1
|
Nakagawa N. The neuronal Golgi in neural circuit formation and reorganization. Front Neural Circuits 2024; 18:1504422. [PMID: 39703196 PMCID: PMC11655203 DOI: 10.3389/fncir.2024.1504422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The Golgi apparatus is a central hub in the intracellular secretory pathway. By positioning in the specific intracellular region and transporting materials to spatially restricted compartments, the Golgi apparatus contributes to the cell polarity establishment and morphological specification in diverse cell types. In neurons, the Golgi apparatus mediates several essential steps of initial neural circuit formation during early brain development, such as axon-dendrite polarization, neuronal migration, primary dendrite specification, and dendritic arbor elaboration. Moreover, neuronal activity-dependent remodeling of the Golgi structure enables morphological changes in neurons, which provides the cellular basis of circuit reorganization during postnatal critical period. In this review, I summarize recent findings illustrating the unique Golgi positioning and its developmental dynamics in various types of neurons. I also discuss the upstream regulators for the Golgi positioning in neurons, and functional roles of the Golgi in neural circuit formation and reorganization. Elucidating how Golgi apparatus sculpts neuronal connectivity would deepen our understanding of the cellular/molecular basis of neural circuit development and plasticity.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Japan
| |
Collapse
|
2
|
Kim TY, Roychaudhury A, Kim HT, Choi TI, Baek ST, Thyme SB, Kim CH. Impairments of cerebellar structure and function in a zebrafish KO of neuropsychiatric risk gene znf536. Transl Psychiatry 2024; 14:82. [PMID: 38331943 PMCID: PMC10853220 DOI: 10.1038/s41398-024-02806-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Genetic variants in ZNF536 contribute to the risk for neuropsychiatric disorders such as schizophrenia, autism, and others. The role of this putative transcriptional repressor in brain development and function is, however, largely unknown. We generated znf536 knockout (KO) zebrafish and studied their behavior, brain anatomy, and brain function. Larval KO zebrafish showed a reduced ability to compete for food, resulting in decreased total body length and size. This phenotype can be rescued by segregating the homozygous KO larvae from their wild-type and heterozygous siblings, enabling studies of adult homozygous KO animals. In adult KO zebrafish, we observed significant reductions in anxiety-like behavior and social interaction. These znf536 KO zebrafish have decreased cerebellar volume, corresponding to decreased populations of specific neuronal cells, especially in the valvular cerebelli (Va). Finally, using a Tg[mbp:mgfp] line, we identified a previously undetected myelin structure located bilaterally within the Va, which also displayed a reduction in volume and disorganization in KO zebrafish. These findings indicate an important role for ZNF536 in brain development and implicate the cerebellum in the pathophysiology of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tae-Yoon Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | | | - Hyun-Taek Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, 31151, South Korea
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Summer B Thyme
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, USA.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
3
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
4
|
Nakagawa N, Iwasato T. Golgi polarity shift instructs dendritic refinement in the neonatal cortex by mediating NMDA receptor signaling. Cell Rep 2023; 42:112843. [PMID: 37516101 DOI: 10.1016/j.celrep.2023.112843] [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: 03/07/2023] [Revised: 05/15/2023] [Accepted: 07/05/2023] [Indexed: 07/31/2023] Open
Abstract
Dendritic refinement is a critical component of activity-dependent neuronal circuit maturation, through which individual neurons establish specific connectivity with their target axons. Here, we demonstrate that the developmental shift of Golgi polarity is a key process in dendritic refinement. During neonatal development, the Golgi apparatus in layer 4 spiny stellate (SS) neurons in the mouse barrel cortex lose their original apical positioning and acquire laterally polarized distributions. This lateral Golgi polarity, which is oriented toward the barrel center, peaks on postnatal days 5-7 (P5-P7) and disappears by P15, which aligns with the developmental time course of SS neuron dendritic refinement. Genetic ablation of N-methyl-D-aspartate (NMDA) receptors, key players in dendritic refinement, disturbs the lateral Golgi polarity. Golgi polarity manipulation disrupts the asymmetric dendritic projection pattern and the primary-whisker-specific response of SS neurons. Our results elucidate activity-dependent Golgi dynamics and their critical role in developmental neuronal circuit refinement.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics (NIG), Mishima, Shizuoka 411-8540, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan.
