1
|
Odierna GL, Stednitz S, Pruitt A, Arnold J, Hoffman EJ, Scott EK. Generation of stable brain cell cultures from embryonic zebrafish to interrogate phenotypes in zebrafish mutants of neurodevelopmental disorders. J Neurosci Methods 2025; 418:110426. [PMID: 40086601 DOI: 10.1016/j.jneumeth.2025.110426] [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: 12/05/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Zebrafish are a popular model system to study the genetic and neural basis of perception and behavior. Cultured primary neurons provide a complementary tool for such studies, but existing protocols for culturing embryonic zebrafish neurons are limited by short cell survival and low neuronal purity. In this study, we set out to establish a protocol to produce long lived brain cell cultures from zebrafish that could be used to study the mechanistic contributions of genes to neuronal networks. NEW METHOD This protocol improves the viability of embryonic zebrafish primary brain cell cultures. We successfully optimized several parameters to generate long lived mixed cell type or pure neuronal cultures derived from embryonic zebrafish. RESULTS Our optimized protocol produces cultures that form stable networks of neurons expressing the structural hallmarks of mature synaptic connections. As proof of principle, we apply our protocol to explore the cellular consequences of scn1lab loss of function. We find that loss of scn1lab results in increased prevalence of non-neuronal cells consistent with transcriptional signatures from embryonic tissue, providing support for the utility of our protocol. COMPARISON WITH EXISTING METHOD(S) Most existing embryonic zebrafish primary neuron culture protocols describe growing mixed cell types for short durations, with a reported maximum of 9 days in vitro. Here, we describe a protocol that produces cultures viable for over 100 days. CONCLUSIONS The protocol reported in this study raises embryonic zebrafish primary brain cell culture to similar standards observed by well-established methods using cell lines or mammalian tissue.
Collapse
Affiliation(s)
- G Lorenzo Odierna
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia.
| | - Sarah Stednitz
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - April Pruitt
- Child Study Center, Program on Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, United States; Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Joshua Arnold
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Ellen J Hoffman
- Child Study Center, Program on Neurogenetics, Yale School of Medicine, Yale University, New Haven, CT, United States; Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Ethan K Scott
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland, Australia; Department of Anatomy and Physiology, The University of Melbourne, Australia
| |
Collapse
|
2
|
Rerkamnuaychoke W, Sreevidya VS, Svoboda KR. Chloroxylenol and benzethonium chloride exposure alters spinal neuron development and behavior in developing zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:117993. [PMID: 40209347 DOI: 10.1016/j.ecoenv.2025.117993] [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: 09/15/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 04/12/2025]
Abstract
Chloroxylenol (CHX) and benzethonium chloride (BEC) are replacement compounds being used after the ban (U.S. FDA) of triclosan and triclocarban in 2016 from personal care products. These two compounds are also recommended by the World Health Organization (WHO) as disinfectants against COVID-19. Toxicity data for both CHX and BEC are available, however, neurotoxicity studies for both compounds are still limited. Here, we determined the consequences of CHX and BEC exposure in a static exposure paradigm during embryogenesis on neurodevelopment using the zebrafish model (Danio rerio). CHX exposure (1-5 mg/L) dramatically impacted nervous system development without increasing mortality. The exposure altered embryonic motor output, primary motoneuron cell size, spinal interneuron cell size, primary motoneuron (PMN) axon pathfinding and secondary motoneuron (SMN) axon pathfinding. CHX exposure also altered slow muscle fiber development. Changes in neural activity as revealed by the induced changes in embryonic motor output (spontaneous coiling) may underlie the errors in PMN axon pathfinding as well as the changes in spinal interneuron morphology. Errors in SMN axon pathfinding resulting from CHX exposure were directly linked to errors in PMN axon pathfinding. Similar to CHX, BEC exposure (1-5 mg/L) altered embryonic motor output, spinal interneuron development, and slow muscle fiber development. In contrast to CHX, BEC exposure did not alter PMN or SMN axon pathfinding. Moreover, we found that BEC exposure (5 mg/L) from 5 to 96 hpf was lethal, resulting in almost 100 % mortality. Thus, these two triclosan replacement compounds exhibited markedly different modes of toxicity.
Collapse
Affiliation(s)
- Wuttiporn Rerkamnuaychoke
- Joseph J. Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53205, USA
| | - Virinchipuram S Sreevidya
- Joseph J. Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53205, USA
| | - Kurt R Svoboda
- Joseph J. Zilber College of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53205, USA.
| |
Collapse
|
3
|
Kawahara A, Yasojima S, Koiwa J, Fujimaki S, Ito H, Yamada M, Kosaki K, Nishimura Y. Establishment and characterization of adap1-deficient zebrafish. Dev Growth Differ 2025; 67:165-173. [PMID: 40088134 PMCID: PMC11997736 DOI: 10.1111/dgd.70004] [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: 05/18/2024] [Revised: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
The adap1 (ADP-ribosylation factor GTPase-activating protein [ArfGAP] with dual pleckstrin homology [PH] domains 1) gene is predominantly expressed in the mouse brain and is important in neural differentiation and development. However, the functions of adap1 in morphogenesis, locomotor activity, and behaviors in vertebrates are not fully understood. Whole-mount in situ hybridization (WISH) analysis revealed that adap1 was widely expressed in the zebrafish brain, including the forebrain, midbrain, and hindbrain, during early embryogenesis. To investigate the physiological function of the adap1 gene, we generated zebrafish adap1 mutants harboring frameshift mutations around codon 120 of adap1. The adap1 mutants containing homozygous mutant alleles exhibited no apparent morphological abnormalities at 1 day postfertilization (dpf), and the spontaneous coiling and touch response of the adap1 mutants were comparable to those of the wild-type fish. In addition, the expression of neural genes, such as emx1, mbx, and huC, was comparable between the wild-type fish and the adap1 mutants at 1 dpf. The adap1 mutants grew to adulthood without exhibiting any apparent swimming defects. The adult adap1 mutants spent more time than the wild type in the center region of the open field test. In the social behavior test, zebrafish containing the adap1 mutant alleles spent more time than the wild type in the regions near the chambers where novel conspecifics swam. These results imply the involvement of the adap1 gene in regulating approach behavior to visual cues from conspecifics.
Collapse
Affiliation(s)
- Atsuo Kawahara
- Laboratory for Developmental Biology, Center for Medical Education and Sciences, Graduate School of Medical ScienceUniversity of YamanashiYamanashiJapan
| | - Sakyo Yasojima
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuJapan
| | - Junko Koiwa
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuJapan
| | - Saori Fujimaki
- Laboratory for Developmental Biology, Center for Medical Education and Sciences, Graduate School of Medical ScienceUniversity of YamanashiYamanashiJapan
| | - Hiroaki Ito
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuJapan
| | - Mamiko Yamada
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | - Kenjiro Kosaki
- Center for Medical GeneticsKeio University School of MedicineTokyoJapan
| | - Yuhei Nishimura
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuJapan
| |
Collapse
|
4
|
Wang X, Yue M, Cheung JPY, Cheung PWH, Fan Y, Wu M, Wang X, Zhao S, Khanshour AM, Rios JJ, Chen Z, Wang X, Tu W, Chan D, Yuan Q, Qin D, Qiu G, Wu Z, Zhang TJ, Ikegawa S, Wu N, Wise CA, Hu Y, Luk KDK, Song YQ, Gao B. Impaired glycine neurotransmission causes adolescent idiopathic scoliosis. J Clin Invest 2024; 134:e168783. [PMID: 37962965 PMCID: PMC10786698 DOI: 10.1172/jci168783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is the most common form of spinal deformity, affecting millions of adolescents worldwide, but it lacks a defined theory of etiopathogenesis. Because of this, treatment of AIS is limited to bracing and/or invasive surgery after onset. Preonset diagnosis or preventive treatment remains unavailable. Here, we performed a genetic analysis of a large multicenter AIS cohort and identified disease-causing and predisposing variants of SLC6A9 in multigeneration families, trios, and sporadic patients. Variants of SLC6A9, which encodes glycine transporter 1 (GLYT1), reduced glycine-uptake activity in cells, leading to increased extracellular glycine levels and aberrant glycinergic neurotransmission. Slc6a9 mutant zebrafish exhibited discoordination of spinal neural activities and pronounced lateral spinal curvature, a phenotype resembling human patients. The penetrance and severity of curvature were sensitive to the dosage of functional glyt1. Administration of a glycine receptor antagonist or a clinically used glycine neutralizer (sodium benzoate) partially rescued the phenotype. Our results indicate a neuropathic origin for "idiopathic" scoliosis, involving the dysfunction of synaptic neurotransmission and central pattern generators (CPGs), potentially a common cause of AIS. Our work further suggests avenues for early diagnosis and intervention of AIS in preadolescents.
Collapse
Affiliation(s)
- Xiaolu Wang
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ming Yue
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
| | - Prudence Wing Hang Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yanhui Fan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Meicheng Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaojun Wang
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sen Zhao
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Anas M. Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
| | - Jonathan J. Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
- Eugene McDermott Center for Human Growth and Development, Departments of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zheyi Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Tai Po, Hong Kong, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Tai Po, Hong Kong, China
| | - Guixing Qiu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Terry Jianguo Zhang
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Nan Wu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Carol A. Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
- Eugene McDermott Center for Human Growth and Development, Departments of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yong Hu
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
| | - Keith Dip Kei Luk
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - You-Qiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Medicine, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Brain and Cognitive Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Bo Gao
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
- Centre for Translational Stem Cell Biology, Tai Po, Hong Kong, China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
5
|
von Hellfeld R, Gade C, Baumann L, Leist M, Braunbeck T. The sensitivity of the zebrafish embryo coiling assay for the detection of neurotoxicity by compounds with diverse modes of action. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023:10.1007/s11356-023-27662-2. [PMID: 37213015 DOI: 10.1007/s11356-023-27662-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
In the aim to determine neurotoxicity, new methods are being validated, including tests and test batteries comprising in vitro and in vivo approaches. Alternative test models such as the zebrafish (Danio rerio) embryo have received increasing attention, with minor modifications of the fish embryo toxicity test (FET; OECD TG 236) as a tool to assess behavioral endpoints related to neurotoxicity during early developmental stages. The spontaneous tail movement assay, also known as coiling assay, assesses the development of random movement into complex behavioral patterns and has proven sensitive to acetylcholine esterase inhibitors at sublethal concentrations. The present study explored the sensitivity of the assay to neurotoxicants with other modes of action (MoAs). Here, five compounds with diverse MoAs were tested at sublethal concentrations: acrylamide, carbaryl, hexachlorophene, ibuprofen, and rotenone. While carbaryl, hexachlorophene, and rotenone consistently induced severe behavioral alterations by ~ 30 h post fertilization (hpf), acrylamide and ibuprofen expressed time- and/or concentration-dependent effects. At 37-38 hpf, additional observations revealed behavioral changes during dark phases with a strict concentration-dependency. The study documented the applicability of the coiling assay to MoA-dependent behavioral alterations at sublethal concentrations, underlining its potential as a component of a neurotoxicity test battery.
