein lifetimes are enriched in young animals and in brain regions controlling innate behaviors, whereas synapses with long protein lifetimes accumulate during development, are enriched in the cortex and CA1 where memories are stored, and are preferentially preserved in old age. Synapse protein lifetime increases throughout the brain in a mouse model of autism and schizophrenia. Protein lifetime adds a further layer to synapse diversity and enriches prevailing concepts in brain development, aging, and disease.
Collapse
Affiliation(s)
- Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Maximilian Kratschke
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Adrianna Zgraj
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - David G Fricker
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Eleanor J Tuck
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Babis Koniaris
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; School of Computing, Edinburgh Napier University, Edinburgh EH10 5DT, UK
| | - Shekib A Jami
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paula Merino-Serrais
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, UPM, 28223 Madrid, Spain; Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Elodie Husi
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marc Vendrell
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Thomas J O'Dell
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, UPM, 28223 Madrid, Spain; Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; The Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Anthony Holtmaat
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden; Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
167
|
Tuning instability of non-columnar neurons in the salt-and-pepper whisker map in somatosensory cortex. Nat Commun 2022; 13:6611. [PMID: 36329010 PMCID: PMC9633707 DOI: 10.1038/s41467-022-34261-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Rodent sensory cortex contains salt-and-pepper maps of sensory features, whose structure is not fully known. Here we investigated the structure of the salt-and-pepper whisker somatotopic map among L2/3 pyramidal neurons in somatosensory cortex, in awake mice performing one-vs-all whisker discrimination. Neurons tuned for columnar (CW) and non-columnar (non-CW) whiskers were spatially intermixed, with co-tuned neurons forming local (20 µm) clusters. Whisker tuning was markedly unstable in expert mice, with 35-46% of pyramidal cells significantly shifting tuning over 5-18 days. Tuning instability was highly concentrated in non-CW tuned neurons, and thus was structured in the map. Instability of non-CW neurons was unchanged during chronic whisker paralysis and when mice discriminated individual whiskers, suggesting it is an inherent feature. Thus, L2/3 combines two distinct components: a stable columnar framework of CW-tuned cells that may promote spatial perceptual stability, plus an intermixed, non-columnar surround with highly unstable tuning.
Collapse
|
168
|
Svara F, Förster D, Kubo F, Januszewski M, Dal Maschio M, Schubert PJ, Kornfeld J, Wanner AA, Laurell E, Denk W, Baier H. Automated synapse-level reconstruction of neural circuits in the larval zebrafish brain. Nat Methods 2022; 19:1357-1366. [PMID: 36280717 PMCID: PMC9636024 DOI: 10.1038/s41592-022-01621-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 08/22/2022] [Indexed: 12/29/2022]
Abstract
Dense reconstruction of synaptic connectivity requires high-resolution electron microscopy images of entire brains and tools to efficiently trace neuronal wires across the volume. To generate such a resource, we sectioned and imaged a larval zebrafish brain by serial block-face electron microscopy at a voxel size of 14 × 14 × 25 nm3. We segmented the resulting dataset with the flood-filling network algorithm, automated the detection of chemical synapses and validated the results by comparisons to transmission electron microscopic images and light-microscopic reconstructions. Neurons and their connections are stored in the form of a queryable and expandable digital address book. We reconstructed a network of 208 neurons involved in visual motion processing, most of them located in the pretectum, which had been functionally characterized in the same specimen by two-photon calcium imaging. Moreover, we mapped all 407 presynaptic and postsynaptic partners of two superficial interneurons in the tectum. The resource developed here serves as a foundation for synaptic-resolution circuit analyses in the zebrafish nervous system.
Collapse
Affiliation(s)
- Fabian Svara
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Max Planck Institute for Neurobiology of Behavior - caesar, Bonn, Germany
- ariadne.ai ag, Buchrain, Switzerland
| | - Dominique Förster
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Fumi Kubo
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Center for Frontier Research, National Institute of Genetics, Mishima, Japan
| | | | - Marco Dal Maschio
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Jörgen Kornfeld
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Adrian A Wanner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Paul Scherrer Institute (PSI), Villigen, Switzerland
| | - Eva Laurell
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Winfried Denk
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, Martinsried, Germany.
| |
Collapse
|
169
|
Deep tissue multi-photon imaging using adaptive optics with direct focus sensing and shaping. Nat Biotechnol 2022; 40:1663-1671. [PMID: 35697805 DOI: 10.1038/s41587-022-01343-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 04/29/2022] [Indexed: 12/30/2022]
Abstract
High-resolution optical imaging deep in tissues is challenging because of optical aberrations and scattering of light caused by the complex structure of living matter. Here we present an adaptive optics three-photon microscope based on analog lock-in phase detection for focus sensing and shaping (ALPHA-FSS). ALPHA-FSS accurately measures and effectively compensates for both aberrations and scattering induced by specimens and recovers subcellular resolution at depth. A conjugate adaptive optics configuration with remote focusing enables in vivo imaging of fine neuronal structures in the mouse cortex through the intact skull up to a depth of 750 µm below the pia, enabling near-non-invasive high-resolution microscopy in cortex. Functional calcium imaging with high sensitivity and high-precision laser-mediated microsurgery through the intact skull were also demonstrated. Moreover, we achieved in vivo high-resolution imaging of the deep cortex and subcortical hippocampus up to 1.1 mm below the pia within the intact brain.
Collapse
|
170
|
Ma L, Day-Cooney J, Benavides OJ, Muniak MA, Qin M, Ding JB, Mao T, Zhong H. Locomotion activates PKA through dopamine and adenosine in striatal neurons. Nature 2022; 611:762-768. [PMID: 36352228 PMCID: PMC10752255 DOI: 10.1038/s41586-022-05407-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/03/2022] [Indexed: 11/10/2022]
Abstract
The canonical model of striatal function predicts that animal locomotion is associated with the opposing regulation of protein kinase A (PKA) in direct and indirect pathway striatal spiny projection neurons (SPNs) by dopamine1-7. However, the precise dynamics of PKA in dorsolateral SPNs during locomotion remain to be determined. It is also unclear whether other neuromodulators are involved. Here we show that PKA activity in both types of SPNs is essential for normal locomotion. Using two-photon fluorescence lifetime imaging8-10 of a PKA sensor10 through gradient index lenses, we measured PKA activity within individual SPNs of the mouse dorsolateral striatum during locomotion. Consistent with the canonical view, dopamine activated PKA activity in direct pathway SPNs during locomotion through the dopamine D1 receptor. However, indirect pathway SPNs exhibited a greater increase in PKA activity, which was largely abolished through the blockade of adenosine A2A receptors. In agreement with these results, fibre photometry measurements of an adenosine sensor11 revealed an acute increase in extracellular adenosine during locomotion. Functionally, antagonism of dopamine or adenosine receptors resulted in distinct changes in SPN PKA activity, neuronal activity and locomotion. Together, our results suggest that acute adenosine accumulation interplays with dopamine release to orchestrate PKA activity in SPNs and proper striatal function during animal locomotion.
Collapse
Affiliation(s)
- Lei Ma
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Julian Day-Cooney
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Omar Jáidar Benavides
- Department of Neurosurgery and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael A Muniak
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jun B Ding
- Department of Neurosurgery and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
171
|
Sensitive genetically encoded sensors for population and subcellular imaging of cAMP in vivo. Nat Methods 2022; 19:1461-1471. [PMID: 36303019 PMCID: PMC10171401 DOI: 10.1038/s41592-022-01646-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/09/2022] [Indexed: 01/07/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) signaling integrates information from diverse G-protein-coupled receptors, such as neuromodulator receptors, to regulate pivotal biological processes in a cellular-specific and subcellular-specific manner. However, in vivo cellular-resolution imaging of cAMP dynamics remains challenging. Here, we screen existing genetically encoded cAMP sensors and further develop the best performer to derive three improved variants, called cAMPFIREs. Compared with their parental sensor, these sensors exhibit up to 10-fold increased sensitivity to cAMP and a cytosolic distribution. cAMPFIREs are compatible with both ratiometric and fluorescence lifetime imaging and can detect cAMP dynamics elicited by norepinephrine at physiologically relevant, nanomolar concentrations. Imaging of cAMPFIREs in awake mice reveals tonic levels of cAMP in cortical neurons that are associated with wakefulness, modulated by opioids, and differentially regulated across subcellular compartments. Furthermore, enforced locomotion elicits neuron-specific, bidirectional cAMP dynamics. cAMPFIREs also function in Drosophila. Overall, cAMPFIREs may have broad applicability for studying intracellular signaling in vivo.