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics (NIG), Mishima, Shizuoka 411-8540, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Shizuoka 411-8540, Japan.
| |
Collapse
|
5
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Sugihara T, Shimizu T, Koyanagi M, Terakita A, Hibi M. Optogenetic manipulation of Gq- and Gi/o-coupled receptor signaling in neurons and heart muscle cells. eLife 2023; 12:e83974. [PMID: 37589544 PMCID: PMC10435233 DOI: 10.7554/elife.83974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) transmit signals into cells depending on the G protein type. To analyze the functions of GPCR signaling, we assessed the effectiveness of animal G-protein-coupled bistable rhodopsins that can be controlled into active and inactive states by light application using zebrafish. We expressed Gq- and Gi/o-coupled bistable rhodopsins in hindbrain reticulospinal V2a neurons, which are involved in locomotion, or in cardiomyocytes. Light stimulation of the reticulospinal V2a neurons expressing Gq-coupled spider Rh1 resulted in an increase in the intracellular Ca2+ level and evoked swimming behavior. Light stimulation of cardiomyocytes expressing the Gi/o-coupled mosquito Opn3, pufferfish TMT opsin, or lamprey parapinopsin induced cardiac arrest, and the effect was suppressed by treatment with pertussis toxin or barium, suggesting that Gi/o-dependent regulation of inward-rectifier K+ channels controls cardiac function. These data indicate that these rhodopsins are useful for optogenetic control of GPCR-mediated signaling in zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya UniversityNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | | | - Akihisa Terakita
- Graduate School of Science, Osaka Metropolitan UniversityOsakaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| |
Collapse
|
6
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Shimizu T, Hososhima S, Tsunoda SP, Kandori H, Hibi M. Optogenetic manipulation of neuronal and cardiomyocyte functions in zebrafish using microbial rhodopsins and adenylyl cyclases. eLife 2023; 12:e83975. [PMID: 37589546 PMCID: PMC10435232 DOI: 10.7554/elife.83975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
Even though microbial photosensitive proteins have been used for optogenetics, their use should be optimized to precisely control cell and tissue functions in vivo. We exploited GtCCR4 and KnChR, cation channelrhodopsins from algae, BeGC1, a guanylyl cyclase rhodopsin from a fungus, and photoactivated adenylyl cyclases (PACs) from cyanobacteria (OaPAC) or bacteria (bPAC), to control cell functions in zebrafish. Optical activation of GtCCR4 and KnChR in the hindbrain reticulospinal V2a neurons, which are involved in locomotion, induced swimming behavior at relatively short latencies, whereas activation of BeGC1 or PACs achieved it at long latencies. Activation of GtCCR4 and KnChR in cardiomyocytes induced cardiac arrest, whereas activation of bPAC gradually induced bradycardia. KnChR activation led to an increase in intracellular Ca2+ in the heart, suggesting that depolarization caused cardiac arrest. These data suggest that these optogenetic tools can be used to reveal the function and regulation of zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Takashi Shimizu
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shoko Hososhima
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Satoshi P Tsunoda
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| |
Collapse
|
7
|
Pose-Méndez S, Schramm P, Valishetti K, Köster RW. Development, circuitry, and function of the zebrafish cerebellum. Cell Mol Life Sci 2023; 80:227. [PMID: 37490159 PMCID: PMC10368569 DOI: 10.1007/s00018-023-04879-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/30/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
The cerebellum represents a brain compartment that first appeared in gnathostomes (jawed vertebrates). Besides the addition of cell numbers, its development, cytoarchitecture, circuitry, physiology, and function have been highly conserved throughout avian and mammalian species. While cerebellar research in avian and mammals is extensive, systematic investigations on this brain compartment in zebrafish as a teleostian model organism started only about two decades ago, but has provided considerable insight into cerebellar development, physiology, and function since then. Zebrafish are genetically tractable with nearly transparent small-sized embryos, in which cerebellar development occurs within a few days. Therefore, genetic investigations accompanied with non-invasive high-resolution in vivo time-lapse imaging represents a powerful combination for interrogating the behavior and function of cerebellar cells in their complex native environment.
Collapse
Affiliation(s)
- Sol Pose-Méndez
- Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106, Braunschweig, Germany.
| | - Paul Schramm
- Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106, Braunschweig, Germany
| | - Komali Valishetti
- Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106, Braunschweig, Germany
| | - Reinhard W Köster
- Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, 38106, Braunschweig, Germany.