Collapse
Affiliation(s)
- Rebecca von Hellfeld
- School of Biological Sciences, University of Aberdeen, 23 St Machar Drive, Aberdeen, AB24 3UK, UK.
- National Decommissioning Centre, Main Street, Ellon, AB41 6AA, UK.
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany.
| | - Christoph Gade
- School of Biological Sciences, University of Aberdeen, 23 St Machar Drive, Aberdeen, AB24 3UK, UK
- National Decommissioning Centre, Main Street, Ellon, AB41 6AA, UK
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Lisa Baumann
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
- Faculty of Science, Environmental Health & Toxicology, Vrije Universiteit Amsterdam, De Boelelaan 1105, 1081 HV, Amersterdam, Netherlands
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitätsstraße 10, 78464, Constance, Germany
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| |
Collapse
|
6
|
Martin A, Babbitt A, Pickens AG, Pickett BE, Hill JT, Suli A. Single-Cell RNA Sequencing Characterizes the Molecular Heterogeneity of the Larval Zebrafish Optic Tectum. Front Mol Neurosci 2022; 15:818007. [PMID: 35221915 PMCID: PMC8869500 DOI: 10.3389/fnmol.2022.818007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/11/2022] [Indexed: 01/04/2023] Open
Abstract
The optic tectum (OT) is a multilaminated midbrain structure that acts as the primary retinorecipient in the zebrafish brain. Homologous to the mammalian superior colliculus, the OT is responsible for the reception and integration of stimuli, followed by elicitation of salient behavioral responses. While the OT has been the focus of functional experiments for decades, less is known concerning specific cell types, microcircuitry, and their individual functions within the OT. Recent efforts have contributed substantially to the knowledge of tectal cell types; however, a comprehensive cell catalog is incomplete. Here we contribute to this growing effort by applying single-cell RNA Sequencing (scRNA-seq) to characterize the transcriptomic profiles of tectal cells labeled by the transgenic enhancer trap line y304Et(cfos:Gal4;UAS:Kaede). We sequenced 13,320 cells, a 4X cellular coverage, and identified 25 putative OT cell populations. Within those cells, we identified several mature and developing neuronal populations, as well as non-neuronal cell types including oligodendrocytes and microglia. Although most mature neurons demonstrate GABAergic activity, several glutamatergic populations are present, as well as one glycinergic population. We also conducted Gene Ontology analysis to identify enriched biological processes, and computed RNA velocity to infer current and future transcriptional cell states. Finally, we conducted in situ hybridization to validate our bioinformatic analyses and spatially map select clusters. In conclusion, the larval zebrafish OT is a complex structure containing at least 25 transcriptionally distinct cell populations. To our knowledge, this is the first time scRNA-seq has been applied to explore the OT alone and in depth.
Collapse
Affiliation(s)
- Annalie Martin
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
- *Correspondence: Annalie Martin,
| | - Anne Babbitt
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| | - Allison G. Pickens
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| | - Brett E. Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Jonathon T. Hill
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
| | - Arminda Suli
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, United States
- Arminda Suli,
| |
Collapse
|
7
|
Bozzo M, Costa S, Obino V, Bachetti T, Marcenaro E, Pestarino M, Schubert M, Candiani S. Functional Conservation and Genetic Divergence of Chordate Glycinergic Neurotransmission: Insights from Amphioxus Glycine Transporters. Cells 2021; 10:cells10123392. [PMID: 34943900 PMCID: PMC8699752 DOI: 10.3390/cells10123392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/24/2022] Open
Abstract
Glycine is an important neurotransmitter in vertebrates, performing both excitatory and inhibitory actions. Synaptic levels of glycine are tightly controlled by the action of two glycine transporters, GlyT1 and GlyT2, located on the surface of glial cells and neurons, respectively. Only limited information is available on glycinergic neurotransmission in invertebrates, and the evolution of glycinergic neurotransmission is poorly understood. Here, by combining phylogenetic and gene expression analyses, we characterized the glycine transporter complement of amphioxus, an important invertebrate model for studying the evolution of chordates. We show that amphioxus possess three glycine transporter genes. Two of these (GlyT2.1 and GlyT2.2) are closely related to GlyT2 of vertebrates, whereas the third (GlyT) is a member of an ancestral clade of deuterostome glycine transporters. GlyT2.2 expression is predominantly non-neural, whereas GlyT and GlyT2.1 are widely expressed in the amphioxus nervous system and are differentially expressed, respectively, in neurons and glia. Vertebrate glycinergic neurons express GlyT2 and glia GlyT1, suggesting that the evolution of the chordate glycinergic system was accompanied by a paralog-specific inversion of gene expression. Despite this genetic divergence between amphioxus and vertebrates, we found strong evidence for conservation in the role glycinergic neurotransmission plays during larval swimming, the implication being that the neural networks controlling the rhythmic movement of chordate bodies may be homologous.
Collapse
Affiliation(s)
- Matteo Bozzo
- Dipartimento di Scienze della Terra, dell’Ambiente e della Vita (DISTAV), Università degli Studi di Genova, 16132 Genoa, Italy; (S.C.); (T.B.); (M.P.)
- Dipartimento di Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy; (V.O.); (E.M.)
- Correspondence: (M.B.); (S.C.); Tel.: +39-010-335-8043 (M.B.); +39-010-335-8051 (S.C.)
| | - Simone Costa
- Dipartimento di Scienze della Terra, dell’Ambiente e della Vita (DISTAV), Università degli Studi di Genova, 16132 Genoa, Italy; (S.C.); (T.B.); (M.P.)
| | - Valentina Obino
- Dipartimento di Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy; (V.O.); (E.M.)
| | - Tiziana Bachetti
- Dipartimento di Scienze della Terra, dell’Ambiente e della Vita (DISTAV), Università degli Studi di Genova, 16132 Genoa, Italy; (S.C.); (T.B.); (M.P.)
| | - Emanuela Marcenaro
- Dipartimento di Medicina Sperimentale (DIMES), Università degli Studi di Genova, 16132 Genoa, Italy; (V.O.); (E.M.)
| | - Mario Pestarino
- Dipartimento di Scienze della Terra, dell’Ambiente e della Vita (DISTAV), Università degli Studi di Genova, 16132 Genoa, Italy; (S.C.); (T.B.); (M.P.)
| | - Michael Schubert
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), Institut de la Mer de Villefranche, Sorbonne Université, CNRS, 06230 Villefranche-sur-Mer, France;
| | - Simona Candiani
- Dipartimento di Scienze della Terra, dell’Ambiente e della Vita (DISTAV), Università degli Studi di Genova, 16132 Genoa, Italy; (S.C.); (T.B.); (M.P.)
- Correspondence: (M.B.); (S.C.); Tel.: +39-010-335-8043 (M.B.); +39-010-335-8051 (S.C.)
| |
Collapse
|
8
|
The presynaptic glycine transporter GlyT2 is regulated by the Hedgehog pathway in vitro and in vivo. Commun Biol 2021; 4:1197. [PMID: 34663888 PMCID: PMC8523746 DOI: 10.1038/s42003-021-02718-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/22/2021] [Indexed: 01/20/2023] Open
Abstract
The identity of a glycinergic synapse is maintained presynaptically by the activity of a surface glycine transporter, GlyT2, which recaptures glycine back to presynaptic terminals to preserve vesicular glycine content. GlyT2 loss-of-function mutations cause Hyperekplexia, a rare neurological disease in which loss of glycinergic neurotransmission causes generalized stiffness and strong motor alterations. However, the molecular underpinnings controlling GlyT2 activity remain poorly understood. In this work, we identify the Hedgehog pathway as a robust controller of GlyT2 expression and transport activity. Modulating the activation state of the Hedgehog pathway in vitro in rodent primary spinal cord neurons or in vivo in zebrafish embryos induced a selective control in GlyT2 expression, regulating GlyT2 transport activity. Our results indicate that activation of Hedgehog reduces GlyT2 expression by increasing its ubiquitination and degradation. This work describes a new molecular link between the Hedgehog signaling pathway and presynaptic glycine availability. By modulating the activation state of the Hedgehog pathway, de la Rocha-Muñoz et al demonstrate that Hedgehog signaling controls the expression and transport activity of the neuronal glycine transporter GlyT2. This work begins to reveal a potential link between the Hedgehog signaling pathway and presynaptic glycine availability.
Collapse
|
9
|
Venincasa MJ, Randlett O, Sumathipala SH, Bindernagel R, Stark MJ, Yan Q, Sloan SA, Buglo E, Meng QC, Engert F, Züchner S, Kelz MB, Syed S, Dallman JE. Elevated preoptic brain activity in zebrafish glial glycine transporter mutants is linked to lethargy-like behaviors and delayed emergence from anesthesia. Sci Rep 2021; 11:3148. [PMID: 33542258 PMCID: PMC7862283 DOI: 10.1038/s41598-021-82342-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 01/19/2021] [Indexed: 11/17/2022] Open
Abstract
Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.
Collapse
Affiliation(s)
- Michael J Venincasa
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, 69008, Lyon, France
| | - Sureni H Sumathipala
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Richard Bindernagel
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Matthew J Stark
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Qing Yan
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Steven A Sloan
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Elena Buglo
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, 33101, USA
- Dr. John T. MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, 33136, USA
| | - Qing Cheng Meng
- Departments of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, 33101, USA
- Dr. John T. MacDonald Foundation Department of Human Genetics, University of Miami, Miami, FL, 33136, USA
| | - Max B Kelz
- Departments of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sheyum Syed
- Department of Physics, University of Miami, Coral Gables, FL, 33146, USA
| | - Julia E Dallman
- Department of Biology, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA.
| |
Collapse
|
10
|
Bhat S, El-Kasaby A, Freissmuth M, Sucic S. Functional and Biochemical Consequences of Disease Variants in Neurotransmitter Transporters: A Special Emphasis on Folding and Trafficking Deficits. Pharmacol Ther 2020; 222:107785. [PMID: 33310157 PMCID: PMC7612411 DOI: 10.1016/j.pharmthera.2020.107785] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 01/30/2023]
Abstract
Neurotransmitters, such as γ-aminobutyric acid, glutamate, acetyl choline, glycine and the monoamines, facilitate the crosstalk within the central nervous system. The designated neurotransmitter transporters (NTTs) both release and take up neurotransmitters to and from the synaptic cleft. NTT dysfunction can lead to severe pathophysiological consequences, e.g. epilepsy, intellectual disability, or Parkinson’s disease. Genetic point mutations in NTTs have recently been associated with the onset of various neurological disorders. Some of these mutations trigger folding defects in the NTT proteins. Correct folding is a prerequisite for the export of NTTs from the endoplasmic reticulum (ER) and the subsequent trafficking to their pertinent site of action, typically at the plasma membrane. Recent studies have uncovered some of the key features in the molecular machinery responsible for transporter protein folding, e.g., the role of heat shock proteins in fine-tuning the ER quality control mechanisms in cells. The therapeutic significance of understanding these events is apparent from the rising number of reports, which directly link different pathological conditions to NTT misfolding. For instance, folding-deficient variants of the human transporters for dopamine or GABA lead to infantile parkinsonism/dystonia and epilepsy, respectively. From a therapeutic point of view, some folding-deficient NTTs are amenable to functional rescue by small molecules, known as chemical and pharmacological chaperones.
