1
|
Wang Q, Baier H. Eat, flee, freeze: Division of labor in the larval zebrafish visuomotor system. Proc Natl Acad Sci U S A 2025; 122:e2506018122. [PMID: 40324097 DOI: 10.1073/pnas.2506018122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025] Open
Affiliation(s)
- Qing Wang
- Department of Genes-Circuits-Behavior, Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| | - Herwig Baier
- Department of Genes-Circuits-Behavior, Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| |
Collapse
|
2
|
Wasser-Bennett G, Brown AR, Maynard SK, Owen SF, Tyler CR. Critical insights into the potential risks of antipsychotic drugs to fish, including through effects on behaviour. Biol Rev Camb Philos Soc 2025. [PMID: 40355132 DOI: 10.1111/brv.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Antipsychotic drugs (APDs) are a diverse class of neuroactive pharmaceuticals increasingly detected in surface and ground waters globally. Some APDs are classified as posing a high environmental risk, due, in part, to their tendency to bioaccumulate in wildlife, including fish. Additional risk drivers for APDs relate to their behavioural effects, potentially impacting fitness outcomes. However, standard ecotoxicological tests used in environmental risk assessment (ERA) do not currently account for these mechanisms. In this review, we critically appraise the environmental risks of APDs to fish. We begin by reading-across from human and mammalian effects data to standard ecotoxicological effects endpoints in fish. We then explore the wide range of behaviours suitable for ecotoxicological assessment of APDs (and other neuroactive) pharmaceuticals, principally through laboratory studies with zebrafish, and assess the potential for using these behavioural phenotypes to predict adverse individual- and population-level outcomes in wild fish, taking into account phenotypic plasticity. Next, we illustrate the advantages and challenges of measuring and applying behavioural endpoints for fish, including within current regulatory risk assessments. In our final analysis, the implications of relying on apical endpoints for ERA of neuroactive drugs (including APDs) are assessed and recommendations provided for the development of a more refined and tailored mechanistic approach, which would enable more robust assessment of their environmental risk(s).
Collapse
Affiliation(s)
- Gabrielle Wasser-Bennett
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| | - A Ross Brown
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| | - Samuel K Maynard
- AstraZeneca, Global Environment, Macclesfield, Cheshire, SK10 2NA, UK
| | - Stewart F Owen
- AstraZeneca, Global Environment, Macclesfield, Cheshire, SK10 2NA, UK
| | - Charles R Tyler
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, Devon, UK
| |
Collapse
|
3
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2025; 298:123-184. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
4
|
Akiyama C, Sakata S, Ono F. Normal locomotion in zebrafish lacking the sodium channel NaV1.4 suggests that the need for muscle action potentials is not universal. PLoS Biol 2025; 23:e3003137. [PMID: 40273189 PMCID: PMC12021243 DOI: 10.1371/journal.pbio.3003137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Extensive studies over decades have firmly established the concept that action potentials (APs) in muscles are indispensable for muscle contraction. To re-examine the significance of APs, we generated zebrafish lacking APs by editing the scn4aa and scn4ab genes, which together encode NaV1.4 (NaVDKO), using the CRISPR-Cas9 system. Surprisingly, the escape response of NaVDKOs to tactile stimuli, both in the embryonic and adult stages, was indistinguishable from that of wild-type (WT) fish. Ca2+ imaging using the calcium indicator protein GCaMP revealed that myofibers isolated from WT fish could be excited by the application of acetylcholine (ACh), even in the presence of tetrodotoxin (TTX) indicating that NaVs are dispensable for skeletal muscle contraction in zebrafish. Mathematical simulations showed that the end-plate potential was able to elicit a change in membrane potential large enough to activate the dihydropyridine receptors of the entire muscle fiber owing to the small fiber size and the disseminated distribution of neuromuscular synapses in both adults and embryos. Our data demonstrate that NaVs are not essential for muscle contraction in zebrafish and that the physiological significance of NaV1.4 in muscle is not uniform across vertebrates.
Collapse
Affiliation(s)
- Chifumi Akiyama
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Souhei Sakata
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Fumihito Ono
- Department of Physiology, Division of Life Sciences, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
5
|
Yamaguchi H, Guagliardo NA, Bell LA, Yamaguchi M, Matsuoka D, Xu F, Smith JP, Diagne M, Almeida LF, Medrano S, Barrett PQ, Nieh EH, Gomez RA, Sequeira-Lopez MLS. Inhibition of Renin Release, a Crucial Event in Homeostasis, is Mediated by Coordinated Calcium Oscillations within Juxtaglomerular Cell Clusters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.629519. [PMID: 39763801 PMCID: PMC11703171 DOI: 10.1101/2024.12.23.629519] [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: 01/15/2025]
Abstract
BACKGROUND Juxtaglomerular (JG) cells are sensors that control blood pressure (BP) and fluid-electrolyte homeostasis. They are arranged as clusters at the tip of each afferent arteriole. In response to a decrease in BP or extracellular fluid volume, JG cells secrete renin, initiating an enzymatic cascade that culminates in the production of angiotensin II (AngII), a potent vasoconstrictor that restores BP and fluid-electrolyte homeostasis. In turn, AngII exerts negative feedback on renin release concomitantly with increased intracellular Ca2+, preventing excessive circulating renin and hypertension. However, within their native structural organization, the intricacies of intracellular Ca2+ signaling dynamics and their sources remain uncharacterized. METHODS We generated mice expressing the JG cell-specific genetically encoded Ca2+ indicator (GCaMP6f) to investigate Ca2+ dynamics within JG cell clusters ex vivo and in vivo. For ex vivo Ca2+ imaging, acutely prepared kidney slices were perfused continuously with a buffer containing variable Ca2+ and AngII concentrations ± Ca2+ channel inhibitors. For in vivo Ca2+ image capture, native mouse kidneys were imaged in situ using multi-photon microscopy with and without AngII administration. ELISA measurements of renin concentrations determined acute renin secretion ex vivo and in vivo, respectively. RESULTS Ex vivo Ca2+ imaging revealed that JG cells exhibit robust and coordinated intracellular oscillatory signals with cell-cell propagation following AngII stimulation. AngII dose-dependently induced stereotypical burst patterns characterized by consecutive Ca2+ spikes, which inversely correlated with renin secretion. Pharmacological channel inhibition identified key sources of these oscillations: endoplasmic reticulum Ca2+ storage and release, extracellular Ca2+ uptake via ORAI channels, and intercellular communication through gap junctions. Blocking ORAI channels and gap junctions reduced AngII inhibitory effect on renin secretion. In vivo Ca2+ imaging demonstrated robust intracellular and intercellular Ca2+ oscillations within JG cell clusters under physiological conditions, exhibiting spike patterns consistent with those measured in ex vivo preparations. Administration of AngII enhanced the Ca2+ oscillatory signals and suppressed acute renin secretion in vivo. CONCLUSION AngII elicits coordinated intracellular and intercellular Ca2+ oscillations within JG cell clusters, ex vivo and in vivo. The effect is driven by endoplasmic reticulum-derived Ca2+ release, ORAI channels, and gap junctions, leading to suppressed renin secretion.
Collapse
Affiliation(s)
- Hiroki Yamaguchi
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Nick A. Guagliardo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Laura A. Bell
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Manako Yamaguchi
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Daisuke Matsuoka
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Fang Xu
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Jason P. Smith
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Mohamed Diagne
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Lucas F. Almeida
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Silvia Medrano
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Paula Q. Barrett
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Edward H. Nieh
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - R. Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Maria Luisa S. Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
6
|
Peralta M, Dupas A, Larnicol A, Lefebvre O, Goswami R, Stemmelen T, Molitor A, Carapito R, Girardo S, Osmani N, Goetz JG. Endothelial calcium firing mediates the extravasation of metastatic tumor cells. iScience 2025; 28:111690. [PMID: 39898056 PMCID: PMC11787530 DOI: 10.1016/j.isci.2024.111690] [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: 03/13/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025] Open
Abstract
Metastatic dissemination is driven by genetic, biochemical, and biophysical cues that favor the distant colonization of organs and the formation of life-threatening secondary tumors. We have previously demonstrated that endothelial cells (ECs) actively remodel during extravasation by enwrapping arrested tumor cells (TCs) and extruding them from the vascular lumen while maintaining perfusion. In this work, we dissect the cellular and molecular mechanisms driving endothelial remodeling. Using high-resolution intravital imaging in zebrafish embryos, we demonstrate that the actomyosin network of ECs controls tissue remodeling and subsequent TC extravasation. Furthermore, we uncovered that this cytoskeletal remodeling is driven by altered endothelial-calcium (Ca2+) signaling caused by arrested TCs. Accordingly, we demonstrated that the inhibition of voltage-dependent calcium L-type channels impairs extravasation. Lastly, we identified P2X4, TRP, and Piezo1 mechano-gated Ca2+ channels as key mediators of the process. These results further highlight the central role of endothelial remodeling during the extravasation of TCs and open avenues for successful therapeutic targeting.
Collapse
Affiliation(s)
- Marina Peralta
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Amandine Dupas
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Ruchi Goswami
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Tristan Stemmelen
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Anne Molitor
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Raphael Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d’ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091 Strasbourg, France
| | - Salvatore Girardo
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Naël Osmani
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jacky G. Goetz
- Tumor Biomechanics lab, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
7
|
Burkhardt DS, Leyden C, Thomas C, Brysch C, Dehmelt FA, Arrenberg AB. Behavioral and neurophysiological effects of electrical stunning on zebrafish larvae. Lab Anim (NY) 2025; 54:50-58. [PMID: 39870879 PMCID: PMC11790490 DOI: 10.1038/s41684-024-01505-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025]
Abstract
Two methods dominate the way that zebrafish larvae are euthanized after experimental procedures: anesthetic overdose and rapid cooling. Although MS-222 is easy to apply, this anesthetic takes about a minute to act and fish show aversive reactions and interindividual differences, limiting its reliability. Rapid cooling kills larvae after several hours and is not listed as an approved method in the relevant European Union directive. Electrical stunning is a promising alternative euthanasia method for zebrafish but has not yet been fully established. Here we characterize both behavioral and neurophysiological effects of electrical stunning in 4-day-old zebrafish larvae. We identified the electric field characteristics and stimulus duration (50 V/cm alternating current for 32 s) that reliably euthanizes free-swimming larvae and agarose-embedded larvae with an easy-to-implement protocol. Behavioral analysis and calcium neurophysiology show that larvae lose consciousness and stop responding to touch and visual stimuli very quickly (<1 s). Electrically stunned larvae no longer show coordinated brain activity. Their brains instead undergo a series of concerted whole-brain calcium waves over the course of many minutes before the cessation of all brain signals. Consistent with the need to implement the 3R at all stages of animal experimentation, the rapid and reliable euthanasia achieved by electrical stunning has potential for refinement of the welfare of more than 5 million zebrafish used annually in biomedical research worldwide.