| |
Collapse
|
8
|
Louis ED, Martuscello RT, Gionco JT, Hartstone WG, Musacchio JB, Portenti M, McCreary M, Kuo SH, Vonsattel JPG, Faust PL. Histopathology of the cerebellar cortex in essential tremor and other neurodegenerative motor disorders: comparative analysis of 320 brains. Acta Neuropathol 2023; 145:265-283. [PMID: 36607423 PMCID: PMC10461794 DOI: 10.1007/s00401-022-02535-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023]
Abstract
In recent years, numerous morphologic changes have been identified in the essential tremor (ET) cerebellar cortex, distinguishing ET from control brains. These findings have not been fully contextualized within a broader degenerative disease spectrum, thus limiting their interpretability. Building off our prior study and now doubling the sample size, we conducted comparative analyses in a postmortem series of 320 brains on the severity and patterning of cerebellar cortex degenerative changes in ET (n = 100), other neurodegenerative disorders of the cerebellum [spinocerebellar ataxias (SCAs, n = 47, including 13 SCA3 and 34 SCA1, 2, 6, 7, 8, 14); Friedreich's ataxia (FA, n = 13); multiple system atrophy (MSA), n = 29], and other disorders that may involve the cerebellum [Parkinson's disease (PD), n = 62; dystonia, n = 19] versus controls (n = 50). We generated data on 37 quantitative morphologic metrics, grouped into 8 broad categories: Purkinje cell (PC) loss, heterotopic PCs, PC dendritic changes, PC axonal changes (torpedoes), PC axonal changes (other than torpedoes), PC axonal changes (torpedo-associated), basket cell axonal hypertrophy, and climbing fiber-PC synaptic changes. Principal component analysis of z scored raw data across all diagnoses (11,651 data items) revealed that diagnostic groups were not uniform with respect to pathology. Dystonia and PD each differed from controls in only 4/37 and 5/37 metrics, respectively, whereas ET differed in 21, FA in 10, SCA3 in 10, MSA in 21, and SCA1/2/6/7/8/14 in 27. Pathological changes were generally on the milder end of the degenerative spectrum in ET, FA and SCA3, and on the more severe end of that spectrum in SCA1/2/6/7/8/14. Comparative analyses across morphologic categories demonstrated differences in relative expression, defining distinctive patterns of changes in these groups. In summary, we present a robust and reproducible method that identifies somewhat distinctive signatures of degenerative changes in the cerebellar cortex that mark each of these disorders.
Collapse
Affiliation(s)
- Elan D Louis
- Department of Neurology, University of Texas Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813, USA.
| | - Regina T Martuscello
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - John T Gionco
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Whitney G Hartstone
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Jessica B Musacchio
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Marisa Portenti
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| | - Morgan McCreary
- Department of Neurology, University of Texas Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813, USA
| | - Sheng-Han Kuo
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jean-Paul G Vonsattel
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
9
|
Dorigo A, Valishetti K, Hetsch F, Matsui H, Meier JC, Namikawa K, Köster RW. Functional regionalization of the differentiating cerebellar Purkinje cell population occurs in an activity-dependent manner. Front Mol Neurosci 2023; 16:1166900. [PMID: 37181649 PMCID: PMC10174242 DOI: 10.3389/fnmol.2023.1166900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction The cerebellum is organized into functional regions each dedicated to process different motor or sensory inputs for controlling different locomotor behaviors. This functional regionalization is prominent in the evolutionary conserved single-cell layered Purkinje cell (PC) population. Fragmented gene expression domains suggest a genetic organization of PC layer regionalization during cerebellum development. However, the establishment of such functionally specific domains during PC differentiation remained elusive. Methods and results We show the progressive emergence of functional regionalization of PCs from broad responses to spatially restricted regions in zebrafish by means of in vivo Ca2+-imaging during stereotypic locomotive behavior. Moreover, we reveal that formation of new dendritic spines during cerebellar development using in vivo imaging parallels the time course of functional domain development. Pharmacological as well as cell-type specific optogenetic inhibition of PC neuronal activity results in reduced PC dendritic spine density and an altered stagnant pattern of functional domain formation in the PC layer. Discussion Hence, our study suggests that functional regionalization of the PC layer is driven by physiological activity of maturing PCs themselves.