Collapse
Affiliation(s)
- Shreyas Bhat
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Ali El-Kasaby
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology and the Gaston H. Glock Research Laboratories for Exploratory Drug Development, Center of Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
11
|
Gomila AMJ, Rustler K, Maleeva G, Nin-Hill A, Wutz D, Bautista-Barrufet A, Rovira X, Bosch M, Mukhametova E, Petukhova E, Ponomareva D, Mukhamedyarov M, Peiretti F, Alfonso-Prieto M, Rovira C, König B, Bregestovski P, Gorostiza P. Photocontrol of Endogenous Glycine Receptors In Vivo. Cell Chem Biol 2020; 27:1425-1433.e7. [PMID: 32846115 DOI: 10.1016/j.chembiol.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/14/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022]
Abstract
Glycine receptors (GlyRs) are indispensable for maintaining excitatory/inhibitory balance in neuronal circuits that control reflexes and rhythmic motor behaviors. Here we have developed Glyght, a GlyR ligand controlled with light. It is selective over other Cys-loop receptors, is active in vivo, and displays an allosteric mechanism of action. The photomanipulation of glycinergic neurotransmission opens new avenues to understanding inhibitory circuits in intact animals and to developing drug-based phototherapies.
Collapse
Affiliation(s)
- Alexandre M J Gomila
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Karin Rustler
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany
| | - Galyna Maleeva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France
| | - Alba Nin-Hill
- University of Barcelona, Department of Inorganic and Organic Chemistry, Institute of Theoretical Chemistry (IQTCUB), Barcelona 08028, Spain
| | - Daniel Wutz
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany
| | - Antoni Bautista-Barrufet
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Xavier Rovira
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Miquel Bosch
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
| | - Elvira Mukhametova
- Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France; Kazan Federal University, Open Lab of Motor Neurorehabilitation, Kazan, Russia
| | - Elena Petukhova
- Institute of Neurosciences, Kazan State Medical University, Kazan, Russia
| | - Daria Ponomareva
- Institute of Neurosciences, Kazan State Medical University, Kazan, Russia
| | | | - Franck Peiretti
- Aix Marseille Université, INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Mercedes Alfonso-Prieto
- Institute for Advanced Simulation IAS-5 and Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, 52425 Jülich, Germany; Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Carme Rovira
- University of Barcelona, Department of Inorganic and Organic Chemistry, Institute of Theoretical Chemistry (IQTCUB), Barcelona 08028, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08003 Spain.
| | - Burkhard König
- University of Regensburg, Institute of Organic Chemistry, Regensburg 93053, Germany.
| | - Piotr Bregestovski
- Aix-Marseille Université, INSERM, INS, Institut de Neurosciences des Systèmes, Marseille 13005, France; Institute of Neurosciences, Kazan State Medical University, Kazan, Russia.
| | - Pau Gorostiza
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona 08003 Spain; CIBER-BBN, Madrid 28001 Spain.
| |
Collapse
|
12
|
Identification of the hypertension drug niflumic acid as a glycine receptor inhibitor. Sci Rep 2020; 10:13999. [PMID: 32814817 PMCID: PMC7438329 DOI: 10.1038/s41598-020-70983-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/29/2020] [Indexed: 11/08/2022] Open
Abstract
Glycine is one of the major neurotransmitters in the brainstem and the spinal cord. Glycine binds to and activates glycine receptors (GlyRs), increasing Cl- conductance at postsynaptic sites. This glycinergic synaptic transmission contributes to the generation of respiratory rhythm and motor patterns. Strychnine inhibits GlyR by binding to glycine-binding site, while picrotoxin blocks GlyR by binding to the channel pore. We have previously reported that bath application of strychnine to zebrafish embryos causes bilateral muscle contractions in response to tactile stimulation. To explore the drug-mediated inhibition of GlyRs, we screened a chemical library of ~ 1,000 approved drugs and pharmacologically active molecules by observing touch-evoked response of zebrafish embryos in the presence of drugs. We found that exposure of zebrafish embryos to nifedipine (an inhibitor of voltage-gated calcium channel) or niflumic acid (an inhibitor of cyclooxygenase 2) caused bilateral muscle contractions just like strychnine-treated embryos showed. We then assayed strychnine, picrotoxin, nifedipine, and niflumic acid for concentration-dependent inhibition of glycine-mediated currents of GlyRs in oocytes and calculated IC50s. The results indicate that all of them concentration-dependently inhibit GlyR in the order of strychnine > picrotoxin > nifedipine > niflumic acid.
Collapse
|
13
|
Behavioral and brain- transcriptomic synchronization between the two opponents of a fighting pair of the fish Betta splendens. PLoS Genet 2020; 16:e1008831. [PMID: 32555673 PMCID: PMC7299326 DOI: 10.1371/journal.pgen.1008831] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 05/05/2020] [Indexed: 01/13/2023] Open
Abstract
Conspecific male animals fight for resources such as food and mating opportunities but typically stop fighting after assessing their relative fighting abilities to avoid serious injuries. Physiologically, how the fighting behavior is controlled remains unknown. Using the fighting fish Betta splendens, we studied behavioral and brain-transcriptomic changes during the fight between the two opponents. At the behavioral level, surface-breathing, and biting/striking occurred only during intervals between mouth-locking. Eventually, the behaviors of the two opponents became synchronized, with each pair showing a unique behavioral pattern. At the physiological level, we examined the expression patterns of 23,306 brain transcripts using RNA-sequencing data from brains of fighting pairs after a 20-min (D20) and a 60-min (D60) fight. The two opponents in each D60 fighting pair showed a strong gene expression correlation, whereas those in D20 fighting pairs showed a weak correlation. Moreover, each fighting pair in the D60 group showed pair-specific gene expression patterns in a grade of membership analysis (GoM) and were grouped as a pair in the heatmap clustering. The observed pair-specific individualization in brain-transcriptomic synchronization (PIBS) suggested that this synchronization provides a physiological basis for the behavioral synchronization. An analysis using the synchronized genes in fighting pairs of the D60 group found genes enriched for ion transport, synaptic function, and learning and memory. Brain-transcriptomic synchronization could be a general phenomenon and may provide a new cornerstone with which to investigate coordinating and sustaining social interactions between two interacting partners of vertebrates. Agonistic encounters induce changes in the brain and behavior, but their underlying molecular mechanisms remain poorly understood. The fighting fish Betta splendens are small freshwater fish that are well known for their aggressiveness and are widely used to study aggression. Here, by measuring aggressive behavior displays (bite/strike/surface-breathing) between two opponents during fighting, we demonstrate that the two opponents in each fighting pair showed similar fighting configurations by influencing each other. In addition, we compared brain gene expression between opponents and showed synchronization of gene expression within a fighting pair, leading to pair-specific synchronization in genes associated with ion transport, synapse function, and learning and memory. This study presents the possibility that similar behaviors in pairs of animals under similar conditions may trigger synchronizing waves of transcription between the individuals, providing a hint to support the idea that fighting behaviors contain cooperative aspects at the molecular level.
Collapse
|
14
|
Ma M, Kler S, Pan YA. Structural Neural Connectivity Analysis in Zebrafish With Restricted Anterograde Transneuronal Viral Labeling and Quantitative Brain Mapping. Front Neural Circuits 2020; 13:85. [PMID: 32038180 PMCID: PMC6989443 DOI: 10.3389/fncir.2019.00085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022] Open
Abstract
The unique combination of small size, translucency, and powerful genetic tools makes larval zebrafish a uniquely useful vertebrate system to investigate normal and pathological brain structure and function. While functional connectivity can now be assessed by optical imaging (via fluorescent calcium or voltage reporters) at the whole-brain scale, it remains challenging to systematically determine structural connections and identify connectivity changes during development or disease. To address this, we developed Tracer with Restricted Anterograde Spread (TRAS), a novel vesicular stomatitis virus (VSV)-based neural circuit labeling approach. TRAS makes use of replication-incompetent VSV (VSVΔG) and a helper virus (lentivirus) to enable anterograde transneuronal spread between efferent axons and their direct postsynaptic targets but restricts further spread to downstream areas. We integrated TRAS with the Z-Brain zebrafish 3D atlas for quantitative connectivity analysis and identified targets of the retinal and habenular efferent projections, in patterns consistent with previous reports. We compared retinofugal connectivity patterns between wild-type and down syndrome cell adhesion molecule-like 1 (dscaml1) mutant zebrafish and revealed differences in topographical distribution. These results demonstrate the utility of TRAS for quantitative structural connectivity analysis that would be valuable for detecting novel efferent targets and mapping connectivity changes underlying neurological or behavioral deficits.