Collapse
|
172
|
Meyer E, Rieder P, Gobbo D, Candido G, Scheller A, de Oliveira RMW, Kirchhoff F. Cannabidiol Exerts a Neuroprotective and Glia-Balancing Effect in the Subacute Phase of Stroke. Int J Mol Sci 2022; 23:12886. [PMID: 36361675 PMCID: PMC9659180 DOI: 10.3390/ijms232112886] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022] Open
Abstract
Pharmacological agents limiting secondary tissue loss and improving functional outcomes after stroke are still limited. Cannabidiol (CBD), the major non-psychoactive component of Cannabis sativa, has been proposed as a neuroprotective agent against experimental cerebral ischemia. The effects of CBD mostly relate to the modulation of neuroinflammation, including glial activation. To investigate the effects of CBD on glial cells after focal ischemia in vivo, we performed time-lapse imaging of microglia and astroglial Ca2+ signaling in the somatosensory cortex in the subacute phase of stroke by in vivo two-photon laser-scanning microscopy using transgenic mice with microglial EGFP expression and astrocyte-specific expression of the genetically encoded Ca2+ sensor GCaMP3. CBD (10 mg/kg, intraperitoneally) prevented ischemia-induced neurological impairment, reducing the neurological deficit score from 2.0 ± 1.2 to 0.8 ± 0.8, and protected against neurodegeneration, as shown by the reduction (more than 70%) in Fluoro-Jade C staining (18.8 ± 7.5 to 5.3 ± 0.3). CBD reduced ischemia-induced microglial activation assessed by changes in soma area and total branch length, and exerted a balancing effect on astroglial Ca2+ signals. Our findings indicate that the neuroprotective effects of CBD may occur in the subacute phase of ischemia, and reinforce its strong anti-inflammatory property. Nevertheless, its mechanism of action on glial cells still requires further studies.
Collapse
Affiliation(s)
- Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Phillip Rieder
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Gabriella Candido
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Rúbia Maria Weffort de Oliveira
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| |
Collapse
|
173
|
Zhu H, Shen W, Luo C, Liu F. An integrated microfluidic device for multiplexed imaging of spatial gene expression patterns of Drosophila embryos. LAB ON A CHIP 2022; 22:4081-4092. [PMID: 36165088 DOI: 10.1039/d2lc00514j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
To reveal the underlying mechanism of the biological function of multicellular systems, it is important to obtain comprehensive spatial gene expression profiles. Among the emerging single-cell spatial-omics techniques, immunofluorescence (IF)-based iterative multiplexed imaging is a promising approach. However, the conventional method is usually costly, time-consuming, labor-intensive, and has low throughput. Moreover, it has yet to be demonstrated in intact multicellular organisms. Here, we developed an integrated microfluidic system to overcome these challenges for quantitatively measuring multiple protein profiles sequentially in situ in the same Drosophila embryo. We designed an array of hydrodynamic trapping sites to automatically capture over ten Drosophila embryos with orientation selectivity at more than 90% trapping rates. We also optimized the geometry of confinement and the on-chip IF protocol to achieve the same high signal-to-noise ratio as the off-chip traditional IF experiments. Moreover, we developed an efficient de-staining protocol by combining on-chip antibody stripping and fluorophore bleaching. Using the same secondary antibody to sequentially stain different genes, we confirmed that the de-stained genes have no detectable interference with the subsequently stained genes, and the gene expression profiles are preserved after multiple cycles of staining and de-staining processes. This preliminary test shows that our newly developed integrated microfluidic system can be a powerful tool for multiplexed imaging of Drosophila embryos. Our work opens a new avenue to design microfluidic chips for multicellular organisms and single-cell spatial-omics techniques.
Collapse
Affiliation(s)
- Hongcun Zhu
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, China.
| | - Wenting Shen
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, China.
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou, Zhejiang, China
| | - Feng Liu
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Health Science & Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China
| |
Collapse
|
174
|
Dürst CD, Wiegert JS, Schulze C, Helassa N, Török K, Oertner TG. Vesicular release probability sets the strength of individual Schaffer collateral synapses. Nat Commun 2022; 13:6126. [PMID: 36253353 PMCID: PMC9576736 DOI: 10.1038/s41467-022-33565-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 09/22/2022] [Indexed: 12/24/2022] Open
Abstract
Information processing in the brain is controlled by quantal release of neurotransmitters, a tightly regulated process. From ultrastructural analysis, it is known that presynaptic boutons along single axons differ in the number of vesicles docked at the active zone. It is not clear whether the probability of these vesicles to get released (pves) is homogenous or also varies between individual boutons. Here, we optically measure evoked transmitter release at individual Schaffer collateral synapses at different calcium concentrations, using the genetically encoded glutamate sensor iGluSnFR. Fitting a binomial model to measured response amplitude distributions allowed us to extract the quantal parameters N, pves, and q. We find that Schaffer collateral boutons typically release single vesicles under low pves conditions and switch to multivesicular release in high calcium saline. The potency of individual boutons is highly correlated with their vesicular release probability while the number of releasable vesicles affects synaptic output only under high pves conditions.
Collapse
Affiliation(s)
- Céline D Dürst
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
- Department of Basic Neurosciences, Center for Neurosciences (CMU), University of Geneva, 1211, Geneva, Switzerland
| | - J Simon Wiegert
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
| | - Christian Schulze
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
| | - Nordine Helassa
- Cell Biology and Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, SW17 0RE, UK
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Katalin Török
- Cell Biology and Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, SW17 0RE, UK
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany.
| |
Collapse
|
175
|
Meyer DJ, Díaz-García CM, Nathwani N, Rahman M, Yellen G. The Na +/K + pump dominates control of glycolysis in hippocampal dentate granule cells. eLife 2022; 11:e81645. [PMID: 36222651 PMCID: PMC9592084 DOI: 10.7554/elife.81645] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular ATP that is consumed to perform energetically expensive tasks must be replenished by new ATP through the activation of metabolism. Neuronal stimulation, an energetically demanding process, transiently activates aerobic glycolysis, but the precise mechanism underlying this glycolysis activation has not been determined. We previously showed that neuronal glycolysis is correlated with Ca2+ influx, but is not activated by feedforward Ca2+ signaling (Díaz-García et al., 2021a). Since ATP-powered Na+ and Ca2+ pumping activities are increased following stimulation to restore ion gradients and are estimated to consume most neuronal ATP, we aimed to determine if they are coupled to neuronal glycolysis activation. By using two-photon imaging of fluorescent biosensors and dyes in dentate granule cell somas of acute mouse hippocampal slices, we observed that production of cytoplasmic NADH, a byproduct of glycolysis, is strongly coupled to changes in intracellular Na+, while intracellular Ca2+ could only increase NADH production if both forward Na+/Ca2+ exchange and Na+/K+ pump activity were intact. Additionally, antidromic stimulation-induced intracellular [Na+] increases were reduced >50% by blocking Ca2+ entry. These results indicate that neuronal glycolysis activation is predominantly a response to an increase in activity of the Na+/K+ pump, which is strongly potentiated by Na+ influx through the Na+/Ca2+ exchanger during extrusion of Ca2+ following stimulation.