Collapse
Affiliation(s)
- Alessandro Dorigo
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Komali Valishetti
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Hetsch
- Cell Physiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
- Institute of Pathophysiology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Hideaki Matsui
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jochen C. Meier
- Cell Physiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kazuhiko Namikawa
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- *Correspondence: Kazuhiko Namikawa,
| | - Reinhard W. Köster
- Cellular and Molecular Neurobiology, Technische Universität Braunschweig, Braunschweig, Germany
- Reinhard W. Köster,
| |
Collapse
|
10
|
Hasegawa K, Matsui TK, Kondo J, Kuwako KI. N-WASP-Arp2/3 signaling controls multiple steps of dendrite maturation in Purkinje cells in vivo. Development 2022; 149:285127. [PMID: 36469048 DOI: 10.1242/dev.201214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
During neural development, the actin filament network must be precisely regulated to form elaborate neurite structures. N-WASP tightly controls actin polymerization dynamics by activating an actin nucleator Arp2/3. However, the importance of N-WASP-Arp2/3 signaling in the assembly of neurite architecture in vivo has not been clarified. Here, we demonstrate that N-WASP-Arp2/3 signaling plays a crucial role in the maturation of cerebellar Purkinje cell (PC) dendrites in vivo in mice. N-WASP was expressed and activated in developing PCs. Inhibition of Arp2/3 and N-WASP from the beginning of dendrite formation severely disrupted the establishment of a single stem dendrite, which is a characteristic basic structure of PC dendrites. Inhibition of Arp2/3 after stem dendrite formation resulted in hypoplasia of the PC dendritic tree. Cdc42, an upstream activator of N-WASP, is required for N-WASP-Arp2/3 signaling-mediated PC dendrite maturation. In addition, overactivation of N-WASP is also detrimental to dendrite formation in PCs. These findings reveal that proper activation of N-WASP-Arp2/3 signaling is crucial for multiple steps of PC dendrite maturation in vivo.
Collapse
Affiliation(s)
- Koichi Hasegawa
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Takeshi K Matsui
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Junpei Kondo
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Ken-Ichiro Kuwako
- Department of Neural and Muscular Physiology, School of Medicine, Shimane University, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| |
Collapse
|
11
|
Purkinje cells located in the adult zebrafish valvula cerebelli exhibit variable functional responses. Sci Rep 2021; 11:18408. [PMID: 34526620 PMCID: PMC8443705 DOI: 10.1038/s41598-021-98035-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023] Open
Abstract
Purkinje cells are critically involved in processing the cerebellar functions by shaping and coordinating commands that they receive. Here, we demonstrate experimentally that in the adult zebrafish valvular part of the cerebellum, the Purkinje cells exhibited variable firing and functional responses and allowed the categorization into three firing classes. Compared with the Purkinje cells in the corpus cerebelli, the valvular Purkinje cells receive weak and occasional input from the inferior olive and are not active during locomotion. Together, our findings expand further the regional functional differences of the Purkinje cell population and expose their non-locomotor functionality.
Collapse
|
12
|
Sitaraman S, Yadav G, Agarwal V, Jabeen S, Verma S, Jadhav M, Thirumalai V. Gjd2b-mediated gap junctions promote glutamatergic synapse formation and dendritic elaboration in Purkinje neurons. eLife 2021; 10:68124. [PMID: 34346310 PMCID: PMC8382294 DOI: 10.7554/elife.68124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Gap junctions between neurons serve as electrical synapses, in addition to conducting metabolites and signaling molecules. During development, early-appearing gap junctions are thought to prefigure chemical synapses, which appear much later. We present evidence for this idea at a central, glutamatergic synapse and provide some mechanistic insights. Loss or reduction in the levels of the gap junction protein Gjd2b decreased the frequency of glutamatergic miniature excitatory postsynaptic currents (mEPSCs) in cerebellar Purkinje neurons (PNs) in larval zebrafish. Ultrastructural analysis in the molecular layer showed decreased synapse density. Further, mEPSCs had faster kinetics and larger amplitudes in mutant PNs, consistent with their stunted dendritic arbors. Time-lapse microscopy in wild-type and mutant PNs reveals that Gjd2b puncta promote the elongation of branches and that CaMKII may be a critical mediator of this process. These results demonstrate that Gjd2b-mediated gap junctions regulate glutamatergic synapse formation and dendritic elaboration in PNs.