Collapse
Affiliation(s)
- Manxiu Ma
- Center for Neurobiology Research, Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, United States.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Stanislav Kler
- Center for Neurobiology Research, Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, United States.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Y Albert Pan
- Center for Neurobiology Research, Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, United States.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States.,Department of Psychiatry and Behavioral Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
15
|
Wan Y, Wei Z, Looger LL, Koyama M, Druckmann S, Keller PJ. Single-Cell Reconstruction of Emerging Population Activity in an Entire Developing Circuit. Cell 2019; 179:355-372.e23. [PMID: 31564455 PMCID: PMC7055533 DOI: 10.1016/j.cell.2019.08.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 07/03/2019] [Accepted: 08/21/2019] [Indexed: 01/04/2023]
Abstract
Animal survival requires a functioning nervous system to develop during embryogenesis. Newborn neurons must assemble into circuits producing activity patterns capable of instructing behaviors. Elucidating how this process is coordinated requires new methods that follow maturation and activity of all cells across a developing circuit. We present an imaging method for comprehensively tracking neuron lineages, movements, molecular identities, and activity in the entire developing zebrafish spinal cord, from neurogenesis until the emergence of patterned activity instructing the earliest spontaneous motor behavior. We found that motoneurons are active first and form local patterned ensembles with neighboring neurons. These ensembles merge, synchronize globally after reaching a threshold size, and finally recruit commissural interneurons to orchestrate the left-right alternating patterns important for locomotion in vertebrates. Individual neurons undergo functional maturation stereotypically based on their birth time and anatomical origin. Our study provides a general strategy for reconstructing how functioning circuits emerge during embryogenesis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Yinan Wan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Ziqiang Wei
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Minoru Koyama
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Shaul Druckmann
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA; Department of Neurobiology, Stanford University, Stanford, CA, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Philipp J Keller
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| |
Collapse
|
16
|
Samarut E, Chalopin D, Riché R, Allard M, Liao M, Drapeau P. Individual knock out of glycine receptor alpha subunits identifies a specific requirement of glra1 for motor function in zebrafish. PLoS One 2019; 14:e0216159. [PMID: 31048868 PMCID: PMC6497278 DOI: 10.1371/journal.pone.0216159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/15/2019] [Indexed: 11/18/2022] Open
Abstract
Glycine receptors (GlyRs) are ligand-gated chloride channels mediating inhibitory neurotransmission in the brain stem and spinal cord. They function as pentamers composed of alpha and beta subunits for which 5 genes have been identified in human (GLRA1, GLRA2, GLRA3, GLRA4, GLRB). Several in vitro studies showed that the pentameric subtype composition as well as its stoichiometry influence the distribution and the molecular function of the receptor. Moreover, mutations in some of these genes are involved in different human conditions ranging from tinnitus to epilepsy and hyperekplexia, suggesting distinct functions of the different subunits. Although the beta subunit is essential for synaptic clustering of the receptor, the specific role of each alpha subtype is still puzzling in vivo. The zebrafish genome encodes for five glycine receptor alpha subunits (glra1, glra2, glra3, glra4a, glra4b) thus offering a model of choice to investigate the respective role of each subtype on general motor behaviour. After establishing a phylogeny of GlyR subunit evolution between human and zebrafish, we checked the temporal expression pattern of these transcripts during embryo development. Interestingly, we found that glra1 is the only maternally transmitted alpha subunit. We also showed that the expression of the different GlyR subunits starts at different time points during development. Lastly, in order to decipher the role of each alpha subunit on the general motor behaviour of the fish, we knocked out individually each alpha subunit by CRISPR/Cas9-targeted mutagenesis. Surprisingly, we found that knocking out any of the alpha2, 3, a4a or a4b subunit did not lead to any obvious developmental or motor phenotype. However, glra1-/- (hitch) embryos depicted a strong motor dysfunction from 3 days, making them incapable to swim and thus leading to their premature death. Our results infer a strong functional redundancy between alpha subunits and confirm the central role played by glra1 for proper inhibitory neurotransmission controlling locomotion. The genetic tools we developed here will be of general interest for further studies aiming at dissecting the role of GlyRs in glycinergic transmission in vivo and the hitch mutant (hic) is of specific relevance as a new model of hyperekplexia.
Collapse
Affiliation(s)
- Eric Samarut
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- DanioDesign Inc., Montréal, QC, Canada
- Modelis Inc., Montréal, QC, Canada
- * E-mail:
| | - Domitille Chalopin
- UnivLyon, ENS de Lyon, Université de Lyon, CNRS UMR5239, INSERM U1210, Lyon, France
| | - Raphaëlle Riché
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Marc Allard
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Meijiang Liao
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Pierre Drapeau
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- DanioDesign Inc., Montréal, QC, Canada
| |
Collapse
|
17
|
Riché R, Liao M, Pena IA, Leung KY, Lepage N, Greene NDE, Sarafoglou K, Schimmenti LA, Drapeau P, Samarut É. Glycine decarboxylase deficiency-induced motor dysfunction in zebrafish is rescued by counterbalancing glycine synaptic level. JCI Insight 2018; 3:124642. [PMID: 30385710 DOI: 10.1172/jci.insight.124642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/19/2018] [Indexed: 11/17/2022] Open
Abstract
Glycine encephalopathy (GE), or nonketotic hyperglycinemia (NKH), is a rare recessive genetic disease caused by defective glycine cleavage and characterized by increased accumulation of glycine in all tissues. Here, based on new case reports of GLDC loss-of-function mutations in GE patients, we aimed to generate a zebrafish model of severe GE in order to unravel the molecular mechanism of the disease. Using CRISPR/Cas9, we knocked out the gldc gene and showed that gldc-/- fish recapitulate GE on a molecular level and present a motor phenotype reminiscent of severe GE symptoms. The molecular characterization of gldc-/- mutants showed a broad metabolic disturbance affecting amino acids and neurotransmitters other than glycine, with lactic acidosis at stages preceding death. Although a transient imbalance was found in cell proliferation in the brain of gldc-/- zebrafish, the main brain networks were not affected, thus suggesting that GE pathogenicity is mainly due to metabolic defects. We confirmed that the gldc-/- hypotonic phenotype is due to NMDA and glycine receptor overactivation, and demonstrated that gldc-/- larvae depict exacerbated hyperglycinemia at these synapses. Remarkably, we were able to rescue the motor dysfunction of gldc-/- larvae by counterbalancing pharmacologically or genetically the level of glycine at the synapse.
Collapse
Affiliation(s)
- Raphaëlle Riché
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| | - Meijiang Liao
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada
| | - Izabella A Pena
- Children's Hospital of Eastern Ontario Research Institute and Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario, Canada
| | - Kit-Yi Leung
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nathalie Lepage
- Children's Hospital of Eastern Ontario Research Institute and Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario, Canada
| | - Nicolas DE Greene
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Kyriakie Sarafoglou
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lisa A Schimmenti
- Department of Otorhinolaryngology.,Department of Pediatrics, and.,Department of Clinical Genomics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Pierre Drapeau
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada.,DanioDesign Inc., Montréal, Quebec, Canada
| | - Éric Samarut
- Research Center of the University of Montreal Hospital Center (CRCHUM), Department of Neurosciences, Université de Montréal, Montreal, Quebec, Canada.,DanioDesign Inc., Montréal, Quebec, Canada
| |
Collapse
|
18
|
Severi KE, Böhm UL, Wyart C. Investigation of hindbrain activity during active locomotion reveals inhibitory neurons involved in sensorimotor processing. Sci Rep 2018; 8:13615. [PMID: 30206288 PMCID: PMC6134141 DOI: 10.1038/s41598-018-31968-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/30/2018] [Indexed: 11/14/2022] Open
Abstract
Locomotion in vertebrates relies on motor circuits in the spinal cord receiving inputs from the hindbrain to execute motor commands while dynamically integrating proprioceptive sensory feedback. The spatial organization of the neuronal networks driving locomotion in the hindbrain and role of inhibition has not been extensively investigated. Here, we mapped neuronal activity with single-cell resolution in the hindbrain of restrained transgenic Tg(HuC:GCaMP5G) zebrafish larvae swimming in response to whole-field visual motion. We combined large-scale population calcium imaging in the hindbrain with simultaneous high-speed recording of the moving tail in animals where specific markers label glycinergic inhibitory neurons. We identified cells whose activity preferentially correlates with the visual stimulus or motor activity and used brain registration to compare data across individual larvae. We then morphed calcium imaging data onto the zebrafish brain atlas to compare with known transgenic markers. We report cells localized in the cerebellum whose activity is shut off by the onset of the visual stimulus, suggesting these cells may be constitutively active and silenced during sensorimotor processing. Finally, we discover that the activity of a medial stripe of glycinergic neurons in the domain of expression of the transcription factor engrailed1b is highly correlated with the onset of locomotion. Our efforts provide a high-resolution, open-access dataset for the community by comparing our functional map of the hindbrain to existing open-access atlases and enabling further investigation of this population's role in locomotion.
Collapse
Affiliation(s)
- Kristen E Severi
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm, CNRS, AP-HP, F-75013, Paris, France
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Urs L Böhm
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm, CNRS, AP-HP, F-75013, Paris, France
- Dept. of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Claire Wyart
- Institut du Cerveau et de la Moelle épinière, ICM, Sorbonne Université, Inserm, CNRS, AP-HP, F-75013, Paris, France.
| |
Collapse
|
19
|
Bedell V, Buglo E, Marcato D, Pylatiuk C, Mikut R, Stegmaier J, Scudder W, Wray M, Züchner S, Strähle U, Peravali R, Dallman JE. Zebrafish: A Pharmacogenetic Model for Anesthesia. Methods Enzymol 2018; 602:189-209. [PMID: 29588029 PMCID: PMC10559369 DOI: 10.1016/bs.mie.2018.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
General anesthetics are small molecules that interact with and effect the function of many different proteins to promote loss of consciousness, amnesia, and sometimes, analgesia. Owing to the complexity of this state transition and the transient nature of these drug/protein interactions, anesthetics can be difficult to study. The zebrafish is an emerging model for the discovery of both new genes required for the response to and side effects of anesthesia. Here we discuss the tools available to manipulate the zebrafish genome, including both genetic screens and genome engineering approaches. Additionally, there are various robust behavior assays available to study anesthetic and other drug responses. These assays are available for single-gene study or high throughput for genetic or drug discovery. Finally, we present a case study of using propofol as an anesthetic in the zebrafish. These techniques and protocols make the zebrafish a powerful model to study anesthetic mechanisms and drug discovery.
Collapse
Affiliation(s)
- Victoria Bedell
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States.
| | - Elena Buglo
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, United States; Dr. John T. MacDonald Foundation, University of Miami, Miami, FL, United States; University of Miami, Coral Gables, FL, United States
| | - Daniel Marcato
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Christian Pylatiuk
- Institute of Applied Computer Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute of Applied Computer Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Johannes Stegmaier
- Institute of Applied Computer Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Will Scudder
- University of Miami, Coral Gables, FL, United States
| | - Maxwell Wray
- University of Miami, Coral Gables, FL, United States
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, United States; Dr. John T. MacDonald Foundation, University of Miami, Miami, FL, United States
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ravindra Peravali
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | | |
Collapse
|
20
|
Freifeld L, Odstrcil I, Förster D, Ramirez A, Gagnon JA, Randlett O, Costa EK, Asano S, Celiker OT, Gao R, Martin-Alarcon DA, Reginato P, Dick C, Chen L, Schoppik D, Engert F, Baier H, Boyden ES. Expansion microscopy of zebrafish for neuroscience and developmental biology studies. Proc Natl Acad Sci U S A 2017; 114:E10799-E10808. [PMID: 29162696 PMCID: PMC5740639 DOI: 10.1073/pnas.1706281114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Expansion microscopy (ExM) allows scalable imaging of preserved 3D biological specimens with nanoscale resolution on fast diffraction-limited microscopes. Here, we explore the utility of ExM in the larval and embryonic zebrafish, an important model organism for the study of neuroscience and development. Regarding neuroscience, we found that ExM enabled the tracing of fine processes of radial glia, which are not resolvable with diffraction-limited microscopy. ExM further resolved putative synaptic connections, as well as molecular differences between densely packed synapses. Finally, ExM could resolve subsynaptic protein organization, such as ring-like structures composed of glycine receptors. Regarding development, we used ExM to characterize the shapes of nuclear invaginations and channels, and to visualize cytoskeletal proteins nearby. We detected nuclear invagination channels at late prophase and telophase, potentially suggesting roles for such channels in cell division. Thus, ExM of the larval and embryonic zebrafish may enable systematic studies of how molecular components are configured in multiple contexts of interest to neuroscience and developmental biology.