Collapse
Affiliation(s)
- David-Samuel Burkhardt
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Claire Leyden
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Carina Thomas
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Christian Brysch
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Florian Alexander Dehmelt
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany
| | - Aristides B Arrenberg
- Werner Reichardt Centre for Integrative Neuroscience and Institute for Neurobiology, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
8
|
Coomer CE, Naumova D, Talay M, Zolyomi B, Snell NJ, Sorkaç A, Chanchu JM, Cheng J, Roman I, Li J, Robson D, McLean DL, Barnea G, Halpern ME. Transsynaptic labeling and transcriptional control of zebrafish neural circuits. Nat Neurosci 2025; 28:189-200. [PMID: 39702668 DOI: 10.1038/s41593-024-01815-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/30/2024] [Indexed: 12/21/2024]
Abstract
Deciphering the connectome, the ensemble of synaptic connections that underlie brain function, is a central goal of neuroscience research. Here we report the in vivo mapping of connections between presynaptic and postsynaptic partners in zebrafish, by adapting the trans-Tango genetic approach that was first developed for anterograde transsynaptic tracing in Drosophila. Neural connections were visualized between synaptic partners in larval retina, brain and spinal cord and followed over development. The specificity of labeling was corroborated by functional experiments in which optogenetic activation of presynaptic spinal cord interneurons elicited responses in known motor neuronal postsynaptic targets, as measured by trans-Tango-dependent expression of a genetically encoded calcium indicator or by electrophysiology. Transsynaptic signaling through trans-Tango reveals synaptic connections in the zebrafish nervous system, providing a valuable in vivo tool to monitor and interrogate neural circuits over time.
Collapse
Affiliation(s)
- Cagney E Coomer
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Daria Naumova
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Mustafa Talay
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Howard Hughes Medical Institute, Department of Molecular and Cellular Biology, Harvard University, Boston, MA, USA
| | - Bence Zolyomi
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Nathaniel J Snell
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Altar Sorkaç
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Jean Michel Chanchu
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Ji Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana Roman
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Jennifer Li
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Drew Robson
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - David L McLean
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Gilad Barnea
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Marnie E Halpern
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Islam T, Torigoe M, Tanimoto Y, Okamoto H. Adult zebrafish can learn Morris water maze-like tasks in a two-dimensional virtual reality system. CELL REPORTS METHODS 2024; 4:100863. [PMID: 39317191 PMCID: PMC11573742 DOI: 10.1016/j.crmeth.2024.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/11/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
Virtual reality (VR) has emerged as a powerful tool for investigating neural mechanisms of decision-making, spatial cognition, and navigation. In many head-fixed VRs for rodents, animals locomote on spherical treadmills that provide rotation information in two axes to calculate two-dimensional (2D) movement. On the other hand, zebrafish in a submerged head-fixed VR can move their tail to enable movement in 2D VR environment. This motivated us to create a VR system for adult zebrafish to enable 2D movement consisting of forward translation and rotations calculated from tail movement. Besides presenting the VR system, we show that zebrafish can learn a virtual Morris water maze-like (VMWM) task in which finding an invisible safe zone was necessary for the zebrafish to avoid an aversive periodic mild electric shock. Results show high potential for our VR system to be combined with optical imaging for future studies to investigate spatial learning and navigation.
Collapse
Affiliation(s)
- Tanvir Islam
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
| | - Makio Torigoe
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Yuki Tanimoto
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan; School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatu-cho, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Hitoshi Okamoto
- RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan; School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatu-cho, Shinjuku-ku, Tokyo 169-8555, Japan; Institute of Neuropsychiatry, 91 Benten-cho, Shinjuku-ku, Tokyo 162-0851, Japan.
| |
Collapse
|
10
|
Liu Y, Chen Y, Duffy CR, VanLeuven AJ, Byers JB, Schriever HC, Ball RE, Carpenter JM, Gunderson CE, Filipov NM, Ma P, Kner PA, Lauderdale JD. Decreased GABA levels during development result in increased connectivity in the larval zebrafish tectum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612511. [PMID: 39314470 PMCID: PMC11419034 DOI: 10.1101/2024.09.11.612511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
γ-aminobutyric acid (GABA) is an abundant neurotransmitter that plays multiple roles in the vertebrate central nervous system (CNS). In the early developing CNS, GABAergic signaling acts to depolarize cells. It mediates several aspects of neural development, including cell proliferation, neuronal migration, neurite growth, and synapse formation, as well as the development of critical periods. Later in CNS development, GABAergic signaling acts in an inhibitory manner when it becomes the predominant inhibitory neurotransmitter in the brain. This behavior switch occurs due to changes in chloride/cation transporter expression. Abnormalities of GABAergic signaling appear to underlie several human neurological conditions, including seizure disorders. However, the impact of reduced GABAergic signaling on brain development has been challenging to study in mammals. Here we take advantage of zebrafish and light sheet imaging to assess the impact of reduced GABAergic signaling on the functional circuitry in the larval zebrafish optic tectum. Zebrafish have three gad genes: two gad1 paralogs known as gad1a and gad1b, and gad2. The gad1b and gad2 genes are expressed in the developing optic tectum. Null mutations in gad1b significantly reduce GABA levels in the brain and increase electrophysiological activity in the optic tectum. Fast light sheet imaging of genetically encoded calcium indicator (GCaMP)-expressing gab1b null larval zebrafish revealed patterns of neural activity that were different than either gad1b-normal larvae or gad1b-normal larvae acutely exposed to pentylenetetrazole (PTZ). These results demonstrate that reduced GABAergic signaling during development increases functional connectivity and concomitantly hyper-synchronization of neuronal networks.
Collapse
Affiliation(s)
- Yang Liu
- School of Electrical and Computer Engineering, The University of Georgia, Athens, GA 30602, USA
| | - Yongkai Chen
- Department of Statistics, The University of Georgia, Athens, GA 30602, USA
| | - Carly R Duffy
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Ariel J VanLeuven
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - John Branson Byers
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Hannah C Schriever
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Rebecca E Ball
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Jessica M Carpenter
- Department of Physiology and Pharmacology, The University of Georgia, College of Veterinary Medicine, Athens, GA, 30602, USA
- Neuroscience Division of the Biomedical and Translational Sciences Institute, The University of Georgia, Athens, GA 30602, USA
| | - Chelsea E Gunderson
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
| | - Nikolay M Filipov
- Department of Physiology and Pharmacology, The University of Georgia, College of Veterinary Medicine, Athens, GA, 30602, USA
| | - Ping Ma
- Department of Statistics, The University of Georgia, Athens, GA 30602, USA
| | - Peter A Kner
- School of Electrical and Computer Engineering, The University of Georgia, Athens, GA 30602, USA
| | - James D Lauderdale
- Department of Cellular Biology, The University of Georgia, Athens, GA 30602, USA
- Neuroscience Division of the Biomedical and Translational Sciences Institute, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
11
|
Luu P, Nadtochiy A, Zanon M, Moreno N, Messina A, Petrazzini MEM, Torres Perez JV, Keomanee-Dizon K, Jones M, Brennan CH, Vallortigara G, Fraser SE, Truong TV. Neural Basis of Number Sense in Larval Zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610552. [PMID: 39290349 PMCID: PMC11406567 DOI: 10.1101/2024.08.30.610552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Number sense, the ability to discriminate the quantity of objects, is crucial for survival. To understand how neurons work together and develop to mediate number sense, we used two-photon fluorescence light sheet microscopy to capture the activity of individual neurons throughout the brain of larval Danio rerio, while displaying a visual number stimulus to the animal. We identified number-selective neurons as early as 3 days post-fertilization and found a proportional increase of neurons tuned to larger quantities after 3 days. We used machine learning to predict the stimulus from the neuronal activity and observed that the prediction accuracy improves with age. We further tested ethanol's effect on number sense and found a decrease in number-selective neurons in the forebrain, suggesting cognitive impairment. These findings are a significant step towards understanding neural circuits devoted to discrete magnitudes and our methodology to track single-neuron activity across the whole brain is broadly applicable to other fields in neuroscience.
Collapse
Affiliation(s)
- Peter Luu
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Anna Nadtochiy
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Mirko Zanon
- Centre for Mind/Brain Sciences, University of Trento, Rovereto, Italy
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Noah Moreno
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Andrea Messina
- Centre for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | | | - Jose Vicente Torres Perez
- School of Biological and Behavioral Sciences, Queen Mary University of London, London, United Kingdom
| | - Kevin Keomanee-Dizon
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
- Joseph Henry Laboratories of Physics, Princeton University, Princeton, NJ, USA
| | - Matthew Jones
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
| | - Caroline H Brennan
- School of Biological and Behavioral Sciences, Queen Mary University of London, London, United Kingdom
| | | | - Scott E Fraser
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Thai V Truong
- Translational Imaging Center, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Lebas M, Chinigò G, Courmont E, Bettaieb L, Machmouchi A, Goveia J, Beatovic A, Van Kerckhove J, Robil C, Angulo FS, Vedelago M, Errerd A, Treps L, Gao V, Delgado De la Herrán HC, Mayeuf-Louchart A, L’homme L, Chamlali M, Dejos C, Gouyer V, Garikipati VNS, Tomar D, Yin H, Fukui H, Vinckier S, Stolte A, Conradi LC, Infanti F, Lemonnier L, Zeisberg E, Luo Y, Lin L, Desseyn JL, Pickering G, Kishore R, Madesh M, Dombrowicz D, Perocchi F, Staels B, Pla AF, Gkika D, Cantelmo AR. Integrated single-cell RNA-seq analysis reveals mitochondrial calcium signaling as a modulator of endothelial-to-mesenchymal transition. SCIENCE ADVANCES 2024; 10:eadp6182. [PMID: 39121218 PMCID: PMC11313856 DOI: 10.1126/sciadv.adp6182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/08/2024] [Indexed: 08/11/2024]
Abstract
Endothelial cells (ECs) are highly plastic, capable of differentiating into various cell types. Endothelial-to-mesenchymal transition (EndMT) is crucial during embryonic development and contributes substantially to vascular dysfunction in many cardiovascular diseases (CVDs). While targeting EndMT holds therapeutic promise, understanding its mechanisms and modulating its pathways remain challenging. Using single-cell RNA sequencing on three in vitro EndMT models, we identified conserved gene signatures. We validated original regulators in vitro and in vivo during embryonic heart development and peripheral artery disease. EndMT induction led to global expression changes in all EC subtypes rather than in mesenchymal clusters. We identified mitochondrial calcium uptake as a key driver of EndMT; inhibiting mitochondrial calcium uniporter (MCU) prevented EndMT in vitro, and conditional Mcu deletion in ECs blocked mesenchymal activation in a hind limb ischemia model. Tissues from patients with critical limb ischemia with EndMT features exhibited significantly elevated endothelial MCU. These findings highlight MCU as a regulator of EndMT and a potential therapeutic target.