Collapse
Affiliation(s)
- Dylan J Meyer
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | | | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mahia Rahman
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
176
|
Geng J, Tang Y, Yu Z, Gao Y, Li W, Lu Y, Wang B, Zhou H, Li P, Liu N, Wang P, Fan Y, Yang Y, Guo ZV, Liu X. Chronic Ca 2+ imaging of cortical neurons with long-term expression of GCaMP-X. eLife 2022; 11:e76691. [PMID: 36196992 PMCID: PMC9699699 DOI: 10.7554/elife.76691] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Dynamic Ca2+ signals reflect acute changes in membrane excitability, and also mediate signaling cascades in chronic processes. In both cases, chronic Ca2+ imaging is often desired, but challenged by the cytotoxicity intrinsic to calmodulin (CaM)-based GCaMP, a series of genetically-encoded Ca2+ indicators that have been widely applied. Here, we demonstrate the performance of GCaMP-X in chronic Ca2+ imaging of cortical neurons, where GCaMP-X by design is to eliminate the unwanted interactions between the conventional GCaMP and endogenous (apo)CaM-binding proteins. By expressing in adult mice at high levels over an extended time frame, GCaMP-X showed less damage and improved performance in two-photon imaging of sensory (whisker-deflection) responses or spontaneous Ca2+ fluctuations, in comparison with GCaMP. Chronic Ca2+ imaging of one month or longer was conducted for cultured cortical neurons expressing GCaMP-X, unveiling that spontaneous/local Ca2+ transients progressively developed into autonomous/global Ca2+ oscillations. Along with the morphological indices of neurite length and soma size, the major metrics of oscillatory Ca2+, including rate, amplitude and synchrony were also examined. Dysregulations of both neuritogenesis and Ca2+ oscillations became discernible around 2-3 weeks after virus injection or drug induction to express GCaMP in newborn or mature neurons, which were exacerbated by stronger or prolonged expression of GCaMP. In contrast, neurons expressing GCaMP-X were significantly less damaged or perturbed, altogether highlighting the unique importance of oscillatory Ca2+ to neural development and neuronal health. In summary, GCaMP-X provides a viable solution for Ca2+ imaging applications involving long-time and/or high-level expression of Ca2+ probes.
Collapse
Affiliation(s)
- Jinli Geng
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Yingjun Tang
- Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Medicine, Tsinghua UniversityBeijingChina
| | - Zhen Yu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Yunming Gao
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Wenxiang Li
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Yitong Lu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Bo Wang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| | - Huiming Zhou
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
- Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Medicine, Tsinghua UniversityBeijingChina
| | - Ping Li
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
| | - Nan Liu
- Center for Life Sciences, School of Life Sciences, Yunnan UniversityKunmingChina
| | - Ping Wang
- Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang UniversityHangzhouChina
| | - Yubo Fan
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
| | - Yaxiong Yang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
| | - Zengcai V Guo
- Tsinghua-Peking Joint Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Medicine, Tsinghua UniversityBeijingChina
| | - Xiaodong Liu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang UniversityBeijingChina
- X-Laboratory for Ion-Channel Engineering, Beihang UniversityBeijingChina
| |
Collapse
|
177
|
Rübel O, Tritt A, Ly R, Dichter BK, Ghosh S, Niu L, Baker P, Soltesz I, Ng L, Svoboda K, Frank L, Bouchard KE. The Neurodata Without Borders ecosystem for neurophysiological data science. eLife 2022; 11:e78362. [PMID: 36193886 PMCID: PMC9531949 DOI: 10.7554/elife.78362] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/13/2022] [Indexed: 01/21/2023] Open
Abstract
The neurophysiology of cells and tissues are monitored electrophysiologically and optically in diverse experiments and species, ranging from flies to humans. Understanding the brain requires integration of data across this diversity, and thus these data must be findable, accessible, interoperable, and reusable (FAIR). This requires a standard language for data and metadata that can coevolve with neuroscience. We describe design and implementation principles for a language for neurophysiology data. Our open-source software (Neurodata Without Borders, NWB) defines and modularizes the interdependent, yet separable, components of a data language. We demonstrate NWB's impact through unified description of neurophysiology data across diverse modalities and species. NWB exists in an ecosystem, which includes data management, analysis, visualization, and archive tools. Thus, the NWB data language enables reproduction, interchange, and reuse of diverse neurophysiology data. More broadly, the design principles of NWB are generally applicable to enhance discovery across biology through data FAIRness.
Collapse
Affiliation(s)
- Oliver Rübel
- Scientific Data Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Andrew Tritt
- Applied Mathematics and Computational Research Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | - Ryan Ly
- Scientific Data Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
| | | | - Satrajit Ghosh
- McGovern Institute for Brain Research, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical SchoolBostonUnited States
| | | | - Pamela Baker
- Allen Institute for Brain ScienceSeattleUnited States
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford UniversityStanfordUnited States
| | - Lydia Ng
- Allen Institute for Brain ScienceSeattleUnited States
| | - Karel Svoboda
- Allen Institute for Brain ScienceSeattleUnited States
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Loren Frank
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Kavli Institute for Fundamental NeuroscienceSan FranciscoUnited States
- Departments of Physiology and Psychiatry University of California, San FranciscoSan FranciscoUnited States
| | - Kristofer E Bouchard
- Scientific Data Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Kavli Institute for Fundamental NeuroscienceSan FranciscoUnited States
- Biological Systems and Engineering Division, Lawrence Berkeley National LaboratoryBerkeleyUnited States
- Helen Wills Neuroscience Institute and Redwood Center for Theoretical Neuroscience, University of California, BerkeleyBerkeleyUnited States
- Weill NeurohubBerkeleyUnited States
| |
Collapse
|
178
|
Park A, Croset V, Otto N, Agarwal D, Treiber CD, Meschi E, Sims D, Waddell S. Gliotransmission of D-serine promotes thirst-directed behaviors in Drosophila. Curr Biol 2022; 32:3952-3970.e8. [PMID: 35963239 PMCID: PMC9616736 DOI: 10.1016/j.cub.2022.07.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/04/2022] [Accepted: 07/15/2022] [Indexed: 12/13/2022]
Abstract
Thirst emerges from a range of cellular changes that ultimately motivate an animal to consume water. Although thirst-responsive neuronal signals have been reported, the full complement of brain responses is unclear. Here, we identify molecular and cellular adaptations in the brain using single-cell sequencing of water-deprived Drosophila. Water deficiency primarily altered the glial transcriptome. Screening the regulated genes revealed astrocytic expression of the astray-encoded phosphoserine phosphatase to bi-directionally regulate water consumption. Astray synthesizes the gliotransmitter D-serine, and vesicular release from astrocytes is required for drinking. Moreover, dietary D-serine rescues aay-dependent drinking deficits while facilitating water consumption and expression of water-seeking memory. D-serine action requires binding to neuronal NMDA-type glutamate receptors. Fly astrocytes contribute processes to tripartite synapses, and the proportion of astrocytes that are themselves activated by glutamate increases with water deprivation. We propose that thirst elevates astrocytic D-serine release, which awakens quiescent glutamatergic circuits to enhance water procurement.
Collapse
Affiliation(s)
- Annie Park
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Vincent Croset
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK; Department of Biosciences, Durham University, Durham DH1 3LE, UK.
| | - Nils Otto
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Devika Agarwal
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK; MRC Computational Genomics Analysis and Training Programme (CGAT), MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Christoph D Treiber
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Eleonora Meschi
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - David Sims
- MRC Computational Genomics Analysis and Training Programme (CGAT), MRC Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Scott Waddell
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK.
| |
Collapse
|
179
|
Lee JJ, Krumin M, Harris KD, Carandini M. Task specificity in mouse parietal cortex. Neuron 2022; 110:2961-2969.e5. [PMID: 35963238 PMCID: PMC9616730 DOI: 10.1016/j.neuron.2022.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/16/2022] [Accepted: 07/15/2022] [Indexed: 11/26/2022]
Abstract
Parietal cortex is implicated in a variety of behavioral processes, but it is unknown whether and how its individual neurons participate in multiple tasks. We trained head-fixed mice to perform two visual decision tasks involving a steering wheel or a virtual T-maze and recorded from the same parietal neurons during these two tasks. Neurons that were active during the T-maze task were typically inactive during the steering-wheel task and vice versa. Recording from the same neurons in the same apparatus without task stimuli yielded the same specificity as in the task, suggesting that task specificity depends on physical context. To confirm this, we trained some mice in a third task combining the steering wheel context with the visual environment of the T-maze. This hybrid task engaged the same neurons as those engaged in the steering-wheel task. Thus, participation by neurons in mouse parietal cortex is task specific, and this specificity is determined by physical context.