Collapse
Affiliation(s)
- Sahana Sitaraman
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Gnaneshwar Yadav
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Vandana Agarwal
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Shaista Jabeen
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Shivangi Verma
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Meha Jadhav
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Vatsala Thirumalai
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
13
|
Elsaey MA, Namikawa K, Köster RW. Genetic Modeling of the Neurodegenerative Disease Spinocerebellar Ataxia Type 1 in Zebrafish. Int J Mol Sci 2021; 22:7351. [PMID: 34298970 PMCID: PMC8306488 DOI: 10.3390/ijms22147351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 12/26/2022] Open
Abstract
Dominant spinocerebellar ataxias (SCAs) are progredient neurodegenerative diseases commonly affecting the survival of Purkinje cells (PCs) in the human cerebellum. Spinocerebellar ataxia type 1 (SCA1) is caused by the mutated ataxin1 (Atx1) gene product, in which a polyglutamine stretch encoded by CAG repeats is extended in affected SCA1 patients. As a monogenetic disease with the Atx1-polyQ protein exerting a gain of function, SCA1 can be genetically modelled in animals by cell type-specific overexpression. We have established a transgenic PC-specific SCA1 model in zebrafish coexpressing the fluorescent reporter protein mScarlet together with either human wild type Atx1[30Q] as control or SCA1 patient-derived Atx1[82Q]. SCA1 zebrafish display an age-dependent PC degeneration starting at larval stages around six weeks postfertilization, which continuously progresses during further juvenile and young adult stages. Interestingly, PC degeneration is observed more severely in rostral than in caudal regions of the PC population. Although such a neuropathology resulted in no gross locomotor control deficits, SCA1-fish with advanced PC loss display a reduced exploratory behaviour. In vivo imaging in this SCA1 model may help to better understand such patterned PC death known from PC neurodegeneration diseases, to elucidate disease mechanisms and to provide access to neuroprotective compound characterization in vivo.
Collapse
Affiliation(s)
- Mohamed A. Elsaey
- Division of Cellular & Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany;
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Kazuhiko Namikawa
- Division of Cellular & Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany;
| | - Reinhard W. Köster
- Division of Cellular & Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Spielmannstraße 7, 38106 Braunschweig, Germany;
| |
Collapse
|
14
|
Involvement of Cerebellar Neural Circuits in Active Avoidance Conditioning in Zebrafish. eNeuro 2021; 8:ENEURO.0507-20.2021. [PMID: 33952613 PMCID: PMC8184220 DOI: 10.1523/eneuro.0507-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/20/2021] [Accepted: 03/28/2021] [Indexed: 12/15/2022] Open
Abstract
When animals repeatedly receive a combination of neutral conditional stimulus (CS) and aversive unconditional stimulus (US), they learn the relationship between CS and US, and show conditioned fear responses after CS. They show passive responses such as freezing or panic movements (classical or Pavlovian fear conditioning), or active behavioral responses to avoid aversive stimuli (active avoidance). Previous studies suggested the roles of the cerebellum in classical fear conditioning but it remains elusive whether the cerebellum is involved in active avoidance conditioning. In this study, we analyzed the roles of cerebellar neural circuits during active avoidance in adult zebrafish. When pairs of CS (light) and US (electric shock) were administered to wild-type zebrafish, about half of them displayed active avoidance. The expression of botulinum toxin, which inhibits the release of neurotransmitters, in cerebellar granule cells (GCs) or Purkinje cells (PCs) did not affect conditioning-independent swimming behaviors, but did inhibit active avoidance conditioning. Nitroreductase (NTR)-mediated ablation of PCs in adult zebrafish also impaired active avoidance. Furthermore, the inhibited transmission of GCs or PCs resulted in reduced fear-conditioned Pavlovian fear responses. Our findings suggest that the zebrafish cerebellum plays an active role in active avoidance conditioning.
Collapse
|
15
|
Zebrafish as an animal model for biomedical research. Exp Mol Med 2021; 53:310-317. [PMID: 33649498 PMCID: PMC8080808 DOI: 10.1038/s12276-021-00571-5] [Citation(s) in RCA: 214] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Zebrafish have several advantages compared to other vertebrate models used in modeling human diseases, particularly for large-scale genetic mutant and therapeutic compound screenings, and other biomedical research applications. With the impactful developments of CRISPR and next-generation sequencing technology, disease modeling in zebrafish is accelerating the understanding of the molecular mechanisms of human genetic diseases. These efforts are fundamental for the future of precision medicine because they provide new diagnostic and therapeutic solutions. This review focuses on zebrafish disease models for biomedical research, mainly in developmental disorders, mental disorders, and metabolic diseases.
Collapse
|
16
|
Alexander CJ, Barzik M, Fujiwara I, Remmert K, Wang YX, Petralia RS, Friedman TB, Hammer JA. Myosin 18Aα targets the guanine nucleotide exchange factor β-Pix to the dendritic spines of cerebellar Purkinje neurons and promotes spine maturation. FASEB J 2021; 35:e21092. [PMID: 33378124 PMCID: PMC8357457 DOI: 10.1096/fj.202001449r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/24/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is β-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, β-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with β-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of β-Pix's GEF activity.