Collapse
Affiliation(s)
- Mathilde Lebas
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Giorgia Chinigò
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Torino, Italy
| | - Evan Courmont
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Louay Bettaieb
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Amani Machmouchi
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | | | | | | | - Cyril Robil
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Fabiola Silva Angulo
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Mauro Vedelago
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Alina Errerd
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
- Molecular Biosciences/Cancer Biology Program, Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lucas Treps
- Nantes Université, INSERM UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France
| | - Vance Gao
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | | | - Alicia Mayeuf-Louchart
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Laurent L’homme
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Mohamed Chamlali
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Camille Dejos
- INSERM, U1003 - PHYCEL - Physiologie Cellulaire, Université de Lille, F-59000 Lille, France
| | - Valérie Gouyer
- Université de Lille, Inserm, CHU Lille, U1286 Infinite, F-59000 Lille, France
| | - Venkata Naga Srikanth Garikipati
- Aging + Cardiovascular Discovery Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Dhanendra Tomar
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Hao Yin
- Robarts Research Institute, Western University, London, Canada
| | - Hajime Fukui
- National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology (CCB), VIB and Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Anneke Stolte
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany
| | | | - Loic Lemonnier
- INSERM, U1003 - PHYCEL - Physiologie Cellulaire, Université de Lille, F-59000 Lille, France
| | - Elisabeth Zeisberg
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- DZHK German Center for Cardiovascular Research, Partner Site Lower Saxony, Göttingen, Germany
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jean-Luc Desseyn
- Université de Lille, Inserm, CHU Lille, U1286 Infinite, F-59000 Lille, France
| | - Geoffrey Pickering
- Robarts Research Institute, Western University, London, Canada
- Department of Medicine, Biochemistry, and Medical Biophysics, Western University, London, Canada
| | - Raj Kishore
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140 USA
| | - Muniswamy Madesh
- Department of Medicine, Center for Mitochondrial Medicine, Division of Cardiology, University of Texas Health San Antonio, San Antonio, TX 78229 USA
| | - David Dombrowicz
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Fabiana Perocchi
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Munich, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Bart Staels
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Alessandra Fiorio Pla
- Department of Life Sciences and Systems Biology, University of Torino, 10123 Torino, Italy
- INSERM, U1003 - PHYCEL - Physiologie Cellulaire, Université de Lille, F-59000 Lille, France
| | - Dimitra Gkika
- Université de Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Anna Rita Cantelmo
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| |
Collapse
|
13
|
Ito S, Uchida N. Selective decision-making and collective behavior of fish by the motion of visual attention. PNAS NEXUS 2024; 3:pgae264. [PMID: 39045016 PMCID: PMC11264410 DOI: 10.1093/pnasnexus/pgae264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/23/2024] [Indexed: 07/25/2024]
Abstract
Collective motion provides a spectacular example of self-organization in Nature. Visual information plays a crucial role among various types of information in determining interactions. Recently, experiments have revealed that organisms such as fish and insects selectively utilize a portion, rather than the entirety, of visual information. Here, focusing on fish, we propose an agent-based model where the direction of attention is guided by visual stimuli received from the images of nearby fish. Our model reproduces a branching phenomenon where a fish selectively follows a specific individual as the distance between two or three nearby fish increases. Furthermore, our model replicates various patterns of collective motion in a group of agents, such as vortex, polarized school, swarm, and turning. We also discuss the topological nature of the visual interaction, as well as the positional distribution of nearby fish and the map of pairwise and three-body interactions induced by them. Through a comprehensive comparison with existing experimental results, we clarify the roles of visual interactions and issues to be resolved by other forms of interactions.
Collapse
Affiliation(s)
- Susumu Ito
- Department of Physics, Tohoku University, Sendai 980-8578, Japan
| | - Nariya Uchida
- Department of Physics, Tohoku University, Sendai 980-8578, Japan
| |
Collapse
|
14
|
McDonald KO, Lyons NMA, Gray LKC, Xu JB, Schoderboeck L, Hughes SM, Basak I. Transcription Factor-Mediated Generation of Dopaminergic Neurons from Human iPSCs-A Comparison of Methods. Cells 2024; 13:1016. [PMID: 38920646 PMCID: PMC11201854 DOI: 10.3390/cells13121016] [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: 02/23/2024] [Revised: 05/23/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
Dopaminergic neurons are the predominant brain cells affected in Parkinson's disease. With the limited availability of live human brain dopaminergic neurons to study pathological mechanisms of Parkinson's disease, dopaminergic neurons have been generated from human-skin-cell-derived induced pluripotent stem cells. Originally, induced pluripotent stem-cell-derived dopaminergic neurons were generated using small molecules. These neurons took more than two months to mature. However, the transcription-factor-mediated differentiation of induced pluripotent stem cells has revealed quicker and cheaper methods to generate dopaminergic neurons. In this study, we compared and contrasted three protocols to generate induced pluripotent stem-cell-derived dopaminergic neurons using transcription-factor-mediated directed differentiation. We deviated from the established protocols using lentivirus transduction to stably integrate different transcription factors into the AAVS1 safe harbour locus of induced pluripotent stem cells. We used different media compositions to generate more than 90% of neurons in the culture, out of which more than 85% of the neurons were dopaminergic neurons within three weeks. Therefore, from our comparative study, we reveal that a combination of transcription factors along with small molecule treatment may be required to generate a pure population of human dopaminergic neurons.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Indranil Basak
- Department of Biochemistry, Brain Health Research Centre and Genetics Otago, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
15
|
Spikol ED, Cheng J, Macurak M, Subedi A, Halpern ME. Genetically defined nucleus incertus neurons differ in connectivity and function. eLife 2024; 12:RP89516. [PMID: 38819436 PMCID: PMC11142643 DOI: 10.7554/elife.89516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
The nucleus incertus (NI), a conserved hindbrain structure implicated in the stress response, arousal, and memory, is a major site for production of the neuropeptide relaxin-3. On the basis of goosecoid homeobox 2 (gsc2) expression, we identified a neuronal cluster that lies adjacent to relaxin 3a (rln3a) neurons in the zebrafish analogue of the NI. To delineate the characteristics of the gsc2 and rln3a NI neurons, we used CRISPR/Cas9 targeted integration to drive gene expression specifically in each neuronal group, and found that they differ in their efferent and afferent connectivity, spontaneous activity, and functional properties. gsc2 and rln3a NI neurons have widely divergent projection patterns and innervate distinct subregions of the midbrain interpeduncular nucleus (IPN). Whereas gsc2 neurons are activated more robustly by electric shock, rln3a neurons exhibit spontaneous fluctuations in calcium signaling and regulate locomotor activity. Our findings define heterogeneous neurons in the NI and provide new tools to probe its diverse functions.
Collapse
Affiliation(s)
- Emma D Spikol
- Department of Molecular and Systems Biology, Geisel School of Medicine at DartmouthHanoverUnited States
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ji Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at DartmouthHanoverUnited States
- Department of Biology, Johns Hopkins UniversityBaltimoreUnited States
| | - Michelle Macurak
- Department of Biology, Johns Hopkins UniversityBaltimoreUnited States
| | - Abhignya Subedi
- Department of Biology, Johns Hopkins UniversityBaltimoreUnited States
| | - Marnie E Halpern
- Department of Molecular and Systems Biology, Geisel School of Medicine at DartmouthHanoverUnited States
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Biology, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
16
|
Anzai T, Myatt GJ, Hall F, Finney B, Nakagawa K, Iwata H, Anzai R, Dickinson A, Freer M, Nakae D, Onodera H, Matsuyama T. Drug review process advancement and required manufacturer and contract research oraganization responses. J Toxicol Pathol 2024; 37:45-53. [PMID: 38584971 PMCID: PMC10995435 DOI: 10.1293/tox.2023-0106] [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: 09/08/2023] [Accepted: 11/09/2023] [Indexed: 04/09/2024] Open
Abstract
The United States Senate passed the "FDA Modernization Act 2.0." on September 29, 2022. Although the effectiveness of this Bill, which aims to eliminate the mandatory use of laboratory animals in new drug development, is limited, it represents a significant trend that will change the shape of drug applications in the United States and other countries. However, pharmaceutical companies have not taken major steps towards the complete elimination of animal testing from the standpoint of product safety, where they prioritize patient safety. Nonetheless, society is becoming increasingly opposed to animal testing, and efforts will be made to use fewer animals and conduct fewer animal tests as a natural and reasonable response. These changes eventually alter the shape of new drug applications. Based on the assumption that fewer animal tests will be conducted or fewer animals will be used in testing, this study explored bioinformatics and new technologies as alternatives to compensate for reduced information and provide a picture of how future new drug applications may look. The authors also discuss the directions that pharmaceutical companies and nonclinical contract research organizations should adopt to promote the replacement, reduction, and refinement of animals used in research, teaching, testing, and exhibitions.
Collapse
Affiliation(s)
- Takayuki Anzai
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Glenn J. Myatt
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Frances Hall
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Brenda Finney
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Kenshi Nakagawa
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Hijiri Iwata
- LunaPath Laboratory of Toxicologic Pathology, 3-5-1
Aoihigashi, Naka-ku Hamamatsu-shi, Shizuoka 433-8114, Japan
| | - Reo Anzai
- Tokyo University of the Arts, 5000 Omonma, Toride-shi,
Ibaraki 302-0001, Japan
| | - Anne Dickinson
- Newcastle University, Newcastle upon Tyne, NE1 7RU, United
Kingdom
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Matthew Freer
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Dai Nakae
- Teikyo Heisei University, 4-1 Uruido-Minami, Ichihara-shi,
Chiba 290-0193, Japan
| | - Hiroshi Onodera
- National Institute of Health Sciences, 3-25-26 Tono-machi,
Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takaaki Matsuyama
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| |
Collapse
|
17
|
Schmidt AR, Placer HJ, Muhammad IM, Shephard R, Patrick RL, Saurborn T, Horstick EJ, Bergeron SA. Transcriptional control of visual neural circuit development by GS homeobox 1. PLoS Genet 2024; 20:e1011139. [PMID: 38669217 PMCID: PMC11051655 DOI: 10.1371/journal.pgen.1011139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 01/16/2024] [Indexed: 04/28/2024] Open
Abstract
As essential components of gene expression networks, transcription factors regulate neural circuit assembly. The homeobox transcription factor encoding gene, gs homeobox 1 (gsx1), is expressed in the developing visual system; however, no studies have examined its role in visual system formation. In zebrafish, retinal ganglion cell (RGC) axons that transmit visual information to the brain terminate in ten arborization fields (AFs) in the optic tectum (TeO), pretectum (Pr), and thalamus. Pretectal AFs (AF1-AF9) mediate distinct visual behaviors, yet we understand less about their development compared to AF10 in the TeO. Using gsx1 zebrafish mutants, immunohistochemistry, and transgenic lines, we observed that gsx1 is required for vesicular glutamate transporter, Tg(slc17a6b:DsRed), expression in the Pr, but not overall neuron number. gsx1 mutants have normal eye morphology, yet they exhibit impaired visual ability during prey capture. RGC axon volume in the gsx1 mutant Pr and TeO is reduced, and AF7 that is active during feeding is missing which is consistent with reduced hunting performance. Timed laser ablation of Tg(slc17a6b:DsRed)-positive cells reveals that they are necessary for AF7 formation. This work is the first to implicate gsx1 in establishing cell identity and functional neural circuits in the visual system.