Collapse
Affiliation(s)
- Julie J Lee
- UCL Institute of Ophthalmology, University College London, Gower Street, London WC1E 6AE, UK.
| | - Michael Krumin
- UCL Institute of Ophthalmology, University College London, Gower Street, London WC1E 6AE, UK
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, Gower Street, London WC1E 6AE, UK
| | - Matteo Carandini
- UCL Institute of Ophthalmology, University College London, Gower Street, London WC1E 6AE, UK
| |
Collapse
|
180
|
Gonzalez-Suarez AD, Zavatone-Veth JA, Chen J, Matulis CA, Badwan BA, Clark DA. Excitatory and inhibitory neural dynamics jointly tune motion detection. Curr Biol 2022; 32:3659-3675.e8. [PMID: 35868321 PMCID: PMC9474608 DOI: 10.1016/j.cub.2022.06.075] [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: 01/09/2022] [Revised: 05/03/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022]
Abstract
Neurons integrate excitatory and inhibitory signals to produce their outputs, but the role of input timing in this integration remains poorly understood. Motion detection is a paradigmatic example of this integration, since theories of motion detection rely on different delays in visual signals. These delays allow circuits to compare scenes at different times to calculate the direction and speed of motion. Different motion detection circuits have different velocity sensitivity, but it remains untested how the response dynamics of individual cell types drive this tuning. Here, we sped up or slowed down specific neuron types in Drosophila's motion detection circuit by manipulating ion channel expression. Altering the dynamics of individual neuron types upstream of motion detectors increased their sensitivity to fast or slow visual motion, exposing distinct roles for excitatory and inhibitory dynamics in tuning directional signals, including a role for the amacrine cell CT1. A circuit model constrained by functional data and anatomy qualitatively reproduced the observed tuning changes. Overall, these results reveal how excitatory and inhibitory dynamics together tune a canonical circuit computation.
Collapse
Affiliation(s)
| | - Jacob A Zavatone-Veth
- Department of Physics, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Juyue Chen
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | | | - Bara A Badwan
- School of Engineering and Applied Science, Yale University, New Haven, CT 06511, USA
| | - Damon A Clark
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA; Department of Physics, Yale University, New Haven, CT 06511, USA; Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06511, USA; Department of Neuroscience, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
181
|
Hwang FJ, Roth RH, Wu YW, Sun Y, Kwon DK, Liu Y, Ding JB. Motor learning selectively strengthens cortical and striatal synapses of motor engram neurons. Neuron 2022; 110:2790-2801.e5. [PMID: 35809573 PMCID: PMC9464700 DOI: 10.1016/j.neuron.2022.06.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/21/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022]
Abstract
Learning and consolidation of new motor skills require plasticity in the motor cortex and striatum, two key motor regions of the brain. However, how neurons undergo synaptic changes and become recruited during motor learning to form a memory engram remains unknown. Here, we train mice on a motor learning task and use a genetic approach to identify and manipulate behavior-relevant neurons selectively in the primary motor cortex (M1). We find that the degree of M1 engram neuron reactivation correlates with motor performance. We further demonstrate that learning-induced dendritic spine reorganization specifically occurs in these M1 engram neurons. In addition, we find that motor learning leads to an increase in the strength of M1 engram neuron outputs onto striatal spiny projection neurons (SPNs) and that these synapses form clusters along SPN dendrites. These results identify a highly specific synaptic plasticity during the formation of long-lasting motor memory traces in the corticostriatal circuit.
Collapse
Affiliation(s)
- Fuu-Jiun Hwang
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yu-Wei Wu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yue Sun
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Destany K Kwon
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Yu Liu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
182
|
Lai HM, Tang Y, Lau ZYH, Campbell RAA, Yau JCN, Chan CCY, Chan DCW, Wong TY, Wong HKT, Yan LYC, Wu WKK, Wong SH, Kwok KW, Wing YK, Lam HHN, Ng HK, Mrsic-Flogel TD, Mok VCT, Chan JYK, Ko H. Antibody stabilization for thermally accelerated deep immunostaining. Nat Methods 2022; 19:1137-1146. [PMID: 36050489 PMCID: PMC9467915 DOI: 10.1038/s41592-022-01569-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/27/2022] [Indexed: 01/02/2023]
Abstract
Antibodies have diverse applications due to their high reaction specificities but are sensitive to denaturation when a higher working temperature is required. We have developed a simple, highly scalable and generalizable chemical approach for stabilizing off-the-shelf antibodies against thermal and chemical denaturation. We demonstrate that the stabilized antibodies (termed SPEARs) can withstand up to 4 weeks of continuous heating at 55 °C and harsh denaturants, and apply our method to 33 tested antibodies. SPEARs enable flexible applications of thermocycling and denaturants to dynamically modulate their binding kinetics, reaction equilibrium, macromolecular diffusivity and aggregation propensity. In particular, we show that SPEARs permit the use of a thermally facilitated three-dimensional immunolabeling strategy (termed ThICK staining), achieving whole mouse brain immunolabeling within 72 h, as well as nearly fourfold deeper penetration with threefold less antibodies in human brain tissue. With faster deep-tissue immunolabeling and broad compatibility with tissue processing and clearing methods without the need for any specialized equipment, we anticipate the wide applicability of ThICK staining with SPEARs for deep immunostaining.
Collapse
Grants
- Croucher Innovation Award (CIA20CU01) from the Croucher Foundation Faculty Innovation Awards (FIA2017/B/01) from the Faculty of Medicine, CUHK Gerald Choa Neuroscience Centre, the Margaret K. L. Cheung Research Centre for Parkinsonism Management, Faculty of Medicine, CUHK Midstream Research Programme for Universities (MRP/048/20) of Innovation and Technology Council (ITC) of Hong Kong Collaborative Research Fund (C6027-19GF & C7074-21GF) and the Area of Excellence Scheme (AoE/M-604/16) of the UGC of Hong Kong Excellent Young Scientists Fund from the National Natural Science Foundation of China
- Midstream Research Programme for Universities (MRP/048/20) of the Innovation and Technology Council (ITC) of Hong Kong Faculty Innovation Awards (FIA2020/B/01) from the Faculty of Medicine, CUHK Gerald Choa Neuroscience Centre, the Margaret K. L. Cheung Research Centre for Parkinsonism Management, Faculty of Medicine, CUHK
- Innovation and Technology Support Programme (ITS/435/18FX) of the ITC of Hong Kong
- Innovation and Technology Support Programme (ITS/435/18FX) of the ITC of Hong Kong General Research Fund (GRF14108818) of the University Grants Committee (UGC) of Hong Kong
- Gerald Choa Neuroscience Centre, the Margaret K. L. Cheung Research Centre for Parkinsonism Management, Faculty of Medicine, CUHK
Collapse
Affiliation(s)
- Hei Ming Lai
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Yumi Tang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zachary Y H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Robert A A Campbell
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Juno C N Yau
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Caleb C Y Chan
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Danny C W Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Shatin, Hong Kong
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Tin Yan Wong
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Harriet K T Wong
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Leo Y C Yan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Shatin, Hong Kong
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Sunny H Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Nanyang Avenue, Singapore
| | - Ka-Wai Kwok
- Department of Mechanical Engineering, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yun-Kwok Wing
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Henry H N Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Thomas D Mrsic-Flogel
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Vincent C T Mok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jason Y K Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Ko
- Department of Psychiatry, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Gerald Choa Neuroscience Centre, The Chinese University of Hong Kong, Shatin, Hong Kong.
- Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
183
|
Yuan Y, Lu F. A Flexible Chamber for Time-Lapse Live-Cell Imaging with Stimulated Raman Scattering Microscopy. J Vis Exp 2022:10.3791/64449. [PMID: 36121285 PMCID: PMC9631341 DOI: 10.3791/64449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2024] Open
Abstract
Stimulated Raman scattering (SRS) microscopy is a label-free chemical imaging technology. Live-cell imaging with SRS has been demonstrated for many biological and biomedical applications. However, long-term time-lapse SRS imaging of live cells has not been widely adopted. SRS microscopy often uses a high numerical aperture (NA) water-immersion objective and a high NA oil-immersion condenser to achieve high-resolution imaging. In this case, the gap between the objective and the condenser is only a few millimeters. Therefore, most commercial stage-top environmental chambers cannot be used for SRS imaging because of their large thickness with a rigid glass cover. This paper describes the design and fabrication of a flexible chamber that can be used for time-lapse live-cell imaging with transmitted SRS signal detection on an upright microscope frame. The flexibility of the chamber is achieved by using a soft material - a thin natural rubber film. The new enclosure and chamber design can be easily added to an existing SRS imaging setup. The testing and preliminary results demonstrate that the flexible chamber system enables stable, long-term, time-lapse SRS imaging of live cells, which can be used for various bioimaging applications in the future.
Collapse
Affiliation(s)
- Yuhao Yuan
- Department of Biomedical Engineering, Binghamton University, State University of New York
| | - Fake Lu
- Department of Biomedical Engineering, Binghamton University, State University of New York;
| |
Collapse
|
184
|
Weiler S, Guggiana Nilo D, Bonhoeffer T, Hübener M, Rose T, Scheuss V. Functional and structural features of L2/3 pyramidal cells continuously covary with pial depth in mouse visual cortex. Cereb Cortex 2022; 33:3715-3733. [PMID: 36017976 PMCID: PMC10068292 DOI: 10.1093/cercor/bhac303] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Pyramidal cells of neocortical layer 2/3 (L2/3 PyrCs) integrate signals from numerous brain areas and project throughout the neocortex. These PyrCs show pial depth-dependent functional and structural specializations, indicating participation in different functional microcircuits. However, whether these depth-dependent differences result from separable PyrC subtypes or whether their features display a continuum correlated with pial depth is unknown. Here, we assessed the stimulus selectivity, electrophysiological properties, dendritic morphology, and excitatory and inhibitory connectivity across the depth of L2/3 in the binocular visual cortex of mice. We find that the apical, but not the basal dendritic tree structure, varies with pial depth, which is accompanied by variation in subthreshold electrophysiological properties. Lower L2/3 PyrCs receive increased input from L4, while upper L2/3 PyrCs receive a larger proportion of intralaminar input. In vivo calcium imaging revealed a systematic change in visual responsiveness, with deeper PyrCs showing more robust responses than superficial PyrCs. Furthermore, deeper PyrCs are more driven by contralateral than ipsilateral eye stimulation. Importantly, the property value transitions are gradual, and L2/3 PyrCs do not display discrete subtypes based on these parameters. Therefore, L2/3 PyrCs' multiple functional and structural properties systematically correlate with their depth, forming a continuum rather than discrete subtypes.
Collapse
Affiliation(s)
- Simon Weiler
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Großhaderner Str. 2, Planegg 82152, Germany.,Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, 25 Howland Street, London W1T 4JG, United Kingdom
| | - Drago Guggiana Nilo
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Max Planck Institute for Biological Intelligence, in foundation, Martinsried, Germany
| | - Tobias Bonhoeffer
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Max Planck Institute for Biological Intelligence, in foundation, Martinsried, Germany
| | - Mark Hübener
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Max Planck Institute for Biological Intelligence, in foundation, Martinsried, Germany
| | - Tobias Rose
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Institute for Experimental Epileptology and Cognition Research, University of Bonn, Venusberg-Campus 1, Bonn 53127, Germany
| | - Volker Scheuss
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, Martinsried 82152, Germany.,Department of Psychiatry, Ludwig-Maximilians-Universität München, Nussbaumstr. 7, München 80336, Germany
| |
Collapse
|
185
|
Spampinato GLB, Ronzitti E, Zampini V, Ferrari U, Trapani F, Khabou H, Agraval A, Dalkara D, Picaud S, Papagiakoumou E, Marre O, Emiliani V. All-optical inter-layers functional connectivity investigation in the mouse retina. CELL REPORTS METHODS 2022; 2:100268. [PMID: 36046629 PMCID: PMC9421538 DOI: 10.1016/j.crmeth.2022.100268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/03/2022] [Accepted: 07/19/2022] [Indexed: 06/01/2023]
Abstract
We developed a multi-unit microscope for all-optical inter-layers circuits interrogation. The system performs two-photon (2P) functional imaging and 2P multiplexed holographic optogenetics at axially distinct planes. We demonstrated the capability of the system to map, in the mouse retina, the functional connectivity between rod bipolar cells (RBCs) and ganglion cells (GCs) by activating single or defined groups of RBCs while recording the evoked response in the GC layer with cell-type specificity and single-cell resolution. We then used a logistic model to probe the functional connectivity between cell types by deriving the "cellular receptive field" describing how RBCs impact each GC type. With the capability to simultaneously image and control neuronal activity at axially distinct planes, the system enables a precise interrogation of multi-layered circuits. Understanding this information transfer is a promising avenue to dissect complex neural circuits and understand the neural basis of computations.
Collapse
Affiliation(s)
| | - Emiliano Ronzitti
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Valeria Zampini
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Ulisse Ferrari
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Francesco Trapani
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Hanen Khabou
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | | | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | | | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Valentina Emiliani
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| |
Collapse
|
186
|
DePiero VJ, Borghuis BG. Phase advancing is a common property of multiple neuron classes in the mouse retina. eNeuro 2022; 9:ENEURO.0270-22.2022. [PMID: 35995559 PMCID: PMC9450563 DOI: 10.1523/eneuro.0270-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Behavioral interactions with moving objects are challenged by response latencies within the sensory and motor nervous systems. In vision, the combined latency from phototransduction and synaptic transmission from the retina to central visual areas amounts to 50-100 ms, depending on stimulus conditions. Time required for generating appropriate motor output adds to this latency and further compounds the behavioral delay. Neuronal adaptations that help counter sensory latency within the retina have been demonstrated in some species, but how general these specializations are, and where in the circuitry they originate, remains unclear. To address this, we studied the timing of object motion-evoked responses at multiple signaling stages within the mouse retina using two-photon fluorescence calcium and glutamate imaging, targeted whole-cell electrophysiology, and computational modeling. We found that both ON and OFF-type ganglion cells, as well as the bipolar cells that innervate them, temporally advance the position encoding of a moving object and so help counter the inherent signaling delay in the retina. Model simulations show that this predictive capability is a direct consequence of the spatial extent of the cells' linear visual receptive field, with no apparent specialized circuits that help predict beyond it.Significance StatementSignal transduction and synaptic transmission within sensory signaling pathways costs time. Not a lot of time, just tens to a few hundred milliseconds depending on the sensory system, but enough to challenge fast behavioral interactions under dynamic stimulus conditions, like catching a moving fly. To counter neuronal delays, nervous systems of many species use anticipatory mechanisms. One such mechanism in the mammalian visual system helps predict the future position of a moving target through a process called phase advancing. Here we ask for functionally diverse neuron populations in the mouse retina how common is phase advancing and demonstrate that it is common and generated at multiple signaling stages.
Collapse
Affiliation(s)
- Victor J DePiero
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Bart G Borghuis
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
187
|
Moroni M, Brondi M, Fellin T, Panzeri S. SmaRT2P: a software for generating and processing smart line recording trajectories for population two-photon calcium imaging. Brain Inform 2022; 9:18. [PMID: 35927517 PMCID: PMC9352634 DOI: 10.1186/s40708-022-00166-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022] Open
Abstract
Two-photon fluorescence calcium imaging allows recording the activity of large neural populations with subcellular spatial resolution, but it is typically characterized by low signal-to-noise ratio (SNR) and poor accuracy in detecting single or few action potentials when large number of neurons are imaged. We recently showed that implementing a smart line scanning approach using trajectories that optimally sample the regions of interest increases both the SNR fluorescence signals and the accuracy of single spike detection in population imaging in vivo. However, smart line scanning requires highly specialised software to design recording trajectories, interface with acquisition hardware, and efficiently process acquired data. Furthermore, smart line scanning needs optimized strategies to cope with movement artefacts and neuropil contamination. Here, we develop and validate SmaRT2P, an open-source, user-friendly and easy-to-interface Matlab-based software environment to perform optimized smart line scanning in two-photon calcium imaging experiments. SmaRT2P is designed to interface with popular acquisition software (e.g., ScanImage) and implements novel strategies to detect motion artefacts, estimate neuropil contamination, and minimize their impact on functional signals extracted from neuronal population imaging. SmaRT2P is structured in a modular way to allow flexibility in the processing pipeline, requiring minimal user intervention in parameter setting. The use of SmaRT2P for smart line scanning has the potential to facilitate the functional investigation of large neuronal populations with increased SNR and accuracy in detecting the discharge of single and few action potentials.