Collapse
Affiliation(s)
- Christopher J Alexander
- Molecular Cell Biology Laboratory, Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Melanie Barzik
- Laboratory of Molecular Genetics, NIDCD, NIH, Bethesda, MD, USA
| | - Ikuko Fujiwara
- Graduate School of Science, Osaka City University, Osaka, Japan
| | | | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD, NIH, Betheda, MD, USA
| | | | | | - John A Hammer
- Molecular Cell Biology Laboratory, Cell and Developmental Biology Center, NHLBI, NIH, Bethesda, MD, USA
| |
Collapse
|
17
|
Wang J, Fourriere L, Gleeson PA. Local Secretory Trafficking Pathways in Neurons and the Role of Dendritic Golgi Outposts in Different Cell Models. Front Mol Neurosci 2020; 13:597391. [PMID: 33324160 PMCID: PMC7726432 DOI: 10.3389/fnmol.2020.597391] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/18/2022] Open
Abstract
A fundamental characteristic of neurons is the relationship between the architecture of the polarized neuron and synaptic transmission between neurons. Intracellular membrane trafficking is paramount to establish and maintain neuronal structure; perturbation in trafficking results in defects in neurodevelopment and neurological disorders. Given the physical distance from the cell body to the distal sites of the axon and dendrites, transport of newly synthesized membrane proteins from the central cell body to their functional destination at remote, distal sites represents a conundrum. With the identification of secretory organelles in dendrites, including endoplasmic reticulum (ER) and Golgi outposts (GOs), recent studies have proposed local protein synthesis and trafficking distinct from the conventional anterograde transport pathways of the cell body. A variety of different model organisms, including Drosophila, zebrafish, and rodents, have been used to probe the organization and function of the local neuronal secretory network. Here, we review the evidence for local secretory trafficking pathways in dendrites in a variety of cell-based neuronal systems and discuss both the similarities and differences in the organization and role of the local secretory organelles, especially the GOs. In addition, we identify the gaps in the current knowledge and the potential advances using human induced pluripotent stem cells (iPSCs) in defining local membrane protein trafficking in human neurons and in understanding the molecular basis of neurological diseases.
Collapse
Affiliation(s)
- Jingqi Wang
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Lou Fourriere
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul A Gleeson
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Hsieh JY, Ulrich BN, Issa FA, Lin MCA, Brown B, Papazian DM. Infant and adult SCA13 mutations differentially affect Purkinje cell excitability, maturation, and viability in vivo. eLife 2020; 9:57358. [PMID: 32644043 PMCID: PMC7386905 DOI: 10.7554/elife.57358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022] Open
Abstract
Mutations in KCNC3, which encodes the Kv3.3 K+ channel, cause spinocerebellar ataxia 13 (SCA13). SCA13 exists in distinct forms with onset in infancy or adulthood. Using zebrafish, we tested the hypothesis that infant- and adult-onset mutations differentially affect the excitability and viability of Purkinje cells in vivo during cerebellar development. An infant-onset mutation dramatically and transiently increased Purkinje cell excitability, stunted process extension, impaired dendritic branching and synaptogenesis, and caused rapid cell death during cerebellar development. Reducing excitability increased early Purkinje cell survival. In contrast, an adult-onset mutation did not significantly alter basal tonic firing in Purkinje cells, but reduced excitability during evoked high frequency spiking. Purkinje cells expressing the adult-onset mutation matured normally and did not degenerate during cerebellar development. Our results suggest that differential changes in the excitability of cerebellar neurons contribute to the distinct ages of onset and timing of cerebellar degeneration in infant- and adult-onset SCA13.
Collapse
Affiliation(s)
- Jui-Yi Hsieh
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brittany N Ulrich
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Fadi A Issa
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Meng-Chin A Lin
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brandon Brown
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Brain Research Institute, UCLA, Los Angeles, United States.,Molecular Biology Institute, UCLA, Los Angeles, United States
| |
Collapse
|
19
|
Functionally distinct Purkinje cell types show temporal precision in encoding locomotion. Proc Natl Acad Sci U S A 2020; 117:17330-17337. [PMID: 32632015 PMCID: PMC7382291 DOI: 10.1073/pnas.2005633117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Purkinje cells, the principal neurons of cerebellar computations, are believed to comprise a uniform neuronal population of cells, each with similar functional properties. Here, we show an undiscovered heterogeneity of adult zebrafish Purkinje cells, revealing the existence of anatomically and functionally distinct cell types. Dual patch-clamp recordings showed that the cerebellar circuit contains all Purkinje cell types that cross-communicate extensively using chemical and electrical synapses. Further activation of spinal central pattern generators (CPGs) revealed unique phase-locked activity from each Purkinje cell type during the locomotor cycle. Thus, we show intricately organized Purkinje cell networks in the adult zebrafish cerebellum that encode the locomotion rhythm differentially, and we suggest that these organizational properties may also apply to other cerebellar functions.