Collapse
Affiliation(s)
- Alexandra R. Schmidt
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Haiden J. Placer
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Ishmael M. Muhammad
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Rebekah Shephard
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Regina L. Patrick
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Taylor Saurborn
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
| | - Eric J. Horstick
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
- Department of Neuroscience, West Virginia University, Morgantown, West Virgina, United States of America
| | - Sadie A. Bergeron
- Department of Biology, West Virginia University, Morgantown, West Virgina, United States of America
- Department of Neuroscience, West Virginia University, Morgantown, West Virgina, United States of America
| |
Collapse
|
18
|
Schevenels G, Cabochette P, America M, Vandenborne A, De Grande L, Guenther S, He L, Dieu M, Christou B, Vermeersch M, Germano RFV, Perez-Morga D, Renard P, Martin M, Vanlandewijck M, Betsholtz C, Vanhollebeke B. A brain-specific angiogenic mechanism enabled by tip cell specialization. Nature 2024; 628:863-871. [PMID: 38570687 PMCID: PMC11041701 DOI: 10.1038/s41586-024-07283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 03/07/2024] [Indexed: 04/05/2024]
Abstract
Vertebrate organs require locally adapted blood vessels1,2. The gain of such organotypic vessel specializations is often deemed to be molecularly unrelated to the process of organ vascularization. Here, opposing this model, we reveal a molecular mechanism for brain-specific angiogenesis that operates under the control of Wnt7a/b ligands-well-known blood-brain barrier maturation signals3-5. The control mechanism relies on Wnt7a/b-dependent expression of Mmp25, which we find is enriched in brain endothelial cells. CRISPR-Cas9 mutagenesis in zebrafish reveals that this poorly characterized glycosylphosphatidylinositol-anchored matrix metalloproteinase is selectively required in endothelial tip cells to enable their initial migration across the pial basement membrane lining the brain surface. Mechanistically, Mmp25 confers brain invasive competence by cleaving meningeal fibroblast-derived collagen IV α5/6 chains within a short non-collagenous region of the central helical part of the heterotrimer. After genetic interference with the pial basement membrane composition, the Wnt-β-catenin-dependent organotypic control of brain angiogenesis is lost, resulting in properly patterned, yet blood-brain-barrier-defective cerebrovasculatures. We reveal an organ-specific angiogenesis mechanism, shed light on tip cell mechanistic angiodiversity and thereby illustrate how organs, by imposing local constraints on angiogenic tip cells, can select vessels matching their distinctive physiological requirements.
Collapse
Affiliation(s)
- Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Pauline Cabochette
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michelle America
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Arnaud Vandenborne
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Line De Grande
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim, Germany
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Marc Dieu
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Basile Christou
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Raoul F V Germano
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Perez-Morga
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Patricia Renard
- Mass Spectrometry Facility (MaSUN), University of Namur, Namur, Belgium
| | - Maud Martin
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
| | - Michael Vanlandewijck
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium.
| |
Collapse
|
19
|
Chai Y, Qi K, Wu Y, Li D, Tan G, Guo Y, Chu J, Mu Y, Shen C, Wen Q. All-optical interrogation of brain-wide activity in freely swimming larval zebrafish. iScience 2024; 27:108385. [PMID: 38205255 PMCID: PMC10776927 DOI: 10.1016/j.isci.2023.108385] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 01/12/2024] Open
Abstract
We introduce an all-optical technique that enables volumetric imaging of brain-wide calcium activity and targeted optogenetic stimulation of specific brain regions in unrestrained larval zebrafish. The system consists of three main components: a 3D tracking module, a dual-color fluorescence imaging module, and a real-time activity manipulation module. Our approach uses a sensitive genetically encoded calcium indicator in combination with a long Stokes shift red fluorescence protein as a reference channel, allowing the extraction of Ca2+ activity from signals contaminated by motion artifacts. The method also incorporates rapid 3D image reconstruction and registration, facilitating real-time selective optogenetic stimulation of different regions of the brain. By demonstrating that selective light activation of the midbrain regions in larval zebrafish could reliably trigger biased turning behavior and changes of brain-wide neural activity, we present a valuable tool for investigating the causal relationship between distributed neural circuit dynamics and naturalistic behavior.
Collapse
Affiliation(s)
- Yuming Chai
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Kexin Qi
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Yubin Wu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Daguang Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Guodong Tan
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Yuqi Guo
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology and Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology and Center for Biomedical Optics and Molecular Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yu Mu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chen Shen
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| | - Quan Wen
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Center for Physical Sciences at the Microscale, Center for Integrative Imaging, University of Science and Technology of China, Hefei, China
| |
Collapse
|
20
|
Uribe-Arias A, Rozenblat R, Vinepinsky E, Marachlian E, Kulkarni A, Zada D, Privat M, Topsakalian D, Charpy S, Candat V, Nourin S, Appelbaum L, Sumbre G. Radial astrocyte synchronization modulates the visual system during behavioral-state transitions. Neuron 2023; 111:4040-4057.e6. [PMID: 37863038 PMCID: PMC10783638 DOI: 10.1016/j.neuron.2023.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Glial cells support the function of neurons. Recent evidence shows that astrocytes are also involved in brain computations. To explore whether and how their excitable nature affects brain computations and motor behaviors, we used two-photon Ca2+ imaging of zebrafish larvae expressing GCaMP in both neurons and radial astrocytes (RAs). We found that in the optic tectum, RAs synchronize their Ca2+ transients immediately after the end of an escape behavior. Using optogenetics, ablations, and a genetically encoded norepinephrine sensor, we observed that RA synchronous Ca2+ events are mediated by the locus coeruleus (LC)-norepinephrine circuit. RA synchronization did not induce direct excitation or inhibition of tectal neurons. Nevertheless, it modulated the direction selectivity and the long-distance functional correlations among neurons. This mechanism supports freezing behavior following a switch to an alerted state. These results show that LC-mediated neuro-glial interactions modulate the visual system during transitions between behavioral states.
Collapse
Affiliation(s)
- Alejandro Uribe-Arias
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Rotem Rozenblat
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Ehud Vinepinsky
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Emiliano Marachlian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Anirudh Kulkarni
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - David Zada
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Martin Privat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Diego Topsakalian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Charpy
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Virginie Candat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Nourin
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Germán Sumbre
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France.
| |
Collapse
|
21
|
Nava S, Palma W, Wan X, Oh JY, Gharib S, Wang H, Revanna JS, Tan M, Zhang M, Liu J, Chen CH, Lee JS, Perry B, Sternberg PW. A cGAL-UAS bipartite expression toolkit for Caenorhabditis elegans sensory neurons. Proc Natl Acad Sci U S A 2023; 120:e2221680120. [PMID: 38096407 PMCID: PMC10743456 DOI: 10.1073/pnas.2221680120] [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: 12/26/2022] [Accepted: 10/05/2023] [Indexed: 12/18/2023] Open
Abstract
Animals integrate sensory information from the environment and display various behaviors in response to external stimuli. In Caenorhabditis elegans hermaphrodites, 33 types of sensory neurons are responsible for chemosensation, olfaction, and mechanosensation. However, the functional roles of all sensory neurons have not been systematically studied due to the lack of facile genetic accessibility. A bipartite cGAL-UAS system has been previously developed to study tissue- or cell-specific functions in C. elegans. Here, we report a toolkit of new cGAL drivers that can facilitate the analysis of a vast majority of the 60 sensory neurons in C. elegans hermaphrodites. We generated 37 sensory neuronal cGAL drivers that drive cGAL expression by cell-specific regulatory sequences or intersection of two distinct regulatory regions with overlapping expression (split cGAL). Most cGAL-drivers exhibit expression in single types of cells. We also constructed 28 UAS effectors that allow expression of proteins to perturb or interrogate sensory neurons of choice. This cGAL-UAS sensory neuron toolkit provides a genetic platform to systematically study the functions of C. elegans sensory neurons.
Collapse
Affiliation(s)
- Stephanie Nava
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Wilber Palma
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Xuan Wan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Jun Young Oh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Shahla Gharib
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Han Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Jasmin S. Revanna
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Minyi Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Mark Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Jonathan Liu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Chun-Hao Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - James S. Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Barbara Perry
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| | - Paul W. Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125
| |
Collapse
|
22
|
Becker CJ, Cigliola V, Gillotay P, Rich A, De Simone A, Han Y, Di Talia S, Poss KD. In toto imaging of glial JNK signaling during larval zebrafish spinal cord regeneration. Development 2023; 150:dev202076. [PMID: 37997694 PMCID: PMC10753585 DOI: 10.1242/dev.202076] [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: 06/12/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023]
Abstract
Identification of signaling events that contribute to innate spinal cord regeneration in zebrafish can uncover new targets for modulating injury responses of the mammalian central nervous system. Using a chemical screen, we identify JNK signaling as a necessary regulator of glial cell cycling and tissue bridging during spinal cord regeneration in larval zebrafish. With a kinase translocation reporter, we visualize and quantify JNK signaling dynamics at single-cell resolution in glial cell populations in developing larvae and during injury-induced regeneration. Glial JNK signaling is patterned in time and space during development and regeneration, decreasing globally as the tissue matures and increasing in the rostral cord stump upon transection injury. Thus, dynamic and regional regulation of JNK signaling help to direct glial cell behaviors during innate spinal cord regeneration.
Collapse
Affiliation(s)
- Clayton J. Becker
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Valentina Cigliola
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose, 06100 Nice, France
| | - Pierre Gillotay
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ashley Rich
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alessandro De Simone
- Department of Genetics and Evolution, University of Geneva, 1211 Geneva, Switzerland
| | - Yanchao Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College, Soochow University, Suzhou, 215006 Jiangsu, China
| | - Stefano Di Talia
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D. Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
23
|
Chen Y, Pang S, Li J, Lu Y, Gao C, Xiao Y, Chen M, Wang M, Ren X. Genetically encoded protein sensors for metal ion detection in biological systems: a review and bibliometric analysis. Analyst 2023; 148:5564-5581. [PMID: 37872814 DOI: 10.1039/d3an01412f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Metal ions are indispensable elements in living organisms and are associated with regulating various biological processes. An imbalance in metal ion content can lead to disorders in normal physiological functions of the human body and cause various diseases. Genetically encoded fluorescent protein sensors have the advantages of low biotoxicity, high specificity, and a long imaging time in vivo and have become a powerful tool to visualize or quantify the concentration level of biomolecules in vivo and in vitro, temporal and spatial distribution, and life activity process. This review analyzes the development status and current research hotspots in the field of genetically encoded fluorescent protein sensors by bibliometric analysis. Based on the results of bibliometric analysis, the research progress of genetically encoded fluorescent protein sensors for metal ion detection is reviewed, and the construction strategies, physicochemical properties, and applications of such sensors in biological imaging are summarized.