Collapse
Affiliation(s)
- Monica Moroni
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems, UniTn, Istituto Italiano Di Tecnologia, 38068, Rovereto, Italy.
| | - Marco Brondi
- Optical Approaches to Brain Function Laboratory, Istituto Italiano Di Tecnologia, 16163, Genoa, Italy.,Department of Biomedical Sciences-UNIPD, Università Degli Studi Di Padova, 35121, Padua, Italy.,Padova Neuroscience Center (PNC), Università Degli Studi Di Padova, 35129, Padua, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano Di Tecnologia, 16163, Genoa, Italy
| | - Stefano Panzeri
- Neural Computation Laboratory, Center for Neuroscience and Cognitive Systems, UniTn, Istituto Italiano Di Tecnologia, 38068, Rovereto, Italy. .,Department of Excellence for Neural Information Processing, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany.
| |
Collapse
|
188
|
Fieblinger T, Perez-Alvarez A, Lamothe-Molina PJ, Gee CE, Oertner TG. Presynaptic cGMP sets synaptic strength in the striatum and is important for motor learning. EMBO Rep 2022; 23:e54361. [PMID: 35735260 PMCID: PMC9346481 DOI: 10.15252/embr.202154361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
The striatum is a subcortical brain region responsible for the initiation and termination of voluntary movements. Striatal spiny projection neurons receive major excitatory synaptic input from neocortex and thalamus, and cyclic nucleotides have long been known to play important roles in striatal function. Yet, the precise mechanism of action is unclear. Here, we combine optogenetic stimulation, 2‐photon imaging, and genetically encoded scavengers to dissect the regulation of striatal synapses in mice. Our data show that excitatory striatal inputs are tonically depressed by phosphodiesterases (PDEs), in particular PDE1. Blocking PDE activity boosts presynaptic calcium entry and glutamate release, leading to strongly increased synaptic transmission. Although PDE1 degrades both cAMP and cGMP, we uncover that the concentration of cGMP, not cAMP, controls the gain of striatal inputs. Disturbing this gain control mechanism in vivo impairs motor skill learning in mice. The tight dependence of striatal excitatory synapses on PDE1 and cGMP offers a new perspective on the molecular mechanisms regulating striatal activity.
Collapse
Affiliation(s)
- Tim Fieblinger
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alberto Perez-Alvarez
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Rapp OptoElectronic GmbH, Wedel, Germany
| | - Paul J Lamothe-Molina
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine E Gee
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
189
|
Yildirim M, Delepine C, Feldman D, Pham VA, Chou S, Ip J, Nott A, Tsai LH, Ming GL, So PTC, Sur M. Label-free three-photon imaging of intact human cerebral organoids for tracking early events in brain development and deficits in Rett syndrome. eLife 2022; 11:78079. [PMID: 35904330 PMCID: PMC9337854 DOI: 10.7554/elife.78079] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/08/2022] [Indexed: 12/20/2022] Open
Abstract
Human cerebral organoids are unique in their development of progenitor-rich zones akin to ventricular zones from which neuronal progenitors differentiate and migrate radially. Analyses of cerebral organoids thus far have been performed in sectioned tissue or in superficial layers due to their high scattering properties. Here, we demonstrate label-free three-photon imaging of whole, uncleared intact organoids (~2 mm depth) to assess early events of early human brain development. Optimizing a custom-made three-photon microscope to image intact cerebral organoids generated from Rett Syndrome patients, we show defects in the ventricular zone volumetric structure of mutant organoids compared to isogenic control organoids. Long-term imaging live organoids reveals that shorter migration distances and slower migration speeds of mutant radially migrating neurons are associated with more tortuous trajectories. Our label-free imaging system constitutes a particularly useful platform for tracking normal and abnormal development in individual organoids, as well as for screening therapeutic molecules via intact organoid imaging.
Collapse
Affiliation(s)
- Murat Yildirim
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Neuroscience, Cleveland Clinic Lerner Research Institute, Cleveland, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Chloe Delepine
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Danielle Feldman
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Vincent A Pham
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Stephanie Chou
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Jacque Ip
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alexi Nott
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Li-Huei Tsai
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Peter T C So
- Deparment of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Mriganka Sur
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
190
|
Voitov I, Mrsic-Flogel TD. Cortical feedback loops bind distributed representations of working memory. Nature 2022; 608:381-389. [PMID: 35896749 PMCID: PMC9365695 DOI: 10.1038/s41586-022-05014-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 06/22/2022] [Indexed: 11/16/2022]
Abstract
Working memory—the brain’s ability to internalize information and use it flexibly to guide behaviour—is an essential component of cognition. Although activity related to working memory has been observed in several brain regions1–3, how neural populations actually represent working memory4–7 and the mechanisms by which this activity is maintained8–12 remain unclear13–15. Here we describe the neural implementation of visual working memory in mice alternating between a delayed non-match-to-sample task and a simple discrimination task that does not require working memory but has identical stimulus, movement and reward statistics. Transient optogenetic inactivations revealed that distributed areas of the neocortex were required selectively for the maintenance of working memory. Population activity in visual area AM and premotor area M2 during the delay period was dominated by orderly low-dimensional dynamics16,17 that were, however, independent of working memory. Instead, working memory representations were embedded in high-dimensional population activity, present in both cortical areas, persisted throughout the inter-stimulus delay period, and predicted behavioural responses during the working memory task. To test whether the distributed nature of working memory was dependent on reciprocal interactions between cortical regions18–20, we silenced one cortical area (AM or M2) while recording the feedback it received from the other. Transient inactivation of either area led to the selective disruption of inter-areal communication of working memory. Therefore, reciprocally interconnected cortical areas maintain bound high-dimensional representations of working memory. Experiments in mice alternating between a visual working memory task and a task that is independent of working memory provide insight into the neural representation of working memory and the distributed nature of its maintenance.
Collapse
Affiliation(s)
- Ivan Voitov
- Sainsbury Wellcome Centre, University College London, London, UK. .,Biozentrum, University of Basel, Basel, Switzerland.