Collapse
Affiliation(s)
- Limor Freifeld
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Iris Odstrcil
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Dominique Förster
- Department Genes-Circuits-Behavior, Max Planck Institute of Neurobiology, Martinsried 82152, Germany
| | - Alyson Ramirez
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - James A Gagnon
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Owen Randlett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Emma K Costa
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139
| | - Shoh Asano
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Orhan T Celiker
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139
| | - Ruixuan Gao
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139
| | | | - Paul Reginato
- Department of Biological Engineering, MIT, Cambridge, MA 02139
- Department of Genetics, Harvard Medical School, Cambridge, MA 02138
| | - Cortni Dick
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
| | - Linlin Chen
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139
- Neuroscience Program, Wellesley College, Wellesley, MA 02481
| | - David Schoppik
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016
- Neuroscience Institute, New York University School of Medicine, New York NY 10016
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Herwig Baier
- Department Genes-Circuits-Behavior, Max Planck Institute of Neurobiology, Martinsried 82152, Germany
| | - Edward S Boyden
- Media Lab, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139;
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139
- McGovern Institute for Brain Research, MIT, Cambridge, MA 02139
- Center for Neurobiological Engineering, MIT, Cambridge, MA 02139
| |
Collapse
|
21
|
Chow DM, Zuchowski KA, Fetcho JR. In Vivo Measurement of Glycine Receptor Turnover and Synaptic Size Reveals Differences between Functional Classes of Motoneurons in Zebrafish. Curr Biol 2017; 27:1173-1183. [PMID: 28416115 DOI: 10.1016/j.cub.2017.03.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/12/2017] [Accepted: 03/15/2017] [Indexed: 01/18/2023]
Abstract
The interplay between binding and unbinding of synaptic receptor proteins at synapses plays an important role in determining receptor concentration and synaptic strength, with known links between changes in binding kinetics and synaptic plasticity. The regulation of such kinetics may subserve the specific functional requirements of neurons in intact circuits. However, the majority of studies of synaptic turnover kinetics have been performed in cultured neurons outside the context of normal circuits, and synaptic receptor turnover has not been measured at individual synaptic sites in vivo. We quantified the distribution of glycinergic receptor dynamics using fluorescence recovery after photoconversion of synapses in intact zebrafish and correlated recovery kinetics to synaptic volume in two functionally distinct classes of cells: primary and secondary motoneurons. The rate of fluorescence recovery after photoconversion decreased with synaptic volume in both types of motoneurons, with larger synapses having slower recovery. Primary motoneurons had both larger synapses and associated slower recovery times than secondary motoneurons. Our results suggest that synaptic kinetics are regulated in concert with synaptic sizes and reflect the functional role played by neurons within their circuit.
Collapse
Affiliation(s)
- Dawnis M Chow
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14850, USA.
| | - Kathryn A Zuchowski
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14850, USA
| | - Joseph R Fetcho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
22
|
Kurolap A, Armbruster A, Hershkovitz T, Hauf K, Mory A, Paperna T, Hannappel E, Tal G, Nijem Y, Sella E, Mahajnah M, Ilivitzki A, Hershkovitz D, Ekhilevitch N, Mandel H, Eulenburg V, Baris HN. Loss of Glycine Transporter 1 Causes a Subtype of Glycine Encephalopathy with Arthrogryposis and Mildly Elevated Cerebrospinal Fluid Glycine. Am J Hum Genet 2016; 99:1172-1180. [PMID: 27773429 DOI: 10.1016/j.ajhg.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/07/2016] [Indexed: 01/02/2023] Open
Abstract
Glycine is a major neurotransmitter that activates inhibitory glycine receptors and is a co-agonist for excitatory glutamatergic N-methyl-D-aspartate (NMDA) receptors. Two transporters, GLYT1 and GLYT2, regulate extracellular glycine concentrations within the CNS. Dysregulation of the extracellular glycine has been associated with hyperekplexia and nonketotic hyperglycinemia. Here, we report four individuals from two families who presented at birth with facial dysmorphism, encephalopathy, arthrogryposis, hypotonia progressing to hypertonicity with startle-like clonus, and respiratory failure. Only one individual survived the respiratory failure and was weaned off ventilation but has significant global developmental delay. Mildly elevated cerebrospinal fluid (CSF) glycine and normal serum glycine were observed in two individuals. In both families, we identified truncating mutations in SLC6A9, encoding GLYT1. We demonstrate that pharmacologic or genetic abolishment of GlyT1 activity in mice leads to mildly elevated glycine in the CSF but not in blood. Additionally, previously reported slc6a9-null mice and zebrafish mutants also display phenotypes consistent with the affected individuals we examined. Our data suggest that truncating SLC6A9 mutations lead to a distinct human neurological syndrome hallmarked by mildly elevated CSF glycine and normal serum glycine.
Collapse
|
23
|
Ogino K, Hirata H. Defects of the Glycinergic Synapse in Zebrafish. Front Mol Neurosci 2016; 9:50. [PMID: 27445686 PMCID: PMC4925712 DOI: 10.3389/fnmol.2016.00050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/13/2016] [Indexed: 12/26/2022] Open
Abstract
Glycine mediates fast inhibitory synaptic transmission. Physiological importance of the glycinergic synapse is well established in the brainstem and the spinal cord. In humans, the loss of glycinergic function in the spinal cord and brainstem leads to hyperekplexia, which is characterized by an excess startle reflex to sudden acoustic or tactile stimulation. In addition, glycinergic synapses in this region are also involved in the regulation of respiration and locomotion, and in the nociceptive processing. The importance of the glycinergic synapse is conserved across vertebrate species. A teleost fish, the zebrafish, offers several advantages as a vertebrate model for research of glycinergic synapse. Mutagenesis screens in zebrafish have isolated two motor defective mutants that have pathogenic mutations in glycinergic synaptic transmission: bandoneon (beo) and shocked (sho). Beo mutants have a loss-of-function mutation of glycine receptor (GlyR) β-subunit b, alternatively, sho mutant is a glycinergic transporter 1 (GlyT1) defective mutant. These mutants are useful animal models for understanding of glycinergic synaptic transmission and for identification of novel therapeutic agents for human diseases arising from defect in glycinergic transmission, such as hyperekplexia or glycine encephalopathy. Recent advances in techniques for genome editing and for imaging and manipulating of a molecule or a physiological process make zebrafish more attractive model. In this review, we describe the glycinergic defective zebrafish mutants and the technical advances in both forward and reverse genetic approaches as well as in vivo visualization and manipulation approaches for the study of the glycinergic synapse in zebrafish.
Collapse
Affiliation(s)
- Kazutoyo Ogino
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University Sagamihara, Japan
| | - Hiromi Hirata
- Department of Chemistry and Biological Science, College of Science and Engineering, Aoyama Gakuin University Sagamihara, Japan
| |
Collapse
|
24
|
An E3-ligase-based method for ablating inhibitory synapses. Nat Methods 2016; 13:673-8. [PMID: 27271196 PMCID: PMC5312699 DOI: 10.1038/nmeth.3894] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/09/2016] [Indexed: 01/08/2023]
Abstract
Although neuronal activity can be modulated using a variety of techniques, there are currently few methods for controlling neuronal connectivity. We introduce a tool (GFE3) that mediates the fast, specific and reversible elimination of inhibitory synaptic inputs onto genetically determined neurons. GFE3 is a fusion between an E3 ligase, which mediates the ubiquitination and rapid degradation of proteins, and a recombinant, antibody-like protein (FingR) that binds to gephyrin. Expression of GFE3 leads to a strong and specific reduction of gephyrin in culture or in vivo and to a substantial decrease in phasic inhibition onto cells that express GFE3. By temporarily expressing GFE3 we showed that inhibitory synapses regrow following ablation. Thus, we have created a simple, reversible method for modulating inhibitory synaptic input onto genetically determined cells.
Collapse
|
25
|
A hybrid electrical/chemical circuit in the spinal cord generates a transient embryonic motor behavior. J Neurosci 2014; 34:9644-55. [PMID: 25031404 DOI: 10.1523/jneurosci.1225-14.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Spontaneous network activity is a highly stereotyped early feature of developing circuits throughout the nervous system, including in the spinal cord. Spinal locomotor circuits produce a series of behaviors during development before locomotion that reflect the continual integration of spinal neurons into a functional network, but how the circuitry is reconfigured is not understood. The first behavior of the zebrafish embryo (spontaneous coiling) is mediated by an electrical circuit that subsequently generates mature locomotion (swimming) as chemical neurotransmission develops. We describe here a new spontaneous behavior, double coiling, that consists of two alternating contractions of the tail in rapid succession. Double coiling was glutamate-dependent and required descending hindbrain excitation, similar to but preceding swimming, making it a discrete intermediary developmental behavior. At the cellular level, motoneurons had a distinctive glutamate-dependent activity pattern that correlated with double coiling. Two glutamatergic interneurons, CoPAs and CiDs, had different activity profiles during this novel behavior. CoPA neurons failed to show changes in activity patterns during the period in which double coiling appears, whereas CiD neurons developed a glutamate-dependent activity pattern that correlated with double coiling and they innervated motoneurons at that time. Additionally, double coils were modified after pharmacological reduction of glycinergic neurotransmission such that embryos produced three or more rapidly alternating coils. We propose that double coiling behavior represents an important transition of the motor network from an electrically coupled spinal cord circuit that produces simple periodic coils to a spinal network driven by descending chemical neurotransmission, which generates more complex behaviors.
Collapse
|
26
|
Moly PK, Ikenaga T, Kamihagi C, Islam AT, Hatta K. Identification of initially appearing glycine-immunoreactive neurons in the embryonic zebrafish brain. Dev Neurobiol 2014; 74:616-32. [DOI: 10.1002/dneu.22158] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 11/26/2013] [Accepted: 11/26/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Pricila Khan Moly
- Graduate School of Life Science; University of Hyogo; 3-2-1 Kouto, Kamigori, Ako-gun Hyogo 678-1297 Japan
| | - Takanori Ikenaga
- Graduate School of Life Science; University of Hyogo; 3-2-1 Kouto, Kamigori, Ako-gun Hyogo 678-1297 Japan
| | - Chihiro Kamihagi
- Graduate School of Life Science; University of Hyogo; 3-2-1 Kouto, Kamigori, Ako-gun Hyogo 678-1297 Japan
| | - A.F.M. Tariqul Islam
- Graduate School of Life Science; University of Hyogo; 3-2-1 Kouto, Kamigori, Ako-gun Hyogo 678-1297 Japan
| | - Kohei Hatta
- Graduate School of Life Science; University of Hyogo; 3-2-1 Kouto, Kamigori, Ako-gun Hyogo 678-1297 Japan
| |
Collapse
|
27
|
Defective escape behavior in DEAH-box RNA helicase mutants improved by restoring glycine receptor expression. J Neurosci 2013; 33:14638-44. [PMID: 24027265 DOI: 10.1523/jneurosci.1157-13.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
RNA helicases regulate RNA metabolism, but their substrate specificity and in vivo function remain largely unknown. We isolated spontaneous mutant zebrafish that exhibit an abnormal dorsal bend at the beginning of tactile-evoked escape swimming. Similar behavioral defects were observed in zebrafish embryos treated with strychnine, which blocks glycine receptors (GlyRs), suggesting that the abnormal motor response in mutants may be attributable to a deficit in glycinergic synaptic transmission. We identified a missense mutation in the gene encoding RNA helicase Dhx37. In Dhx37 mutants, ribosomal RNA levels were unchanged, whereas GlyR α1, α3, and α4a subunit mRNA levels were decreased due to a splicing defect. We found that Dhx37 can interact with GlyR α1, α3, and α4a transcripts but not with the GlyR α2 subunit mRNA. Overexpression of GlyR α1, α3, or α4a subunits in Dhx37-deficient embryos restored normal behavior. Conversely, antisense-mediated knockdown of multiple GlyR α subunits in wild-type embryos was required to recapitulate the Dhx37 mutant phenotype. These results indicate that Dhx37 is specifically required for the biogenesis of a subset of GlyR α subunit mRNAs, thereby regulating glycinergic synaptic transmission and associated motor behaviors. To our knowledge, this is the first identification of pathologically relevant substrates for an RNA helicase.