Collapse
Affiliation(s)
- Yuxueyuan Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - ShuChao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jingya Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yun Lu
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chenxia Gao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanyu Xiao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meiling Chen
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Meng Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin 301617, China
| | - Xiaoliang Ren
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
24
|
Asakawa K, Handa H, Kawakami K. Dysregulated TDP-43 proteostasis perturbs excitability of spinal motor neurons during brainstem-mediated fictive locomotion in zebrafish. Dev Growth Differ 2023; 65:446-452. [PMID: 37452624 PMCID: PMC11520980 DOI: 10.1111/dgd.12879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/24/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Spinal motor neurons (SMNs) are the primary target of degeneration in amyotrophic lateral sclerosis (ALS). Degenerating motor neurons accumulate cytoplasmic TAR DNA-binding protein 43 (TDP-43) aggregates in most ALS cases. This SMN pathology can occur without mutation in the coding sequence of the TDP-43-encoding gene, TARDBP. Whether and how wild-type TDP-43 drives pathological changes in SMNs in vivo remains largely unexplored. In this study, we develop a two-photon calcium imaging setup in which tactile-evoked neural responses of motor neurons in the brainstem and spinal cord can be monitored using the calcium indicator GCaMP. We devise a piezo-assisted tactile stimulator that reproducibly evokes a brainstem descending neuron upon tactile stimulation of the head. A direct comparison between caudal primary motor neurons (CaPs) with or without TDP-43 overexpression in contiguous spinal segments demonstrates that CaPs overexpressing TDP-43 display attenuated Ca2+ transients during fictive escape locomotion evoked by the tactile stimulation. These results show that excessive amounts of TDP-43 protein reduce the neuronal excitability of SMNs and potentially contribute to asymptomatic pathological lesions of SMNs and movement disorders in patients with ALS.
Collapse
Affiliation(s)
- Kazuhide Asakawa
- Laboratory of Molecular and Developmental BiologyNational Institute of GeneticsMishimaJapan
| | - Hiroshi Handa
- Department of Molecular Pharmacology, Center for Future Medical Research, Institute of Medical ScienceTokyo Medical UniversityTokyoJapan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental BiologyNational Institute of GeneticsMishimaJapan
- The Graduate University for Advanced Studies (SOKENDAI)MishimaJapan
| |
Collapse
|
25
|
Eom M, Han S, Park P, Kim G, Cho ES, Sim J, Lee KH, Kim S, Tian H, Böhm UL, Lowet E, Tseng HA, Choi J, Lucia SE, Ryu SH, Rózsa M, Chang S, Kim P, Han X, Piatkevich KD, Choi M, Kim CH, Cohen AE, Chang JB, Yoon YG. Statistically unbiased prediction enables accurate denoising of voltage imaging data. Nat Methods 2023; 20:1581-1592. [PMID: 37723246 PMCID: PMC10555843 DOI: 10.1038/s41592-023-02005-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 08/10/2023] [Indexed: 09/20/2023]
Abstract
Here we report SUPPORT (statistically unbiased prediction utilizing spatiotemporal information in imaging data), a self-supervised learning method for removing Poisson-Gaussian noise in voltage imaging data. SUPPORT is based on the insight that a pixel value in voltage imaging data is highly dependent on its spatiotemporal neighboring pixels, even when its temporally adjacent frames alone do not provide useful information for statistical prediction. Such dependency is captured and used by a convolutional neural network with a spatiotemporal blind spot to accurately denoise voltage imaging data in which the existence of the action potential in a time frame cannot be inferred by the information in other frames. Through simulations and experiments, we show that SUPPORT enables precise denoising of voltage imaging data and other types of microscopy image while preserving the underlying dynamics within the scene.
Collapse
Affiliation(s)
- Minho Eom
- School of Electrical Engineering, KAIST, Daejeon, Republic of Korea
| | - Seungjae Han
- School of Electrical Engineering, KAIST, Daejeon, Republic of Korea
| | - Pojeong Park
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Gyuri Kim
- School of Electrical Engineering, KAIST, Daejeon, Republic of Korea
| | - Eun-Seo Cho
- School of Electrical Engineering, KAIST, Daejeon, Republic of Korea
| | - Jueun Sim
- Department of Materials Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Kang-Han Lee
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Seonghoon Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - He Tian
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Urs L Böhm
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Einstein Center for Neurosciences, NeuroCure Cluster of Excellence, Charité University of Medicine Berlin, Berlin, Germany
| | - Eric Lowet
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Hua-An Tseng
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Jieun Choi
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
| | - Stephani Edwina Lucia
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
| | - Seung Hyun Ryu
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Republic of Korea
| | - Márton Rózsa
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Sunghoe Chang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea
- Graduate School of Nanoscience and Technology, KAIST, Daejeon, Republic of Korea
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Kiryl D Piatkevich
- Research Center for Industries of the Future and School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Myunghwan Choi
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Jae-Byum Chang
- Department of Materials Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Young-Gyu Yoon
- School of Electrical Engineering, KAIST, Daejeon, Republic of Korea.
- KAIST Institute for Health Science and Technology, Daejeon, Republic of Korea.
- Department of Semiconductor System Engineering, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
26
|
Coomer C, Naumova D, Talay M, Zolyomi B, Snell N, Sorkac A, Chanchu JM, Cheng J, Roman I, Li J, Robson D, Barnea G, Halpern ME. Transsynaptic labeling and transcriptional control of zebrafish neural circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535421. [PMID: 37066422 PMCID: PMC10103993 DOI: 10.1101/2023.04.03.535421] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Deciphering the connectome, the ensemble of synaptic connections that underlie brain function is a central goal of neuroscience research. The trans-Tango genetic approach, initially developed for anterograde transsynaptic tracing in Drosophila, can be used to map connections between presynaptic and postsynaptic partners and to drive gene expression in target neurons. Here, we describe the successful adaptation of trans-Tango to visualize neural connections in a living vertebrate nervous system, that of the zebrafish. Connections were validated between synaptic partners in the larval retina and brain. Results were corroborated by functional experiments in which optogenetic activation of retinal ganglion cells elicited responses in neurons of the optic tectum, as measured by trans-Tango-dependent expression of a genetically encoded calcium indicator. Transsynaptic signaling through trans-Tango reveals predicted as well as previously undescribed synaptic connections, providing a valuable in vivo tool to monitor and interrogate neural circuits over time.
Collapse
|
27
|
Hasani H, Sun J, Zhu SI, Rong Q, Willomitzer F, Amor R, McConnell G, Cossairt O, Goodhill GJ. Whole-brain imaging of freely-moving zebrafish. Front Neurosci 2023; 17:1127574. [PMID: 37139528 PMCID: PMC10150962 DOI: 10.3389/fnins.2023.1127574] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
One of the holy grails of neuroscience is to record the activity of every neuron in the brain while an animal moves freely and performs complex behavioral tasks. While important steps forward have been taken recently in large-scale neural recording in rodent models, single neuron resolution across the entire mammalian brain remains elusive. In contrast the larval zebrafish offers great promise in this regard. Zebrafish are a vertebrate model with substantial homology to the mammalian brain, but their transparency allows whole-brain recordings of genetically-encoded fluorescent indicators at single-neuron resolution using optical microscopy techniques. Furthermore zebrafish begin to show a complex repertoire of natural behavior from an early age, including hunting small, fast-moving prey using visual cues. Until recently work to address the neural bases of these behaviors mostly relied on assays where the fish was immobilized under the microscope objective, and stimuli such as prey were presented virtually. However significant progress has recently been made in developing brain imaging techniques for zebrafish which are not immobilized. Here we discuss recent advances, focusing particularly on techniques based on light-field microscopy. We also draw attention to several important outstanding issues which remain to be addressed to increase the ecological validity of the results obtained.
Collapse
Affiliation(s)
- Hamid Hasani
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, United States
| | - Jipeng Sun
- Department of Computer Science, Northwestern University, Evanston, IL, United States
| | - Shuyu I. Zhu
- Departments of Developmental Biology and Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
| | - Qiangzhou Rong
- Departments of Developmental Biology and Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
| | - Florian Willomitzer
- Wyant College of Optical Sciences, University of Arizona, Tucson, AZ, United States
| | - Rumelo Amor
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Gail McConnell
- Centre for Biophotonics, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Oliver Cossairt
- Department of Computer Science, Northwestern University, Evanston, IL, United States
| | - Geoffrey J. Goodhill
- Departments of Developmental Biology and Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
28
|
Zylbertal A, Bianco IH. Recurrent network interactions explain tectal response variability and experience-dependent behavior. eLife 2023; 12:78381. [PMID: 36943029 PMCID: PMC10030118 DOI: 10.7554/elife.78381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/09/2023] [Indexed: 03/23/2023] Open
Abstract
Response variability is an essential and universal feature of sensory processing and behavior. It arises from fluctuations in the internal state of the brain, which modulate how sensory information is represented and transformed to guide behavioral actions. In part, brain state is shaped by recent network activity, fed back through recurrent connections to modulate neuronal excitability. However, the degree to which these interactions influence response variability and the spatial and temporal scales across which they operate, are poorly understood. Here, we combined population recordings and modeling to gain insights into how neuronal activity modulates network state and thereby impacts visually evoked activity and behavior. First, we performed cellular-resolution calcium imaging of the optic tectum to monitor ongoing activity, the pattern of which is both a cause and consequence of changes in network state. We developed a minimal network model incorporating fast, short range, recurrent excitation and long-lasting, activity-dependent suppression that reproduced a hallmark property of tectal activity - intermittent bursting. We next used the model to estimate the excitability state of tectal neurons based on recent activity history and found that this explained a portion of the trial-to-trial variability in visually evoked responses, as well as spatially selective response adaptation. Moreover, these dynamics also predicted behavioral trends such as selective habituation of visually evoked prey-catching. Overall, we demonstrate that a simple recurrent interaction motif can be used to estimate the effect of activity upon the incidental state of a neural network and account for experience-dependent effects on sensory encoding and visually guided behavior.
Collapse
Affiliation(s)
- Asaph Zylbertal
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| | - Isaac H Bianco
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
29
|
Hagio H, Yamamoto N. Ascending Visual Pathways to the Telencephalon in Teleosts with Special Focus on Forebrain Visual Centers, Associated Neural Circuitries, and Evolution. Zoolog Sci 2023; 40:105-118. [PMID: 37042690 DOI: 10.2108/zs220089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/10/2022] [Indexed: 03/08/2023]
Abstract
Visual pathways to the telencephalon in teleost fishes have been studied in detail only in a few species, and their evolutionary history remained unclear. On the basis of our recent studies we propose that there were two visual pathways in the common ancestor of teleosts, while one of them became lost in acanthopterygian fishes that emerged relatively recently. Our in-depth analyses on the connections of visual centers also revealed that there are connections shared with those of mammals, and retinotopic organization of the ascending connections is maintained at least to the level of the diencephalon in the yellowfin goby. The major visual telencephalic center, or the lateral part of the dorsal telencephalon (Dl), shows considerable species differences in the number of regions and cytoarchitecture. In particular, four highly specialized compartments are noted in the Dl of gobies, and we analyzed about 100 species of teleosts to investigate the evolution of the compartments in the Dl, which indicated that four compartments emerged only in Gobiiformes, while there are fewer specialized compartments in some other percomorph lineages. We also discuss the connections of forebrain visual centers with the cerebellum and other lower brain centers and infer possible functions of the circuitries.