Collapse
|
20
|
Ligon C, Cai Y, Buch S, Arikkath J. A selective role for a component of the autophagy pathway in coupling the Golgi apparatus to dendrite polarity in pyramidal neurons. Neurosci Lett 2020; 730:135048. [PMID: 32439477 DOI: 10.1016/j.neulet.2020.135048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022]
Abstract
Pyramidal neurons have a characteristic morphology that is critical to their ability to integrate into functional neural circuits. In addition to axon dendrite polarity, pyramidal neurons also exhibit dendritic polarity such that apical and basolateral dendrites differ in size, structure and inputs. Dendrite polarity in pyramidal neurons coincides with polarity of the Golgi apparatus, a key feature relevant to directed secretory trafficking, both in vitro and in vivo. We identify a novel autophagy based mechanism that uncouples the polarity of the Golgi apparatus from dendrite polarity. Autophagy is a universal cellular pathway that promotes cellular homeostasis via degradation of cellular components. Our data indicate that knockdown of ATG7, a key component of the autophagy mechanism, disrupts the polarity of the Golgi apparatus without impacting dendritic polarity in primary pyramidal neurons, providing the first evidence that dendrite polarity can be uncoupled from Golgi polarity. Interestingly, these effects are restricted to ATG7 knockdown and are not replicated by the knockdown of ATG16L1, another component of the autophagy mechanism. We propose that cellular mechanisms exist to couple Golgi polarity to dendrite polarity. Components of the autophagy mechanism are leveraged to actively couple Golgi polarity to dendrite polarity, thus impacting secretory trafficking into individual dendrites in pyramidal neurons.
Collapse
Affiliation(s)
- Cheryl Ligon
- Developmental Neuroscience, Munroe-Meyer Institute, United States
| | - Yu Cai
- Department of Pharmacology and Experimental Neuroscience University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jyothi Arikkath
- Department of Anatomy, Howard University, Washington D.C, 20059, United States.
| |
Collapse
|
21
|
Velocity storage mechanism drives a cerebellar clock for predictive eye velocity control. Sci Rep 2020; 10:6944. [PMID: 32332917 PMCID: PMC7181809 DOI: 10.1038/s41598-020-63641-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/30/2020] [Indexed: 01/07/2023] Open
Abstract
Predictive motor control is ubiquitously employed in animal kingdom to achieve rapid and precise motor action. In most vertebrates large, moving visual scenes induce an optokinetic response (OKR) control of eye movements to stabilize vision. In goldfish, the OKR was found to be predictive after a prolonged exposure to temporally periodic visual motion. A recent study showed the cerebellum necessary to acquire this predictive OKR (pOKR), but it remained unclear as to whether the cerebellum alone was sufficient. Herein we examined different fish species known to share the basic architecture of cerebellar neuronal circuitry for their ability to acquire pOKR. Carps were shown to acquire pOKR like goldfish while zebrafish and medaka did not, demonstrating the cerebellum alone not to be sufficient. Interestingly, those fish that acquired pOKR were found to exhibit long-lasting optokinetic after nystagmus (OKAN) as opposed to those that didn’t. To directly manipulate OKAN vestibular-neurectomy was performed in goldfish that severely shortened OKAN, but pOKR was acquired comparable to normal animals. These results suggest that the neuronal circuitry producing OKAN, known as the velocity storage mechanism (VSM), is required to acquire pOKR irrespective of OKAN duration. Taken together, we conclude that pOKR is acquired through recurrent cerebellum-brainstem parallel loops in which the cerebellum adjusts VSM signal flow and, in turn, receives appropriately timed eye velocity information to clock visual world motion.