| | | |
Collapse
|
191
|
Kosillo P, Ahmed KM, Aisenberg EE, Karalis V, Roberts BM, Cragg SJ, Bateup HS. Dopamine neuron morphology and output are differentially controlled by mTORC1 and mTORC2. eLife 2022; 11:e75398. [PMID: 35881440 PMCID: PMC9328766 DOI: 10.7554/elife.75398] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
The mTOR pathway is an essential regulator of cell growth and metabolism. Midbrain dopamine neurons are particularly sensitive to mTOR signaling status as activation or inhibition of mTOR alters their morphology and physiology. mTOR exists in two distinct multiprotein complexes termed mTORC1 and mTORC2. How each of these complexes affect dopamine neuron properties, and whether they have similar or distinct functions is unknown. Here, we investigated this in mice with dopamine neuron-specific deletion of Rptor or Rictor, which encode obligatory components of mTORC1 or mTORC2, respectively. We find that inhibition of mTORC1 strongly and broadly impacts dopamine neuron structure and function causing somatodendritic and axonal hypotrophy, increased intrinsic excitability, decreased dopamine production, and impaired dopamine release. In contrast, inhibition of mTORC2 has more subtle effects, with selective alterations to the output of ventral tegmental area dopamine neurons. Disruption of both mTOR complexes leads to pronounced deficits in dopamine release demonstrating the importance of balanced mTORC1 and mTORC2 signaling for dopaminergic function.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Kamran M Ahmed
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Erin E Aisenberg
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Bradley M Roberts
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Stephanie J Cragg
- Department of Physiology, Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg Biohub, San FranciscoSan FranciscoUnited States
| |
Collapse
|
192
|
Yates JL, Scholl B. Unraveling Functional Diversity of Cortical Synaptic Architecture Through the Lens of Population Coding. Front Synaptic Neurosci 2022; 14:888214. [PMID: 35957943 PMCID: PMC9360921 DOI: 10.3389/fnsyn.2022.888214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/21/2022] [Indexed: 11/15/2022] Open
Abstract
The synaptic inputs to single cortical neurons exhibit substantial diversity in their sensory-driven activity. What this diversity reflects is unclear, and appears counter-productive in generating selective somatic responses to specific stimuli. One possibility is that this diversity reflects the propagation of information from one neural population to another. To test this possibility, we bridge population coding theory with measurements of synaptic inputs recorded in vivo with two-photon calcium imaging. We construct a probabilistic decoder to estimate the stimulus orientation from the responses of a realistic, hypothetical input population of neurons to compare with synaptic inputs onto individual neurons of ferret primary visual cortex (V1) recorded with two-photon calcium imaging in vivo. We find that optimal decoding requires diverse input weights and provides a straightforward mapping from the decoder weights to excitatory synapses. Analytically derived weights for biologically realistic input populations closely matched the functional heterogeneity of dendritic spines imaged in vivo with two-photon calcium imaging. Our results indicate that synaptic diversity is a necessary component of information transmission and reframes studies of connectivity through the lens of probabilistic population codes. These results suggest that the mapping from synaptic inputs to somatic selectivity may not be directly interpretable without considering input covariance and highlights the importance of population codes in pursuit of the cortical connectome.
Collapse
Affiliation(s)
- Jacob L. Yates
- Herbert Wertheim School of Optometry and Vision Science, University of California, Berkeley, Berkeley, CA, United States
| | - Benjamin Scholl
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- *Correspondence: Benjamin Scholl
| |
Collapse
|
193
|
Satou C, Neve RL, Oyibo HK, Zmarz P, Huang KH, Arn Bouldoires E, Mori T, Higashijima SI, Keller GB, Friedrich RW. A viral toolbox for conditional and transneuronal gene expression in zebrafish. eLife 2022; 11:e77153. [PMID: 35866706 PMCID: PMC9307271 DOI: 10.7554/elife.77153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022] Open
Abstract
The zebrafish is an important model in systems neuroscience but viral tools to dissect the structure and function of neuronal circuitry are not established. We developed methods for efficient gene transfer and retrograde tracing in adult and larval zebrafish by herpes simplex viruses (HSV1). HSV1 was combined with the Gal4/UAS system to target cell types with high spatial, temporal, and molecular specificity. We also established methods for efficient transneuronal tracing by modified rabies viruses in zebrafish. We demonstrate that HSV1 and rabies viruses can be used to visualize and manipulate genetically or anatomically identified neurons within and across different brain areas of adult and larval zebrafish. An expandable library of viruses is provided to express fluorescent proteins, calcium indicators, optogenetic probes, toxins and other molecular tools. This toolbox creates new opportunities to interrogate neuronal circuits in zebrafish through combinations of genetic and viral approaches.
Collapse
Affiliation(s)
- Chie Satou
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General HospitalCambridgeUnited States
| | - Hassana K Oyibo
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Pawel Zmarz
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Kuo-Hua Huang
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | | | - Takuma Mori
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu UniversityNaganoJapan
| | - Shin-ichi Higashijima
- National Institutes of Natural Sciences, Exploratory Research Center on Life and Living Systems, National Institute for Basic BiologyOkazakiJapan
- Graduate University for Advanced StudiesOkazakiJapan
| | - Georg B Keller
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Faculty of Natural Sciences, University of BaselBaselSwitzerland
| | - Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Faculty of Natural Sciences, University of BaselBaselSwitzerland
| |
Collapse
|
194
|
Fujiwara T, Brotas M, Chiappe ME. Walking strides direct rapid and flexible recruitment of visual circuits for course control in Drosophila. Neuron 2022; 110:2124-2138.e8. [PMID: 35525243 PMCID: PMC9275417 DOI: 10.1016/j.neuron.2022.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/31/2022] [Accepted: 04/08/2022] [Indexed: 12/19/2022]
Abstract
Flexible mapping between activity in sensory systems and movement parameters is a hallmark of motor control. This flexibility depends on the continuous comparison of short-term postural dynamics and the longer-term goals of an animal, thereby necessitating neural mechanisms that can operate across multiple timescales. To understand how such body-brain interactions emerge across timescales to control movement, we performed whole-cell patch recordings from visual neurons involved in course control in Drosophila. We show that the activity of leg mechanosensory cells, propagating via specific ascending neurons, is critical for stride-by-stride steering adjustments driven by the visual circuit, and, at longer timescales, it provides information about the moving body's state to flexibly recruit the visual circuit for course control. Thus, our findings demonstrate the presence of an elegant stride-based mechanism operating at multiple timescales for context-dependent course control. We propose that this mechanism functions as a general basis for the adaptive control of locomotion.
Collapse
Affiliation(s)
- Terufumi Fujiwara
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal
| | - Margarida Brotas
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal
| | - M Eugenia Chiappe
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal.
| |
Collapse
|
195
|
Bugeon S, Duffield J, Dipoppa M, Ritoux A, Prankerd I, Nicoloutsopoulos D, Orme D, Shinn M, Peng H, Forrest H, Viduolyte A, Reddy CB, Isogai Y, Carandini M, Harris KD. A transcriptomic axis predicts state modulation of cortical interneurons. Nature 2022; 607:330-338. [PMID: 35794483 PMCID: PMC9279161 DOI: 10.1038/s41586-022-04915-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Transcriptomics has revealed that cortical inhibitory neurons exhibit a great diversity of fine molecular subtypes1-6, but it is not known whether these subtypes have correspondingly diverse patterns of activity in the living brain. Here we show that inhibitory subtypes in primary visual cortex (V1) have diverse correlates with brain state, which are organized by a single factor: position along the main axis of transcriptomic variation. We combined in vivo two-photon calcium imaging of mouse V1 with a transcriptomic method to identify mRNA for 72 selected genes in ex vivo slices. We classified inhibitory neurons imaged in layers 1-3 into a three-level hierarchy of 5 subclasses, 11 types and 35 subtypes using previously defined transcriptomic clusters3. Responses to visual stimuli differed significantly only between subclasses, with cells in the Sncg subclass uniformly suppressed, and cells in the other subclasses predominantly excited. Modulation by brain state differed at all hierarchical levels but could be largely predicted from the first transcriptomic principal component, which also predicted correlations with simultaneously recorded cells. Inhibitory subtypes that fired more in resting, oscillatory brain states had a smaller fraction of their axonal projections in layer 1, narrower spikes, lower input resistance and weaker adaptation as determined in vitro7, and expressed more inhibitory cholinergic receptors. Subtypes that fired more during arousal had the opposite properties. Thus, a simple principle may largely explain how diverse inhibitory V1 subtypes shape state-dependent cortical processing.