Collapse
Affiliation(s)
- Hanako Hagio
- Laboratory of Fish Biology, Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Naoyuki Yamamoto
- Laboratory of Fish Biology, Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| |
Collapse
|
30
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
31
|
Tanimoto M, Watakabe I, Higashijima SI. Tiltable objective microscope visualizes selectivity for head motion direction and dynamics in zebrafish vestibular system. Nat Commun 2022; 13:7622. [PMID: 36543769 PMCID: PMC9772181 DOI: 10.1038/s41467-022-35190-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/16/2022] [Indexed: 12/24/2022] Open
Abstract
Spatio-temporal information about head orientation and movement is fundamental to the sense of balance and motion. Hair cells (HCs) in otolith organs of the vestibular system transduce linear acceleration, including head tilt and vibration. Here, we build a tiltable objective microscope in which an objective lens and specimen tilt together. With in vivo Ca2+ imaging of all utricular HCs and ganglion neurons during 360° static tilt and vibration in pitch and roll axes, we reveal the direction- and static/dynamic stimulus-selective topographic responses in larval zebrafish. We find that head vibration is preferentially received by striolar HCs, whereas static tilt is preferentially transduced by extrastriolar HCs. Spatially ordered direction preference in HCs is consistent with hair-bundle polarity and is preserved in ganglion neurons through topographic innervation. Together, these results demonstrate topographically organized selectivity for direction and dynamics of head orientation/movement in the vestibular periphery.
Collapse
Affiliation(s)
- Masashi Tanimoto
- Division of Behavioral Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, 444-8787, Japan.
- Neuronal Networks Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi, 444-8787, Japan.
| | - Ikuko Watakabe
- Division of Behavioral Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, 444-8787, Japan
- Neuronal Networks Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi, 444-8787, Japan
| | - Shin-Ichi Higashijima
- Division of Behavioral Neurobiology, National Institute for Basic Biology, Okazaki, Aichi, 444-8787, Japan.
- Neuronal Networks Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), Okazaki, Aichi, 444-8787, Japan.
| |
Collapse
|
32
|
Sterin I, Santos AC, Park S. Neuronal Activity Reporters as Drug Screening Platforms. MICROMACHINES 2022; 13:1500. [PMID: 36144123 PMCID: PMC9504476 DOI: 10.3390/mi13091500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/25/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Understanding how neuronal activity changes and detecting such changes in both normal and disease conditions is of fundamental importance to the field of neuroscience. Neuronal activity plays important roles in the formation and function of both synapses and circuits, and dysregulation of these processes has been linked to a number of debilitating diseases such as autism, schizophrenia, and epilepsy. Despite advances in our understanding of synapse biology and in how it is altered in disease, the development of therapeutics for these diseases has not advanced apace. Many neuronal activity assays have been developed over the years using a variety of platforms and approaches, but major limitations persist. Current assays, such as fluorescence indicators are not designed to monitor neuronal activity over a long time, they are typically low-throughput or lack sensitivity. These are major barriers to the development of new therapies, as drug screening needs to be both high-throughput to screen through libraries of compounds, and longitudinal to detect any effects that may emerge after continued application of the drug. This review will cover existing assays for measuring neuronal activity and highlight a live-cell assay recently developed. This assay can be performed with easily accessible lab equipment, is both scalable and longitudinal, and can be combined with most other established methods.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ana C. Santos
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
33
|
Shin C, Ryu H, Cho ES, Han S, Lee KH, Kim CH, Yoon YG. Three-dimensional fluorescence microscopy through virtual refocusing using a recursive light propagation network. Med Image Anal 2022; 82:102600. [PMID: 36116298 DOI: 10.1016/j.media.2022.102600] [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: 02/18/2022] [Revised: 06/28/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022]
Abstract
Three-dimensional fluorescence microscopy has an intrinsic performance limit set by the number of photons that can be collected from the sample in a given time interval. Here, we extend our earlier work - a recursive light propagation network (RLP-Net) - which is a computational microscopy technique that overcomes such limitations through virtual refocusing that enables volume reconstruction from two adjacent 2-D wide-field fluorescence images. RLP-Net employs a recursive inference scheme in which the network progressively predicts the subsequent planes along the axial direction. This recursive inference scheme reflects that the law of physics for the light propagation remains spatially invariant and therefore a fixed function (i.e., a neural network) for a short distance light propagation can be recursively applied for a longer distance light propagation. In addition, we employ a self-supervised denoising method to enable accurate virtual light propagation over a long distance. We demonstrate the capability of our method through high-speed volumetric imaging of neuronal activity of a live zebrafish brain. The source code used in the paper is available at https://github.com/NICALab/rlpnet.
Collapse
Affiliation(s)
- Changyeop Shin
- School of Electrical Engineering, KAIST, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyun Ryu
- School of Electrical Engineering, KAIST, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Eun-Seo Cho
- School of Electrical Engineering, KAIST, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seungjae Han
- School of Electrical Engineering, KAIST, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kang-Han Lee
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Young-Gyu Yoon
- School of Electrical Engineering, KAIST, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; KAIST Institute for Health Science and Technology, 291, Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
34
|
Edmister ST, Creton R. Modulation of calcineurin signaling during development. Dev Neurobiol 2022; 82:505-516. [PMID: 35785416 PMCID: PMC9922048 DOI: 10.1002/dneu.22895] [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] [Received: 03/31/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/07/2022]
Abstract
Calcineurin signaling pathways are suppressed in Down syndrome (trisomy 21), by overexpression of genes that are located on chromosome 21. Two key genes are the regulator of calcineurin 1 (RCAN1), also called the Down syndrome critical region 1 (DSCR1), and the dual-specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A). The suppressed calcineurin pathway may potentially be restored using small-molecule DYRK inhibitors, which have been proposed as therapeutics in Down syndrome. However, little is known about the benefits and risks of such treatments during various stages of embryonic development, fetal development, and childhood. We examined the modulation of calcineurin signaling during development, using zebrafish as a model system. To mimic suppressed calcineurin signaling in Down syndrome, zebrafish were exposed to the calcineurin inhibitors cyclosporine and tacrolimus during development. We found that suppression of calcineurin signaling changed specific larval behaviors, including activity and responses to acoustic and visual stimuli, depending on the period of exposure. Cotreatment with the DYRK inhibitor proINDY restored a few of these behaviors but also induced a range of adverse side effects including decreased activity and reduced optomotor responses to visual stimuli. Based on these results, we conclude that proINDY has limited benefits and substantial risks when used during development. We propose that zebrafish is an efficient model system for preliminary safety and efficacy tests of other DYRK inhibitors that aim to restore calcineurin signaling during neural development.
Collapse
Affiliation(s)
- Sara Tucker Edmister
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Robbert Creton
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
35
|
Chin JSR, Phan TAN, Albert LT, Keene AC, Duboué ER. Long lasting anxiety following early life stress is dependent on glucocorticoid signaling in zebrafish. Sci Rep 2022; 12:12826. [PMID: 35896563 PMCID: PMC9329305 DOI: 10.1038/s41598-022-16257-5] [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: 03/22/2022] [Accepted: 07/07/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic adversity in early childhood is associated with increased anxiety and a propensity for substance abuse later in adulthood, yet the effects of early life stress (ELS) on brain development remain poorly understood. The zebrafish, Danio rerio, is a powerful model for studying neurodevelopment and stress. Here, we describe a zebrafish model of ELS and identify a role for glucocorticoid signaling during a critical window in development that leads to long-term changes in brain function. Larval fish subjected to chronic stress in early development exhibited increased anxiety-like behavior and elevated glucocorticoid levels later in life. Increased stress-like behavior was only observed when fish were subjected to ELS within a precise time window in early development, revealing a temporal critical window of sensitivity. Moreover, enhanced anxiety-like behavior only emerges after two months post-ELS, revealing a developmentally specified delay in the effects of ELS. ELS leads to increased levels of baseline cortisol, and resulted in a dysregulation of cortisol receptors' mRNA expression, suggesting long-term effects on cortisol signaling. Together, these findings reveal a 'critical window' for ELS to affect developmental reprogramming of the glucocorticoid receptor pathway, resulting in chronic elevated stress.
Collapse
Affiliation(s)
- Jacqueline S R Chin
- Jupiter Life Science Initiative, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33407, USA
| | - Tram-Anh N Phan
- Jupiter Life Science Initiative, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33407, USA
| | - Lydia T Albert
- Jupiter Life Science Initiative, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33407, USA
| | - Alex C Keene
- College of Arts and Sciences, Department of Biology, Texas A&M University, 3258 TAMU, College Station, TX, 77843, USA
| | - Erik R Duboué
- Jupiter Life Science Initiative, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33407, USA.
| |
Collapse
|
36
|
Ando K, Tong L, Peng D, Vázquez-Liébanas E, Chiyoda H, He L, Liu J, Kawakami K, Mochizuki N, Fukuhara S, Grutzendler J, Betsholtz C. KCNJ8/ABCC9-containing K-ATP channel modulates brain vascular smooth muscle development and neurovascular coupling. Dev Cell 2022; 57:1383-1399.e7. [PMID: 35588738 DOI: 10.1016/j.devcel.2022.04.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 12/22/2021] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Loss- or gain-of-function mutations in ATP-sensitive potassium channel (K-ATP)-encoding genes, KCNJ8 and ABCC9, cause human central nervous system disorders with unknown pathogenesis. Here, using mice, zebrafish, and cell culture models, we investigated cellular and molecular causes of brain dysfunctions derived from altered K-ATP channel function. We show that genetic/chemical inhibition or activation of KCNJ8/ABCC9-containing K-ATP channel function leads to brain-selective suppression or promotion of arterial/arteriolar vascular smooth muscle cell (VSMC) differentiation, respectively. We further show that brain VSMCs develop from KCNJ8/ABCC9-containing K-ATP channel-expressing mural cell progenitor and that K-ATP channel cell autonomously regulates VSMC differentiation through modulation of intracellular Ca2+ oscillation via voltage-dependent calcium channels. Consistent with defective VSMC development, Kcnj8 knockout mice showed deficiency in vasoconstrictive capacity and neuronal-evoked vasodilation leading to local hyperemia. Our results demonstrate a role for KCNJ8/ABCC9-containing K-ATP channels in the differentiation of brain VSMC, which in turn is necessary for fine-tuning of cerebral blood flow.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan.
| | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Hirohisa Chiyoda
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan; Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden
| | - Jianping Liu
- Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan; Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka 564-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Science, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8602, Japan
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institute, Campus Flemingsburg, Neo, Blickagången 16, 141 57 Huddinge, Sweden.
| |
Collapse
|
37
|
Fassina L, Assenza MR, Miragoli M, Isidori AM, Naro F, Barbagallo F. Cell Shortening and Calcium Homeostasis Analysis in Adult Cardiomyocytes via a New Software Tool. Biomedicines 2022; 10:biomedicines10030640. [PMID: 35327442 PMCID: PMC8945339 DOI: 10.3390/biomedicines10030640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Intracellular calcium (Ca2+) is the central regulator of heart contractility. Indeed, it couples the electrical signal, which pervades the myocardium, with cardiomyocytes contraction. Moreover, alterations in calcium management are the main factors contributing to the mechanical and electrical dysfunction observed in failing hearts. So, simultaneous analysis of the contractile function and intracellular Ca2+ is indispensable to evaluate cardiomyocytes activity. Intracellular Ca2+ variations and fraction shortening are commonly studied with fluorescent Ca2+ indicator dyes associated with microscopy techniques. However, tracking and dealing with multiple files manually is time-consuming and error-prone and often requires expensive apparatus and software. Here, we announce a new, user-friendly image processing and analysis tool, based on ImageJ-Fiji/MATLAB® software, to evaluate the major cardiomyocyte functional parameters. We succeeded in analyzing fractional cell shortening, Ca2+ transient amplitude, and the kinematics/dynamics parameters of mouse isolated adult cardiomyocytes. The proposed method can be applied to evaluate changes in the Ca2+ cycle and contractile behavior in genetically or pharmacologically induced disease models, in drug screening and other common applications to assess mammalian cardiomyocyte functions.