Collapse
|
28
|
de Juan-Sanz J, Núñez E, Villarejo-López L, Pérez-Hernández D, Rodriguez-Fraticelli AE, López-Corcuera B, Vázquez J, Aragón C. Na+/K+-ATPase is a new interacting partner for the neuronal glycine transporter GlyT2 that downregulates its expression in vitro and in vivo. J Neurosci 2013; 33:14269-81. [PMID: 23986260 PMCID: PMC6618510 DOI: 10.1523/jneurosci.1532-13.2013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 01/16/2023] Open
Abstract
The neuronal glycine transporter GlyT2 plays a fundamental role in the glycinergic neurotransmission by recycling the neurotransmitter to the presynaptic terminal. GlyT2 is the main supplier of glycine for vesicle refilling, a process that is absolutely necessary to preserve quantal glycine content in synaptic vesicles. Alterations in GlyT2 activity modify glycinergic neurotransmission and may underlie several neuromuscular disorders, such as hyperekplexia, myoclonus, dystonia, and epilepsy. Indeed, mutations in the gene encoding GlyT2 are the main presynaptic cause of hyperekplexia in humans and produce congenital muscular dystonia type 2 (CMD2) in Belgian Blue cattle. GlyT2 function is strictly coupled to the sodium electrochemical gradient actively generated by the Na+/K+-ATPase (NKA). GlyT2 cotransports 3Na+/Cl-/glycine generating large rises of Na+ inside the presynaptic terminal that must be efficiently reduced by the NKA to preserve Na+ homeostasis. In this work, we have used high-throughput mass spectrometry to identify proteins interacting with GlyT2 in the CNS. NKA was detected as a putative candidate and through reciprocal coimmunoprecipitations and immunocytochemistry analyses the association between GlyT2 and NKA was confirmed. NKA mainly interacts with the raft-associated active pool of GlyT2, and low and high levels of the specific NKA ligand ouabain modulate the endocytosis and total expression of GlyT2 in neurons. The ouabain-mediated downregulation of GlyT2 also occurs in vivo in two different systems: zebrafish embryos and adult rats, indicating that this NKA-mediated regulatory mechanism is evolutionarily conserved and may play a relevant role in the physiological control of inhibitory glycinergic neurotransmission.
Collapse
Affiliation(s)
- Jaime de Juan-Sanz
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Enrique Núñez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Lucía Villarejo-López
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | | | - Alejo E. Rodriguez-Fraticelli
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz López-Corcuera
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain, and
| | - Carmen Aragón
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 46010 Valencia, Spain
- IdiPAZ-Hospital Universitario La Paz, 28046 Madrid, Spain
| |
Collapse
|
29
|
Anadón R, Rodríguez-Moldes I, Adrio F. Glycine-immunoreactive neurons in the brain of a shark (Scyliorhinus caniculaL.). J Comp Neurol 2013; 521:3057-82. [DOI: 10.1002/cne.23332] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/07/2013] [Accepted: 03/13/2013] [Indexed: 01/12/2023]
Affiliation(s)
- Ramón Anadón
- Department of Cell Biology and Ecology; University of Santiago de Compostela; 15782 Santiago de; Compostela; Spain
| | - Isabel Rodríguez-Moldes
- Department of Cell Biology and Ecology; University of Santiago de Compostela; 15782 Santiago de; Compostela; Spain
| | - Fátima Adrio
- Department of Cell Biology and Ecology; University of Santiago de Compostela; 15782 Santiago de; Compostela; Spain
| |
Collapse
|
30
|
Kondrychyn I, Teh C, Sin M, Korzh V. Stretching morphogenesis of the roof plate and formation of the central canal. PLoS One 2013; 8:e56219. [PMID: 23409159 PMCID: PMC3567028 DOI: 10.1371/journal.pone.0056219] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 01/07/2013] [Indexed: 12/20/2022] Open
Abstract
Background Neurulation is driven by apical constriction of actomyosin cytoskeleton resulting in conversion of the primitive lumen into the central canal in a mechanism driven by F-actin constriction, cell overcrowding and buildup of axonal tracts. The roof plate of the neural tube acts as the dorsal morphogenetic center and boundary preventing midline crossing by neural cells and axons. Methodology/Principal Findings The roof plate zebrafish transgenics expressing cytosolic GFP were used to study and describe development of this structure in vivo for a first time ever. The conversion of the primitive lumen into the central canal causes significant morphogenetic changes of neuroepithelial cells in the dorsal neural tube. We demonstrated that the roof plate cells stretch along the D–V axis in parallel with conversion of the primitive lumen into central canal and its ventral displacement. Importantly, the stretching of the roof plate is well-coordinated along the whole spinal cord and the roof plate cells extend 3× in length to cover 2/3 of the neural tube diameter. This process involves the visco-elastic extension of the roof place cytoskeleton and depends on activity of Zic6 and the Rho-associated kinase (Rock). In contrast, stretching of the floor plate is much less extensive. Conclusions/Significance The extension of the roof plate requires its attachment to the apical complex of proteins at the surface of the central canal, which depends on activity of Zic6 and Rock. The D–V extension of the roof plate may change a range and distribution of morphogens it produces. The resistance of the roof plate cytoskeleton attenuates ventral displacement of the central canal in illustration of the novel mechanical role of the roof plate during development of the body axis.
Collapse
Affiliation(s)
- Igor Kondrychyn
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
| | - Cathleen Teh
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
| | - Melvin Sin
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
| | - Vladimir Korzh
- Institute of Molecular and Cell Biology, A-STAR, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
31
|
Horstick EJ, Linsley JW, Dowling JJ, Hauser MA, McDonald KK, Ashley-Koch A, Saint-Amant L, Satish A, Cui WW, Zhou W, Sprague SM, Stamm DS, Powell CM, Speer MC, Franzini-Armstrong C, Hirata H, Kuwada JY. Stac3 is a component of the excitation-contraction coupling machinery and mutated in Native American myopathy. Nat Commun 2013; 4:1952. [PMID: 23736855 PMCID: PMC4056023 DOI: 10.1038/ncomms2952] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 04/30/2013] [Indexed: 11/09/2022] Open
Abstract
Excitation-contraction coupling, the process that regulates contractions by skeletal muscles, transduces changes in membrane voltage by activating release of Ca(2+) from internal stores to initiate muscle contraction. Defects in excitation-contraction coupling are associated with muscle diseases. Here we identify Stac3 as a novel component of the excitation-contraction coupling machinery. Using a zebrafish genetic screen, we generate a locomotor mutation that is mapped to stac3. We provide electrophysiological, Ca(2+) imaging, immunocytochemical and biochemical evidence that Stac3 participates in excitation-contraction coupling in muscles. Furthermore, we reveal that a mutation in human STAC3 is the genetic basis of the debilitating Native American myopathy (NAM). Analysis of NAM stac3 in zebrafish shows that the NAM mutation decreases excitation-contraction coupling. These findings enhance our understanding of both excitation-contraction coupling and the pathology of myopathies.
Collapse
Affiliation(s)
- Eric J. Horstick
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeremy W. Linsley
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J. Dowling
- Department of Pediatrics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Michael A. Hauser
- Departments of Medicine and Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristin K. McDonald
- Departments of Medicine and Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Allison Ashley-Koch
- Departments of Medicine and Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | - Louis Saint-Amant
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Departement de Pathologie et Biologie Cellulaire, Universite de Montreal, Montreal, Canada H3T 1J4
| | - Akhila Satish
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wilson W. Cui
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weibin Zhou
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Life Science Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shawn M. Sprague
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Demetra S. Stamm
- Department of Internal Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Cynthia M. Powell
- Departments of Pediatrics and Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marcy C. Speer
- Center for Human Genetics, Duke University, Durham, NC 27710, USA
| | - Clara Franzini-Armstrong
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Hiromi Hirata
- National Institute of Genetics, Mishima 411-8540, Japan
| | - John Y. Kuwada
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
32
|
Roy NM, Arpie B, Lugo J, Linney E, Levin ED, Cerutti D. Brief embryonic strychnine exposure in zebrafish causes long-term adult behavioral impairment with indications of embryonic synaptic changes. Neurotoxicol Teratol 2012; 34:587-91. [PMID: 23022260 DOI: 10.1016/j.ntt.2012.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 08/01/2012] [Accepted: 08/01/2012] [Indexed: 10/28/2022]
Abstract
Zebrafish provide a powerful model of the impacts of embryonic toxicant exposure on neural development that may result in long-term behavioral dysfunction. In this study, zebrafish embryos were treated with 1.5mM strychnine for short embryonic time windows to induce transient changes in inhibitory neural signaling, and were subsequently raised in untreated water until adulthood. PCR analysis showed indications that strychnine exposure altered expression of some genes related to glycinergic, GABAergic and glutamatergic neuronal synapses during embryonic development. In adulthood, treated fish showed significant changes in swimming speed and tank diving behavior compared to controls. Taken together, these data show that a short embryonic exposure to a neurotoxicant can alter development of neural synapses and lead to changes in adult behavior.
Collapse
Affiliation(s)
- Nicole M Roy
- Department of Biology, Sacred Heart University, Fairfield, CT, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
A central goal of modern neuroscience is to obtain a mechanistic understanding of higher brain functions under healthy and diseased conditions. Addressing this challenge requires rigorous experimental and theoretical analysis of neuronal circuits. Recent advances in optogenetics, high-resolution in vivo imaging, and reconstructions of synaptic wiring diagrams have created new opportunities to achieve this goal. To fully harness these methods, model organisms should allow for a combination of genetic and neurophysiological approaches in vivo. Moreover, the brain should be small in terms of neuron numbers and physical size. A promising vertebrate organism is the zebrafish because it is small, it is transparent at larval stages and it offers a wide range of genetic tools and advantages for neurophysiological approaches. Recent studies have highlighted the potential of zebrafish for exhaustive measurements of neuronal activity patterns, for manipulations of defined cell types in vivo and for studies of causal relationships between circuit function and behavior. In this article, we summarize background information on the zebrafish as a model in modern systems neuroscience and discuss recent results.