Collapse
Affiliation(s)
- Stéphane Bugeon
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Joshua Duffield
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Mario Dipoppa
- UCL Queen Square Institute of Neurology, University College London, London, UK
- Center for Theoretical Neuroscience, Columbia University, New York, NY, USA
| | - Anne Ritoux
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Isabelle Prankerd
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - David Orme
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Maxwell Shinn
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Han Peng
- Department of Physics, University of Oxford, Oxford, UK
| | - Hamish Forrest
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Aiste Viduolyte
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Charu Bai Reddy
- UCL Queen Square Institute of Neurology, University College London, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Yoh Isogai
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK
| | - Matteo Carandini
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
196
|
Har-Gil H, Golgher L, Kain D, Blinder P. Versatile software and hardware combo enabling photon counting acquisition and real-time display for multiplexing, 2D and continuous 3D two-photon imaging applications. NEUROPHOTONICS 2022; 9:031920. [PMID: 36159710 PMCID: PMC9487143 DOI: 10.1117/1.nph.9.3.031920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
Significance: rPySight brings a flexible and highly customizable open-software platform built around a powerful multichannel digitizer; combined, it enables performing complex photon counting-based experiments. We exploited advanced programming technology to share the photon counting stream with the graphical processing unit (GPU), making possible real-time display of two-dimensional (2D) and three-dimensional (3D) experiments and paving the road for other real-time applications. Aim: Photon counting improves multiphoton imaging by providing better signal-to-noise ratio in photon-deprived applications and is becoming more widely implemented, as indicated by its increasing presence in many microscopy vendor portfolios. Despite the relatively easy access to this technology offered in commercial systems, these remain limited to one or two channels of data and might not enable highly tailored experiments, forcing most researchers to develop their own electronics and code. We set to develop a flexible and open-source interface to a cutting-edge multichannel fast digitizer that can be easily integrated into existing imaging systems. Approach: We selected an advanced multichannel digitizer capable of generating 70M tags/s and wrote an open software application, based on Rust and Python languages, to share the stream of detected events with the GPU, enabling real-time data processing. Results: rPySight functionality was showcased in real-time monitoring of 2D imaging, improved calcium imaging, multiplexing, and 3D imaging through a varifocal lens. We provide a detailed protocol for implementing out-of-the-box rPySight and its related hardware. Conclusions: Applying photon-counting approaches is becoming a fundamental component in recent technical developments that push well beyond existing acquisition speed limitations of classical multiphoton approaches. Given the performance of rPySight, we foresee its use to capture, among others, the joint dynamics of hundreds (if not thousands) of neuronal and vascular elements across volumes, as is likely required to uncover in a much broader sense the hemodynamic transform function.
Collapse
Affiliation(s)
- Hagai Har-Gil
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
- Tel Aviv University, Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv, Israel
| | - Lior Golgher
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
- Tel Aviv University, Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv, Israel
| | - David Kain
- Tel Aviv University, Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv, Israel
| | - Pablo Blinder
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
- Tel Aviv University, Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv, Israel
| |
Collapse
|
197
|
Hong SZ, Mesik L, Grossman CD, Cohen JY, Lee B, Severin D, Lee HK, Hell JW, Kirkwood A. Norepinephrine potentiates and serotonin depresses visual cortical responses by transforming eligibility traces. Nat Commun 2022; 13:3202. [PMID: 35680879 PMCID: PMC9184610 DOI: 10.1038/s41467-022-30827-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 05/19/2022] [Indexed: 11/18/2022] Open
Abstract
Reinforcement allows organisms to learn which stimuli predict subsequent biological relevance. Hebbian mechanisms of synaptic plasticity are insufficient to account for reinforced learning because neuromodulators signaling biological relevance are delayed with respect to the neural activity associated with the stimulus. A theoretical solution is the concept of eligibility traces (eTraces), silent synaptic processes elicited by activity which upon arrival of a neuromodulator are converted into a lasting change in synaptic strength. Previously we demonstrated in visual cortical slices the Hebbian induction of eTraces and their conversion into LTP and LTD by the retroactive action of norepinephrine and serotonin Here we show in vivo in mouse V1 that the induction of eTraces and their conversion to LTP/D by norepinephrine and serotonin respectively potentiates and depresses visual responses. We also show that the integrity of this process is crucial for ocular dominance plasticity, a canonical model of experience-dependent plasticity.
Collapse
Affiliation(s)
- Su Z Hong
- Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Lukas Mesik
- Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Cooper D Grossman
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Jeremiah Y Cohen
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Boram Lee
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Daniel Severin
- Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Hey-Kyoung Lee
- Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Johannes W Hell
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Alfredo Kirkwood
- Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
198
|
Tanaka R, Clark DA. Neural mechanisms to exploit positional geometry for collision avoidance. Curr Biol 2022; 32:2357-2374.e6. [PMID: 35508172 PMCID: PMC9177691 DOI: 10.1016/j.cub.2022.04.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/21/2022] [Accepted: 04/08/2022] [Indexed: 11/21/2022]
Abstract
Visual motion provides rich geometrical cues about the three-dimensional configuration of the world. However, how brains decode the spatial information carried by motion signals remains poorly understood. Here, we study a collision-avoidance behavior in Drosophila as a simple model of motion-based spatial vision. With simulations and psychophysics, we demonstrate that walking Drosophila exhibit a pattern of slowing to avoid collisions by exploiting the geometry of positional changes of objects on near-collision courses. This behavior requires the visual neuron LPLC1, whose tuning mirrors the behavior and whose activity drives slowing. LPLC1 pools inputs from object and motion detectors, and spatially biased inhibition tunes it to the geometry of collisions. Connectomic analyses identified circuitry downstream of LPLC1 that faithfully inherits its response properties. Overall, our results reveal how a small neural circuit solves a specific spatial vision task by combining distinct visual features to exploit universal geometrical constraints of the visual world.
Collapse
Affiliation(s)
- Ryosuke Tanaka
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | - Damon A Clark
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA; Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA; Department of Physics, Yale University, New Haven, CT 06511, USA; Department of Neuroscience, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
199
|
Erlebach E, Ravotto L, Wyss MT, Condrau J, Troxler T, Vinogradov SA, Weber B. Measurement of cerebral oxygen pressure in living mice by two-photon phosphorescence lifetime microscopy. STAR Protoc 2022; 3:101370. [PMID: 35573482 PMCID: PMC9092998 DOI: 10.1016/j.xpro.2022.101370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The ability to quantify partial pressure of oxygen (pO2) is of primary importance for studies of metabolic processes in health and disease. Here, we present a protocol for imaging of oxygen distributions in tissue and vasculature of the cerebral cortex of anesthetized and awake mice. We describe in vivo two-photon phosphorescence lifetime microscopy (2PLM) of oxygen using the probe Oxyphor 2P. This minimally invasive protocol outperforms existing approaches in terms of accuracy, resolution, and imaging depth. For complete details on the use and execution of this protocol, please refer to Esipova et al. (2019). Two-photon phosphorescence imaging of Oxyphor 2P allows for oxygen measurement in vivo Oxygen imaging can be performed in anesthetized or awake, behaving mice Intravenous injection enables oxygen imaging in the vasculature Cisterna magna injection enables extra- and intravascular oxygen imaging in the brain
Collapse
Affiliation(s)
- Eva Erlebach
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Matthias T. Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Jacqueline Condrau
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
| | - Thomas Troxler
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sergei A. Vinogradov
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding author
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zürich, Switzerland
- Corresponding author
| |
Collapse
|
200
|
Koveal D, Rosen PC, Meyer DJ, Díaz-García CM, Wang Y, Cai LH, Chou PJ, Weitz DA, Yellen G. A high-throughput multiparameter screen for accelerated development and optimization of soluble genetically encoded fluorescent biosensors. Nat Commun 2022; 13:2919. [PMID: 35614105 PMCID: PMC9133083 DOI: 10.1038/s41467-022-30685-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/11/2022] [Indexed: 12/30/2022] Open
Abstract
Genetically encoded fluorescent biosensors are powerful tools used to track chemical processes in intact biological systems. However, the development and optimization of biosensors remains a challenging and labor-intensive process, primarily due to technical limitations of methods for screening candidate biosensors. Here we describe a screening modality that combines droplet microfluidics and automated fluorescence imaging to provide an order of magnitude increase in screening throughput. Moreover, unlike current techniques that are limited to screening for a single biosensor feature at a time (e.g. brightness), our method enables evaluation of multiple features (e.g. contrast, affinity, specificity) in parallel. Because biosensor features can covary, this capability is essential for rapid optimization. We use this system to generate a high-performance biosensor for lactate that can be used to quantify intracellular lactate concentrations. This biosensor, named LiLac, constitutes a significant advance in metabolite sensing and demonstrates the power of our screening approach.
Collapse
Affiliation(s)
- Dorothy Koveal
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Paul C Rosen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dylan J Meyer
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Carlos Manlio Díaz-García
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yongcheng Wang
- Department of Physics and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
| | - Li-Heng Cai
- Department of Physics and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Materials Science and Engineering, University of Virginia, Charlottesville, VA, USA
| | - Peter J Chou
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - David A Weitz
- Department of Physics and John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|