Collapse
Affiliation(s)
- Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering (DIII), University of Pavia, 27100 Pavia, Italy;
| | - Maria Rita Assenza
- Institute of Biochemistry and Cell Biology, CNR, 00015 Monterotondo, Italy;
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, 00161 Rome, Italy;
| | - Michele Miragoli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
- Humanitas Research Hospital—IRCCS, 20089 Rozzano, Italy
| | - Andrea M. Isidori
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy;
| | - Fabio Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, 00161 Rome, Italy;
| | - Federica Barbagallo
- Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy;
- Correspondence:
| |
Collapse
|
38
|
Nathan FM, Kibat C, Goel T, Stewart J, Claridge‐Chang A, Mathuru AS. Contingent stimulus delivery assay for zebrafish reveals a role for CCSER1 in alcohol preference. Addict Biol 2022; 27:e13126. [PMID: 35229935 DOI: 10.1111/adb.13126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/02/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022]
Abstract
Alcohol use disorders are complex, multifactorial phenomena with a large footprint within the global burden of diseases. Here, we report the development of an accessible, two-choice self-administration zebrafish assay (SAZA) to study the neurobiology of addiction. Using this assay, we first demonstrated that, although zebrafish avoid higher concentrations of alcohol, they are attracted to low concentrations. Pre-exposure to alcohol did not change this relative preference, but acute exposure to an alcohol deterrent approved for human use decreased alcohol self-administration. A pigment mutant used in whole-brain imaging studies displayed a similar relative alcohol preference profile; however, mutants in CCSER1, a gene associated with alcohol dependence in human genetic studies, showed a reversal in relative preference. The presence of a biphasic response (hormesis) in zebrafish validated a key aspect of vertebrate responses to alcohol. SAZA adds a new dimension for discovering novel alcohol deterrents and studying the neurogenetics of addiction using the zebrafish.
Collapse
Affiliation(s)
| | - Caroline Kibat
- Department of Physiology, YLL School of Medicine National University of Singapore Singapore Singapore
| | - Tanisha Goel
- Department of Physiology, YLL School of Medicine National University of Singapore Singapore Singapore
| | - James Stewart
- Institute of Molecular and Cell Biology Singapore Singapore
- Duke‐NUS Medical School Singapore Singapore
| | - Adam Claridge‐Chang
- Institute of Molecular and Cell Biology Singapore Singapore
- Duke‐NUS Medical School Singapore Singapore
| | - Ajay S. Mathuru
- Yale‐NUS College Singapore Singapore
- Department of Physiology, YLL School of Medicine National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology Singapore Singapore
| |
Collapse
|
39
|
Topographic map formation and the effects of NMDA receptor blockade in the developing visual system. Proc Natl Acad Sci U S A 2022; 119:2107899119. [PMID: 35193956 PMCID: PMC8872792 DOI: 10.1073/pnas.2107899119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2022] [Indexed: 11/18/2022] Open
Abstract
Studying the emergence of topographic organization in sensory maps has been constrained by spatial limitations of traditional anatomical and physiological techniques early in development in many animal models. Here, we have applied a high-resolution, noninvasive, in vivo calcium imaging approach to study the nascent retinotopic map in the larval Xenopus laevis retinotectal system. We performed longitudinal functional imaging of the three-dimensional organization of emerging retinotopic maps and assessed the effects of N-methyl-D-aspartate (NMDA) receptor blockade on map formation. Our results provide insights into early retinotopic map emergence and the role of NMDA receptors in the refinement of topographic gradients. The development of functional topography in the developing brain follows a progression from initially coarse to more precisely organized maps. To examine the emergence of topographically organized maps in the retinotectal system, we performed longitudinal visual receptive field mapping by calcium imaging in the optic tectum of GCaMP6-expressing transgenic Xenopus laevis tadpoles. At stage 42, just 1 d after retinal axons arrived in the optic tectum, a clear retinotopic azimuth map was evident. Animals were imaged over the following week at stages 45 and 48, over which time the tectal neuropil nearly doubled in length and exhibited more precise retinotopic organization. By microinjecting GCaMP6s messenger ribonucleic acid (mRNA) into one blastomere of two-cell stage embryos, we acquired bilateral mosaic tadpoles with GCaMP6s expression in postsynaptic tectal neurons on one side of the animal and in retinal ganglion cell axons crossing to the tectum on the opposite side. Longitudinal observation of retinotopic map emergence revealed the presence of orderly representations of azimuth and elevation as early as stage 42, although presynaptic inputs exhibited relatively less topographic organization than the postsynaptic component for the azimuth axis. Retinotopic gradients in the tectum became smoother between stages 42 and 45. Blocking N-methyl-D-aspartate (NMDA) receptor conductance by rearing tadpoles in MK-801 did not prevent the emergence of retinotopic maps, but it produced more discontinuous topographic gradients and altered receptive field characteristics. These results provide evidence that current through NMDA receptors is dispensable for coarse topographic ordering of retinotectal inputs but does contribute to the fine-scale organization of the retinotectal projection.
Collapse
|
40
|
Terai H, Gwedela MNV, Kawakami K, Aizawa H. Electrophysiological and pharmacological characterization of spreading depolarization in the adult zebrafish tectum. J Neurophysiol 2021; 126:1934-1942. [PMID: 34731067 DOI: 10.1152/jn.00343.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Spreading depolarization (SD) is a slowly propagating wave of neuronal and glial depolarization. A growing number of studies show that SD and SD-like phenomena play a role in neurological disorders such as migraine, stroke, and traumatic brain injury. Despite the clinical importance of SD, its underlying molecular and cellular mechanisms remain elusive, possibly because of insufficient animal model allowing genetic manipulation. Such a model would also allow high-throughput screening for SD-suppressing drug development. To address this, we developed a novel experimental system to study SD using zebrafish. Electrophysiological recordings in the immobilized adult zebrafish revealed that increasing extracellular potassium concentration elicited SD with a large and long-lasting negative shift of direct current (DC) potential in the optic tectum. It also reduced the oscillatory activity in the extracellular field potential and increased the expression of the immediate early gene c-fos. Pharmacological blocking of the N-methyl-d-aspartate (NMDA) glutamate receptor attenuated the propagation of SD, suggesting that glutamatergic neurotransmission mediated tectal SD in zebrafish. Our analyses revealed that the zebrafish tectum and rodent cortex had similar SD kinetics. The current study provides electrophysiological and pharmacological evidence that zebrafish SD and mammal SD are comparable. This zebrafish SD model is suitable for genetic manipulation and cost-effective high-throughput screening. It could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.NEW & NOTEWORTHY Previous studies have implicated spreading depolarization (SD) in stroke and migraine. Here, we demonstrate SD, for the first time, in the adult zebrafish tectum showing waveform kinetics, c-fos expression, and attenuation by N-methyl-d-aspartate glutamate receptor blocker as observed in the rodent cortex. Since the zebrafish is an animal model amenable to genetic manipulation and chemical screening, this result could pave the way to novel diagnostic and therapeutic methods applicable to SD-associated neurological disorders.
Collapse
Affiliation(s)
- Haruhi Terai
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mayeso Naomi Victoria Gwedela
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Shizuoka, Japan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
41
|
Fukui H, Chow RWY, Xie J, Foo YY, Yap CH, Minc N, Mochizuki N, Vermot J. Bioelectric signaling and the control of cardiac cell identity in response to mechanical forces. Science 2021; 374:351-354. [PMID: 34648325 DOI: 10.1126/science.abc6229] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hajime Fukui
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France
| | - Jing Xie
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Yoke Yin Foo
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Choon Hwai Yap
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Department of Bioengineering, Imperial College London, London, UK
| | - Nicolas Minc
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
42
|
Cho ES, Han S, Lee KH, Kim CH, Yoon YG. 3DM: deep decomposition and deconvolution microscopy for rapid neural activity imaging. OPTICS EXPRESS 2021; 29:32700-32711. [PMID: 34615335 DOI: 10.1364/oe.439619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/14/2021] [Indexed: 05/18/2023]
Abstract
We report the development of deep decomposition and deconvolution microscopy (3DM), a computational microscopy method for the volumetric imaging of neural activity. 3DM overcomes the major challenge of deconvolution microscopy, the ill-posed inverse problem. We take advantage of the temporal sparsity of neural activity to reformulate and solve the inverse problem using two neural networks which perform sparse decomposition and deconvolution. We demonstrate the capability of 3DM via in vivo imaging of the neural activity of a whole larval zebrafish brain with a field of view of 1040 µm × 400 µm × 235 µm and with estimated lateral and axial resolutions of 1.7 µm and 5.4 µm, respectively, at imaging rates of up to 4.2 volumes per second.
Collapse
|
43
|
Rodríguez F, Quintero B, Amores L, Madrid D, Salas-Peña C, Salas C. Spatial Cognition in Teleost Fish: Strategies and Mechanisms. Animals (Basel) 2021; 11:2271. [PMID: 34438729 PMCID: PMC8388456 DOI: 10.3390/ani11082271] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 01/25/2023] Open
Abstract
Teleost fish have been traditionally considered primitive vertebrates compared to mammals and birds in regard to brain complexity and behavioral functions. However, an increasing amount of evidence suggests that teleosts show advanced cognitive capabilities including spatial navigation skills that parallel those of land vertebrates. Teleost fish rely on a multiplicity of sensory cues and can use a variety of spatial strategies for navigation, ranging from relatively simple body-centered orientation responses to allocentric or "external world-centered" navigation, likely based on map-like relational memory representations of the environment. These distinct spatial strategies are based on separate brain mechanisms. For example, a crucial brain center for egocentric orientation in teleost fish is the optic tectum, which can be considered an essential hub in a wider brain network responsible for the generation of egocentrically referenced actions in space. In contrast, other brain centers, such as the dorsolateral telencephalic pallium of teleost fish, considered homologue to the hippocampal pallium of land vertebrates, seem to be crucial for allocentric navigation based on map-like spatial memory. Such hypothetical relational memory representations endow fish's spatial behavior with considerable navigational flexibility, allowing them, for example, to perform shortcuts and detours.