Collapse
Affiliation(s)
- Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstr. 66, CH-4058 Basel, Switzerland.
| | | | | |
Collapse
|
34
|
The biological role of the glycinergic synapse in early zebrafish motility. Neurosci Res 2011; 71:1-11. [PMID: 21712054 DOI: 10.1016/j.neures.2011.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/08/2011] [Accepted: 04/15/2011] [Indexed: 01/09/2023]
Abstract
Glycine mediates fast inhibitory neurotransmission in the spinal cord, brainstem and retina. Loss of synaptic glycinergic transmission in vertebrates leads to a severe locomotion defect characterized by an exaggerated startle response accompanied by transient muscle rigidity in response to sudden acoustic or tactile stimuli. Several molecular components of the glycinergic synapse have been characterized as an outcome of genetic and physiological analyses of synaptogenesis in mammals. Recently, the glycinergic synapse has been studied using a forward genetic approach in zebrafish. This review aims to discuss molecular components of the glycinergic synapse, such as glycine receptor subunits, gephyrin, gephyrin-binding proteins and glycine transporters, as well as recent studies relevant to the genetic analysis of the glycinergic synapse in zebrafish.
Collapse
|
35
|
Moly PK, Hatta K. Early glycinergic axon contact with the Mauthner neuron during zebrafish development. Neurosci Res 2011; 70:251-9. [PMID: 21397641 DOI: 10.1016/j.neures.2011.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 03/04/2011] [Accepted: 03/04/2011] [Indexed: 10/18/2022]
Abstract
Glycinergic neurons are the major inhibitory neurons in the vertebrate central nervous system. In teleosts, they play important roles in the escape response by regulating the activity of the Mauthner (M-) cells. Here we studied the contact between glycinergic axons and the M-cells in early zebrafish embryos by double immunostaining with an anti-glycine antibody and the 3A10 antibody that labels M-cells. We also studied a transgenic line, Tg(GlyT2:GFP), in which GFP is expressed under the control of the promoter for the glycine transporter-2 gene. The initial contacts by ascending glycinergic axons on the M-soma were observed within 27h post-fertilization (hpf) on the lateral part of the ventral surface of the M-soma. Stochastic labeling of glycinergic neurons was then performed by injecting a GlyT2:GFP construct into early cleaving eggs. We identified the origin of the earliest glycinergic axons that contact the M-soma as commissural neurons, located in the anterior spinal cord, whose axons ascend along the lateral longitudinal fascicles with a short descending branch. We also found, in the fourth rhombomere, late-developed glycinergic commissural neurons whose axons contact anterior or posterior edge of both M-somas. This study provides the first example of the initial development of an inhibitory network on an identifiable neuron in vertebrates.
Collapse
Affiliation(s)
- Pricila Khan Moly
- Graduate School of Life Science, University of Hyogo, Hyogo 678-1297, Japan
| | | |
Collapse
|
36
|
Adrio F, Rodríguez-Moldes I, Anadón R. Distribution of glycine immunoreactivity in the brain of the Siberian sturgeon (Acipenser baeri): Comparison with γ-aminobutyric acid. J Comp Neurol 2011; 519:1115-42. [DOI: 10.1002/cne.22556] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
37
|
Specific brainstem neurons switch each other into pacemaker mode to drive movement by activating NMDA receptors. J Neurosci 2011; 30:16609-20. [PMID: 21148000 DOI: 10.1523/jneurosci.3695-10.2010] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rhythmic activity is central to brain function. In the vertebrate CNS, the neuronal circuits for breathing and locomotion involve inhibition and also neurons acting as pacemakers, but identifying the neurons responsible has proven difficult. By studying simple hatchling Xenopus laevis tadpoles, we have already identified a population of electrically coupled hindbrain neurons (dINs) that drive swimming. During rhythm generation, dINs release glutamate to excite each other and activate NMDA receptors (NMDARs). The resulting depolarization enables a network mechanism for swimming rhythm generation that depends on reciprocal inhibition between antagonistic right and left sides. Surprisingly, a surgically isolated hemi-CNS without inhibition can still generate swimming-like rhythms. We have now discovered that activation of NMDARs transforms dINs, which normally fire singly to current injection, into pacemakers firing within the normal swimming frequency range (10-25 Hz). When dIN firing is blocked pharmacologically, this NMDAR activation produces 10 Hz membrane potential oscillations that persist when electrical coupling is blocked but not when the voltage-dependent gating of NMDARs by Mg²+ is removed. The NMDA-induced oscillations and pacemaker firing at swimming frequency are unique to the dIN population and do not occur in other spinal neurons. We conclude that NMDAR-mediated self-resetting switches critical neurons that drive swimming into pacemaker mode only during locomotion where it provides an additional, parallel mechanism for rhythm generation. This allows rhythm generation in a half-CNS and raises the possibility that such concealed pacemaker properties may be present underlying rhythm generation in other vertebrate brain networks.
Collapse
|
38
|
Low SE, Zhou W, Choong I, Saint-Amant L, Sprague SM, Hirata H, Cui WW, Hume RI, Kuwada JY. Na(v)1.6a is required for normal activation of motor circuits normally excited by tactile stimulation. Dev Neurobiol 2010; 70:508-22. [PMID: 20225246 DOI: 10.1002/dneu.20791] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A screen for zebrafish motor mutants identified two noncomplementing alleles of a recessive mutation that were named non-active (nav(mi89) and nav(mi130)). nav embryos displayed diminished spontaneous and touch-evoked escape behaviors during the first 3 days of development. Genetic mapping identified the gene encoding Na(V)1.6a (scn8aa) as a potential candidate for nav. Subsequent cloning of scn8aa from the two alleles of nav uncovered two missense mutations in Na(V)1.6a that eliminated channel activity when assayed heterologously. Furthermore, the injection of RNA encoding wild-type scn8aa rescued the nav mutant phenotype indicating that scn8aa was the causative gene of nav. In-vivo electrophysiological analysis of the touch-evoked escape circuit indicated that voltage-dependent inward current was decreased in mechanosensory neurons in mutants, but they were able to fire action potentials. Furthermore, tactile stimulation of mutants activated some neurons downstream of mechanosensory neurons but failed to activate the swim locomotor circuit in accord with the behavioral response of initial escape contractions but no swimming. Thus, mutant mechanosensory neurons appeared to respond to tactile stimulation but failed to initiate swimming. Interestingly fictive swimming could be initiated pharmacologically suggesting that a swim circuit was present in mutants. These results suggested that Na(V)1.6a was required for touch-induced activation of the swim locomotor network.
Collapse
Affiliation(s)
- Sean E Low
- Neuroscience Program, University of Michigan, Ann Arbor, Michigan 48109-1048, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chalphin AV, Saha MS. The specification of glycinergic neurons and the role of glycinergic transmission in development. Front Mol Neurosci 2010; 3:11. [PMID: 20461146 PMCID: PMC2866564 DOI: 10.3389/fnmol.2010.00011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Accepted: 03/23/2010] [Indexed: 12/16/2022] Open
Abstract
Glycine's role as an inhibitory neurotransmitter in the adult vertebrate nervous system has been well characterized in a number of different model organisms. However, a full understanding of glycinergic transmission requires a knowledge of how glycinergic synapses emerge and the role of glycinergic signaling during development. Recent literature has provided a detailed picture of the developmental expression of many of the molecular components that comprise the glycinergic phenotype, namely the glycine transporters and the glycine receptor subunits; the transcriptional networks leading to the expression of this important neurotransmitter phenotype are also being elucidated. An equally important focus of research has revealed the critical role of glycinergic signaling in sculpting many different aspects of neural development. This review examines the current literature detailing the expression patterns of the components of the glycinergic phenotype in various vertebrate model organisms over the course of development and the molecular mechanisms governing the expression of the glycinergic phenotype. The review then surveys the recent work on the role of glycinergic signaling in the developing nervous system and concludes with an overview of areas for further research.
Collapse
|
40
|
Hirata H, Carta E, Yamanaka I, Harvey RJ, Kuwada JY. Defective glycinergic synaptic transmission in zebrafish motility mutants. Front Mol Neurosci 2010; 2:26. [PMID: 20161699 PMCID: PMC2813725 DOI: 10.3389/neuro.02.026.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 11/11/2009] [Indexed: 11/20/2022] Open
Abstract
Glycine is a major inhibitory neurotransmitter in the spinal cord and brainstem. Recently, in vivo analysis of glycinergic synaptic transmission has been pursued in zebrafish using molecular genetics. An ENU mutagenesis screen identified two behavioral mutants that are defective in glycinergic synaptic transmission. Zebrafish bandoneon (beo) mutants have a defect in glrbb, one of the duplicated glycine receptor (GlyR) beta subunit genes. These mutants exhibit a loss of glycinergic synaptic transmission due to a lack of synaptic aggregation of GlyRs. Due to the consequent loss of reciprocal inhibition of motor circuits between the two sides of the spinal cord, motor neurons activate simultaneously on both sides resulting in bilateral contraction of axial muscles of beo mutants, eliciting the so-called 'accordion' phenotype. Similar defects in GlyR subunit genes have been observed in several mammals and are the basis for human hyperekplexia/startle disease. By contrast, zebrafish shocked (sho) mutants have a defect in slc6a9, encoding GlyT1, a glycine transporter that is expressed by astroglial cells surrounding the glycinergic synapse in the hindbrain and spinal cord. GlyT1 mediates rapid uptake of glycine from the synaptic cleft, terminating synaptic transmission. In zebrafish sho mutants, there appears to be elevated extracellular glycine resulting in persistent inhibition of postsynaptic neurons and subsequent reduced motility, causing the 'twitch-once' phenotype. We review current knowledge regarding zebrafish 'accordion' and 'twitch-once' mutants, including beo and sho, and report the identification of a new alpha2 subunit that revises the phylogeny of zebrafish GlyRs.