Collapse
Affiliation(s)
| | | | | | | | | | - Cosme Salas
- Laboratorio de Psicobiología, Universidad de Sevilla, 41018 Sevilla, Spain; (F.R.); (B.Q.); (L.A.); (D.M.); (C.S.-P.)
| |
Collapse
|
44
|
De Faveri F, Marcotti W, Ceriani F. Sensory adaptation at ribbon synapses in the zebrafish lateral line. J Physiol 2021; 599:3677-3696. [PMID: 34047358 PMCID: PMC7612133 DOI: 10.1113/jp281646] [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: 03/15/2021] [Accepted: 05/20/2021] [Indexed: 11/22/2022] Open
Abstract
Adaptation is used by sensory systems to adjust continuously their sensitivity to match changes in environmental stimuli. In the auditory and vestibular systems, the release properties of glutamate-containing vesicles at the hair-cell ribbon synapses play a crucial role in sensory adaptation, thus shaping the neural response to sustained stimulation. How ribbon synapses regulate the release of glutamate and how they modulate afferent responses in vivo is still largely unknown. Here, we have used two-photon imaging and electrophysiology to investigate the synaptic transfer characteristics of the hair cells in the context of sensory adaptation in live zebrafish. Prolonged and repeated water-jet stimulation of the hair-cell stereociliary bundles caused adaptation of the action potential firing rate elicited in the afferent neurons. By monitoring glutamate at ribbon synapses using time-lapse imaging, we identified two kinetically distinct release components: a rapid response that was exhausted within 50-100 ms and a slower and sustained response lasting the entire stimulation. After repeated stimulations, the recovery of the fast component followed a biphasic time course. Depression of glutamate release was largely responsible for the rapid firing rate adaptation recorded in the afferent neurons. However, postsynaptic Ca2+ responses had a slower recovery time course than that of glutamate release, indicating that they are also likely to contribute to the afferent firing adaptation. Hair cells also exhibited a form of adaptation during inhibitory bundle stimulations. We conclude that hair cells have optimised their synaptic machinery to encode prolonged stimuli and to maintain their sensitivity to new incoming stimuli.
Collapse
Affiliation(s)
| | - Walter Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield, UK.,Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Federico Ceriani
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| |
Collapse
|
45
|
Optogenetic Manipulation of Olfactory Responses in Transgenic Zebrafish: A Neurobiological and Behavioral Study. Int J Mol Sci 2021; 22:ijms22137191. [PMID: 34281244 PMCID: PMC8269104 DOI: 10.3390/ijms22137191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/01/2021] [Indexed: 11/18/2022] Open
Abstract
Olfaction is an important neural system for survival and fundamental behaviors such as predator avoidance, food finding, memory formation, reproduction, and social communication. However, the neural circuits and pathways associated with the olfactory system in various behaviors are not fully understood. Recent advances in optogenetics, high-resolution in vivo imaging, and reconstructions of neuronal circuits have created new opportunities to understand such neural circuits. Here, we generated a transgenic zebrafish to manipulate olfactory signal optically, expressing the Channelrhodopsin (ChR2) under the control of the olfactory specific promoter, omp. We observed light-induced neuronal activity of olfactory system in the transgenic fish by examining c-fos expression, and a calcium indicator suggesting that blue light stimulation caused activation of olfactory neurons in a non-invasive manner. To examine whether the photo-activation of olfactory sensory neurons affect behavior of zebrafish larvae, we devised a behavioral choice paradigm and tested how zebrafish larvae choose between two conflicting sensory cues, an aversive odor or the naturally preferred phototaxis. We found that when the conflicting cues (the preferred light and aversive odor) were presented together simultaneously, zebrafish larvae swam away from the aversive odor. However, the transgenic fish with photo-activation were insensitive to the aversive odor and exhibited olfactory desensitization upon optical stimulation of ChR2. These results show that an aversive olfactory stimulus can override phototaxis, and that olfaction is important in decision making in zebrafish. This new transgenic model will be useful for the analysis of olfaction related behaviors and for the dissection of underlying neural circuits.
Collapse
|
46
|
Xu J, Casanave R, Guo S. Larval zebrafish display dynamic learning of aversive stimuli in a constant visual surrounding. ACTA ACUST UNITED AC 2021; 28:228-238. [PMID: 34131054 PMCID: PMC8212779 DOI: 10.1101/lm.053425.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022]
Abstract
Balancing exploration and anti-predation are fundamental to the fitness and survival of all animal species from early life stages. How these basic survival instincts drive learning remains poorly understood. Here, using a light/dark preference paradigm with well-controlled luminance history and constant visual surrounding in larval zebrafish, we analyzed intra- and intertrial dynamics for two behavioral components, dark avoidance and center avoidance. We uncover that larval zebrafish display short-term learning of dark avoidance with initial sensitization followed by habituation; they also exhibit long-term learning that is sensitive to trial interval length. We further show that such stereotyped learning patterns is stimulus-specific, as they are not observed for center avoidance. Finally, we demonstrate at individual levels that long-term learning is under homeostatic control. Together, our work has established a novel paradigm to understand learning, uncovered sequential sensitization and habituation, and demonstrated stimulus specificity, individuality, as well as dynamicity in learning.
Collapse
Affiliation(s)
- Jiale Xu
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA
| | - Romelo Casanave
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, University of California at San Francisico, San Francisco, California 94158, USA.,Program in Human Genetics, University of California at San Francisco, San Francisco, California 94158, USA.,Program in Biological Sciences, University of California at San Francisco, San Francisco, California 94158, USA
| |
Collapse
|
47
|
In vivo calcium imaging reveals disordered interictal network dynamics in epileptic stxbp1b zebrafish. iScience 2021; 24:102558. [PMID: 34142057 PMCID: PMC8184515 DOI: 10.1016/j.isci.2021.102558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
STXBP1 mutations are associated with encephalopathy, developmental delay, intellectual disability, and epilepsy. While neural networks are known to operate at a critical state in the healthy brain, network behavior during pathological epileptic states remains unclear. Examining activity during periods between well-characterized ictal-like events (i.e., interictal period) could provide a valuable step toward understanding epileptic networks. To study these networks in the context of STXBP1 mutations, we combine a larval zebrafish model with in vivo fast confocal calcium imaging and extracellular local field potential recordings. Stxbp1b mutants display transient periods of elevated activity among local clusters of interacting neurons. These network "cascade" events were significantly larger in size and duration in mutants. At mesoscale resolution, cascades exhibit neurodevelopmental abnormalities. At single-cell scale, we describe spontaneous hyper-synchronized neuronal ensembles. That calcium imaging reveals uniquely disordered brain states during periods between pathological ictal-like seizure events is striking and represents a potential interictal biomarker.
Collapse
|
48
|
Friedrich RW, Wanner AA. Dense Circuit Reconstruction to Understand Neuronal Computation: Focus on Zebrafish. Annu Rev Neurosci 2021; 44:275-293. [PMID: 33730512 DOI: 10.1146/annurev-neuro-110220-013050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The dense reconstruction of neuronal wiring diagrams from volumetric electron microscopy data has the potential to generate fundamentally new insights into mechanisms of information processing and storage in neuronal circuits. Zebrafish provide unique opportunities for dynamical connectomics approaches that combine reconstructions of wiring diagrams with measurements of neuronal population activity and behavior. Such approaches have the power to reveal higher-order structure in wiring diagrams that cannot be detected by sparse sampling of connectivity and that is essential for neuronal computations. In the brain stem, recurrently connected neuronal modules were identified that can account for slow, low-dimensional dynamics in an integrator circuit. In the spinal cord, connectivity specifies functional differences between premotor interneurons. In the olfactory bulb, tuning-dependent connectivity implements a whitening transformation that is based on the selective suppression of responses to overrepresented stimulus features. These findings illustrate the potential of dynamical connectomics in zebrafish to analyze the circuit mechanisms underlying higher-order neuronal computations.
Collapse
Affiliation(s)
- Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; .,Faculty of Natural Sciences, University of Basel, 4003 Basel, Switzerland
| | - Adrian A Wanner
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544, USA;
| |
Collapse
|
49
|
Bian Z, Guo T, Jiang S, Chen L, Liu J, Zheng G, Feng B. High-Throughput Functional Characterization of Visceral Afferents by Optical Recordings From Thoracolumbar and Lumbosacral Dorsal Root Ganglia. Front Neurosci 2021; 15:657361. [PMID: 33776645 PMCID: PMC7991386 DOI: 10.3389/fnins.2021.657361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 02/22/2021] [Indexed: 12/27/2022] Open
Abstract
Functional understanding of visceral afferents is important for developing the new treatment to visceral hypersensitivity and pain. The sparse distribution of visceral afferents in dorsal root ganglia (DRGs) has challenged conventional electrophysiological recordings. Alternatively, Ca2+ indicators like GCaMP6f allow functional characterization by optical recordings. Here we report a turnkey microscopy system that enables simultaneous Ca2+ imaging at two parallel focal planes from intact DRG. By using consumer-grade optical components, the microscopy system is cost-effective and can be made broadly available without loss of capacity. It records low-intensity fluorescent signals at a wide field of view (1.9 × 1.3 mm) to cover a whole mouse DRG, with a high pixel resolution of 0.7 micron/pixel, a fast frame rate of 50 frames/sec, and the capability of remote focusing without perturbing the sample. The wide scanning range (100 mm) of the motorized sample stage allows convenient recordings of multiple DRGs in thoracic, lumbar, and sacral vertebrae. As a demonstration, we characterized mechanical neural encoding of visceral afferents innervating distal colon and rectum (colorectum) in GCaMP6f mice driven by VGLUT2 promotor. A post-processing routine is developed for conducting unsupervised detection of visceral afferent responses from GCaMP6f recordings, which also compensates the motion artifacts caused by mechanical stimulation of the colorectum. The reported system offers a cost-effective solution for high-throughput recordings of visceral afferent activities from a large volume of DRG tissues. We anticipate a wide application of this microscopy system to expedite our functional understanding of visceral innervations.
Collapse
Affiliation(s)
- Zichao Bian
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Shaowei Jiang
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Longtu Chen
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Jia Liu
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Guoan Zheng
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Mansfield, CT, United States
| |
Collapse
|
50
|
Gao L, Penglee R, Huang Y, Yi X, Wang X, Liu L, Gong X, Bao B. CRISPR/Cas9-induced nos2b mutant zebrafish display behavioral abnormalities. GENES BRAIN AND BEHAVIOR 2020; 20:e12716. [PMID: 33200539 DOI: 10.1111/gbb.12716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 11/29/2022]
Abstract
The immunomodulatory function of nitric oxide synthase (NOS2) has been extensively studied. However, some behavioral abnormalities caused by its mutations have been found in a few rodent studies, of which the molecular mechanism remains elusive. In this research, we generated nos2b gene knockout zebrafish (nos2bsou2/sou2 ) using CRISPR/Cas9 approach and investigated their behavioral and molecular changes by doing a series of behavioral detections, morphological measurements, and molecular analyses. We found that, compared with nos2b+/+ zebrafish, nos2bsou2/sou2 zebrafish exhibited enhanced motor activity; additionally, nos2bsou2/sou2 zebrafish were characterized by smaller brain size, abnormal structure of optic tectum, reduced mRNA level of presynaptic synaptophysin and postsynaptic homer1, and altered response to sodium nitroprusside/methylphenidate hydrochloride treatment. These findings will likely contribute to future studies of behavioral regulation.
Collapse
Affiliation(s)
- Lei Gao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Rachit Penglee
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Yajuan Huang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xinxin Yi
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaojie Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Liping Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaoling Gong
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Baolong Bao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| |
Collapse
|