Collapse
Affiliation(s)
- Hiromi Hirata
- Graduate School of Science, Nagoya UniversityNagoya, Japan
| | - Eloisa Carta
- Department of Pharmacology, The School of PharmacyLondon, UK
| | - Iori Yamanaka
- Graduate School of Science, Nagoya UniversityNagoya, Japan
| | | | - John Y. Kuwada
- Department of Molecular, Cellular and Developmental Biology, University of MichiganAnn Arbor, MI, USA
| |
Collapse
|
41
|
Abstract
The nervous system can generate rhythms of various frequencies; on the low-frequency side, we have the circuits regulating circadian rhythms with a 24-h period, while on the high-frequency side we have the motor circuits that underlie flight in a hummingbird. Given the ubiquitous nature of rhythms, it is surprising that we know very little of the cellular and molecular mechanisms that produce them in the embryos and of their potential role during the development of neuronal circuits. Recently, zebrafish has been developed as a vertebrate model to study the genetics of neural development. Zebrafish offer several advantages to the study of nervous system development including optical and electrophysiological analysis of neuronal activity even at the earliest embryonic stages. This unique combination of physiology and genetics in the same animal model has led to insights into the development of neuronal networks. This chapter reviews work on the development of zebrafish motor rhythms and speculates on birth and maturation of the circuits that produce them.
Collapse
|
42
|
Ganser LR, Dallman JE. Glycinergic synapse development, plasticity, and homeostasis in zebrafish. Front Mol Neurosci 2009; 2:30. [PMID: 20126315 PMCID: PMC2815536 DOI: 10.3389/neuro.02.030.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 11/26/2009] [Indexed: 11/13/2022] Open
Abstract
The zebrafish glial glycine transporter 1 (GlyT1) mutant provides an animal model in which homeostatic plasticity at glycinergic synapses restores rhythmic motor behaviors. GlyT1 mutants, initially paralyzed by the build-up of the inhibitory neurotransmitter glycine, stage a gradual recovery that is associated with reductions in the strength of evoked glycinergic responses. Gradual motor recovery suggests sequential compensatory mechanisms that culminate in the down-regulation of the neuronal glycine receptor. However, how motor recovery is initiated and how other forms of plasticity contribute to behavioral recovery are still outstanding questions that we discuss in the context of (1) glycinergic synapses as they function in spinal circuits that produce rhythmic motor behaviors, (2) the proteins involved in regulating glycinergic synaptic strength, (3) current models of glycinergic synaptogenesis, and (4) plasticity mechanisms that modulate the strength of glycinergic synapses. Concluding remarks (5) explore the potential for distinct plasticity mechanisms to act in concert at different spatial and temporal scales to achieve a dynamic stability that results in balanced motor behaviors.
Collapse
Affiliation(s)
- Lisa R Ganser
- Department of Biology, University of Miami Coral Gables, FL, USA
| | | |
Collapse
|
43
|
Pietri T, Manalo E, Ryan J, Saint-Amant L, Washbourne P. Glutamate drives the touch response through a rostral loop in the spinal cord of zebrafish embryos. Dev Neurobiol 2009; 69:780-95. [PMID: 19634126 DOI: 10.1002/dneu.20741] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Characterizing connectivity in the spinal cord of zebrafish embryos is not only prerequisite to understanding the development of locomotion, but is also necessary for maximizing the potential of genetic studies of circuit formation in this model system. During their first day of development, zebrafish embryos show two simple motor behaviors. First, they coil their trunks spontaneously, and a few hours later they start responding to touch with contralateral coils. These behaviors are contemporaneous until spontaneous coils become infrequent by 30 h. Glutamatergic neurons are distributed throughout the embryonic spinal cord, but their contribution to these early motor behaviors in immature zebrafish is still unclear. We demonstrate that the kinetics of spontaneous coiling and touch-evoked responses show distinct developmental time courses and that the touch response is dependent on AMPA-type glutamate receptor activation. Transection experiments suggest that the circuits required for touch-evoked responses are confined to the spinal cord and that only the most rostral part of the spinal cord is sufficient for triggering the full response. This rostral sensory connection is presumably established via CoPA interneurons, as they project to the rostral spinal cord. Electrophysiological analysis demonstrates that these neurons receive short latency AMPA-type glutamatergic inputs in response to ipsilateral tactile stimuli. We conclude that touch responses in early embryonic zebrafish arise only after glutamatergic synapses connect sensory neurons and interneurons to the contralateral motor network via a rostral loop. This helps define an elementary circuit that is modified by the addition of sensory inputs, resulting in behavioral transformation.
Collapse
Affiliation(s)
- Thomas Pietri
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
Neurobiologists have long sought to understand how circuits in the nervous system are organized to generate the precise neural outputs that underlie particular behaviours. The motor circuits in the spinal cord that control locomotion, commonly referred to as central pattern generator networks, provide an experimentally tractable model system for investigating how moderately complex ensembles of neurons generate select motor behaviours. The advent of novel molecular and genetic techniques coupled with recent advances in our knowledge of spinal cord development means that a comprehensive understanding of how the motor circuitry is organized and operates may be within our grasp.
Collapse
Affiliation(s)
- Martyn Goulding
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA.
| |
Collapse
|
45
|
Villar-Cerviño V, Barreiro-Iglesias A, Anadón R, Rodicio MC. Development of glycine immunoreactivity in the brain of the sea lamprey: Comparison with γ-aminobutyric acid immunoreactivity. J Comp Neurol 2009; 512:747-67. [DOI: 10.1002/cne.21916] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
A modified acetylcholine receptor delta-subunit enables a null mutant to survive beyond sexual maturation. J Neurosci 2009; 28:13223-31. [PMID: 19052214 DOI: 10.1523/jneurosci.2814-08.2008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The contraction of skeletal muscle is dependent on synaptic transmission through acetylcholine receptors (AChRs) at the neuromuscular junction (NMJ). The lack of an AChR subunit causes a fetal akinesia in humans, leading to death in the first trimester and characteristic features of Fetal Akinesia Deformation Sequences (FADS). A corresponding null mutation of the delta-subunit in zebrafish (sofa potato; sop) leads to the death of embryos around 5 d postfertilization (dpf). In sop(-/-) mutants, we expressed modified delta-subunits, with one (delta1YFP) or two yellow fluorescent protein (delta2YFP) molecules fused at the intracellular loop, under the control of an alpha-actin promoter. AChRs containing these fusion proteins are fluorescent, assemble on the plasma membrane, make clusters under motor neuron endings, and generate synaptic current. We screened for germ-line transmission of the transgene and established a line of sop(-/-) fish stably expressing the delta2YFP. These delta2YFP/sop(-/-) embryos can mount escape behavior close to that of their wild-type siblings. Synaptic currents in these embryos had a smaller amplitude, slower rise time, and slower decay when compared with wild-type fish. Remarkably, these embryos grow to adulthood and display complex behaviors such as feeding and breeding. To the best of our knowledge, this is the first case of a mutant animal corresponding to first trimester lethality in human that has been rescued by a transgene and survived to adulthood. In the rescued fish, a foreign promoter drove the transgene expression and the NMJ had altered synaptic strength. The survival of the transgenic animal delineates requirements for gene therapies of NMJ.
Collapse
|
47
|
Acute nicotine exposure and modulation of a spinal motor circuit in embryonic zebrafish. Toxicol Appl Pharmacol 2008; 239:1-12. [PMID: 19121331 DOI: 10.1016/j.taap.2008.08.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 08/17/2008] [Accepted: 08/19/2008] [Indexed: 11/20/2022]
Abstract
The zebrafish model system is ideal for studying nervous system development. Ultimately, one would like to link the developmental biology to various aspects of behavior. We are studying the consequences of nicotine exposure on nervous system development in zebrafish and have previously shown that chronic nicotine exposure produces paralysis. We also have made observations that the embryos moved in the initial minutes of the exposure as the bend rates of the musculature increased. This nicotine induced behavior manifests as an increase in the rate of spinal musculature bends, which spontaneously begin at approximately 17 h post fertilization. The behavioral observations prompted the systematic characterization of nicotine-induced modulation of zebrafish embryonic motor output; bends of the trunk musculature. We first characterized embryonic motor output in zebrafish embryos with and without their chorions. We then characterized the motor output in embryos raised at 28 degrees C and 25 degrees C. The act of dechorionation along with temperature influenced the embryonic bend rate. We show that nicotine exposure increases embryonic motor output. Nicotine exposure caused the musculature bends to alternate in a left-right-left fashion. Nicotine was able to produce this phenotype in embryos lacking supraspinal input. We then characterize the kinetics of nicotine influx and efflux and demonstrate that nicotine as low as 1 microM can disrupt embryonic physiology. Taken together, these results indicate the presence of nicotinic acetylcholine receptors (nAChRs) associated with embryonic spinal motor circuits early in embryogenesis.
Collapse
|
48
|
The genetics of hyperekplexia: more than startle! Trends Genet 2008; 24:439-47. [DOI: 10.1016/j.tig.2008.06.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/04/2008] [Accepted: 06/04/2008] [Indexed: 11/20/2022]
|
49
|
Mongeon R, Gleason MR, Masino MA, Fetcho JR, Mandel G, Brehm P, Dallman JE. Synaptic homeostasis in a zebrafish glial glycine transporter mutant. J Neurophysiol 2008; 100:1716-23. [PMID: 18715895 DOI: 10.1152/jn.90596.2008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Truncated escape responses characteristic of the zebrafish shocked mutant result from a defective glial glycine transporter (GlyT1). In homozygous GlyT1 mutants, irrigating brain ventricles with glycine-free solution rescues normal swimming. Conversely, elevating brain glycine levels restores motility defects. These experiments are consistent with previous studies that demonstrate regulation of global glycine levels in the CNS as a primary function of GlyT1. As GlyT1 mutants mature, their ability to mount an escape response naturally recovers. To understand the basis of this recovery, we assay synaptic transmission in primary spinal motor neurons by measuring stimulus-evoked postsynaptic potentials. At the peak of the motility defect, inhibitory synaptic potentials are both significantly larger and more prolonged indicating a prominent role for GlyT1 in shaping fast synaptic transmission. However, as GlyT1 mutants naturally regain their ability to swim, the amplitude of inhibitory potentials decreases to below wild-type levels. In parallel with diminishing synaptic potentials, the glycine concentration required to evoke the mutant motility defect increases 61-fold during behavioral recovery. Behavioral recovery is also mirrored by a reduction in the levels of both glycine receptor protein and transcript. These results suggest that increased CNS glycine tolerance and reduced glycine receptor expression in GlyT1 mutants reflect compensatory mechanisms for functional recovery from excess nervous system inhibition.
Collapse
Affiliation(s)
- Rebecca Mongeon
- Howard Hughes Medical Institute, Department of Neurobiology and Behavior, State University of New York, Stony Brook, NY, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
McLean DL, Fetcho JR. Using imaging and genetics in zebrafish to study developing spinal circuits in vivo. Dev Neurobiol 2008; 68:817-34. [PMID: 18383546 DOI: 10.1002/dneu.20617] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Imaging and molecular approaches are perfectly suited to young, transparent zebrafish (Danio rerio), where they have allowed novel functional studies of neural circuits and their links to behavior. Here, we review cutting-edge optical and genetic techniques used to dissect neural circuits in vivo and discuss their application to future studies of developing spinal circuits using living zebrafish. We anticipate that these experiments will reveal general principles governing the assembly of neural circuits that control movements.
Collapse
Affiliation(s)
- David L McLean
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA.
| | | |
Collapse
|