Collapse
|
22
|
Genetic compensation in a stable slc25a46 mutant zebrafish: A case for using F0 CRISPR mutagenesis to study phenotypes caused by inherited disease. PLoS One 2020; 15:e0230566. [PMID: 32208444 PMCID: PMC7092968 DOI: 10.1371/journal.pone.0230566] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/03/2020] [Indexed: 12/22/2022] Open
Abstract
A phenomenon of genetic compensation is commonly observed when an organism with a disease-bearing mutation shows incomplete penetrance of the disease phenotype. Such incomplete phenotypic penetrance, or genetic compensation, is more commonly found in stable knockout models, rather than transient knockdown models. As such, these incidents present a challenge for the disease modeling field, although a deeper understanding of genetic compensation may also hold the key for novel therapeutic interventions. In our study we created a knockout model of slc25a46 gene, which is a recently discovered important player in mitochondrial dynamics, and deleterious mutations in which are known to cause peripheral neuropathy, optic atrophy and cerebellar ataxia. We report a case of genetic compensation in a stable slc25a46 homozygous zebrafish mutant (hereafter referred as “mutant”), in contrast to a penetrant disease phenotype in the first generation (F0) slc25a46 mosaic mutant (hereafter referred as “crispant”), generated with CRISPR/Cas-9 technology. We show that the crispant phenotype is specific and rescuable. By performing mRNA sequencing, we define significant changes in slc25a46 mutant’s gene expression profile, which are largely absent in crispants. We find that among the most significantly altered mRNAs, anxa6 gene stands out as a functionally relevant player in mitochondrial dynamics. We also find that our genetic compensation case does not arise from mechanisms driven by mutant mRNA decay. Our study contributes to the growing evidence of the genetic compensation phenomenon and presents novel insights about Slc25a46 function. Furthermore, our study provides the evidence for the efficiency of F0 CRISPR screens for disease candidate genes, which may be used to advance the field of functional genetics.
Collapse
|
23
|
Simeoli R, Fierabracci A. Insights into the Role of MicroRNAs in the Onset and Development of Diabetic Neuropathy. Int J Mol Sci 2019; 20:ijms20184627. [PMID: 31540445 PMCID: PMC6770207 DOI: 10.3390/ijms20184627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/30/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetic neuropathy is a serious complication of chronic hyperglycemia in diabetes patients. This complication can involve both peripheral sensorimotor and autonomic nervous system. The precise nature of injury to the peripheral nerves mediated by chronic hyperglycemia is unknown; however, several mechanisms have been proposed including polyol pathway activation, enhanced glycation of proteins and lipids, increased oxidative stress, and cytokine release in the site of injury. MicroRNAs (miRNAs) are small non-coding RNAs that mediate RNA interference by post-transcriptionally modulating gene expression and protein synthesis. Therefore, they have been implicated in several developmental, physiological, and pathophysiological processes where they modulate the expression of different proteins. Recently, miRNAs gained an increasing attention also for their role as diagnostic test in many diseases due to their stability in serum and their easy detection. Furthermore, recent studies suggest that miRNAs may be involved in diabetic neuropathy although their role in the onset and the development of this complication is not fully understood. In this review, we discuss the most recent literature providing evidence for miRNAs role in diabetic neuropathy opening new pathways to improve both early diagnosis and treatment of this complication.
Collapse
Affiliation(s)
- Raffaele Simeoli
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
24
|
Chau DDL, Yung KWY, Chan WWL, An Y, Hao Y, Chan HYE, Ngo JCK, Lau KF. Attenuation of amyloid-β generation by atypical protein kinase C-mediated phosphorylation of engulfment adaptor PTB domain containing 1 threonine 35. FASEB J 2019; 33:12019-12035. [PMID: 31373844 DOI: 10.1096/fj.201802825rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Amyloid-β (Aβ) is derived from the proteolytic processing of amyloid precursor protein (APP), and the deposition of extracellular Aβ to form amyloid plaques is a pathologic hallmark of Alzheimer's disease (AD). Although reducing Aβ generation and accumulation has been proposed as a means of treating the disease, adverse side effects and unsatisfactory efficacy have been reported in several clinical trials that sought to lower Aβ levels. Engulfment adaptor phosphotyrosine-binding (PTB) domain containing 1 (GULP1) is a molecular adaptor that has been shown to interact with APP to alter Aβ production. Therefore, the modulation of the GULP1-APP interaction may be an alternative approach to reducing Aβ. However, the mechanisms that regulate GULP1-APP binding remain elusive. As GULP1 is a phosphoprotein, and because phosphorylation is a common mechanism that regulates protein interaction, we anticipated that GULP1 phosphorylation would influence GULP1-APP interaction and thereby Aβ production. We show here that the phosphorylation of GULP1 threonine 35 (T35) reduces GULP1-APP interaction and suppresses the stimulatory effect of GULP1 on APP processing. The residue is phosphorylated by an isoform of atypical PKC (PKCζ). Overexpression of PKCζ reduces both GULP1-APP interaction and GULP1-mediated Aβ generation. Moreover, the activation of PKCζ via insulin suppresses APP processing. In contrast, GULP1-mediated APP processing is enhanced in PKCζ knockout cells. Similarly, PKC ι, another member of atypical PKC, also decreases GULP1-mediated APP processing. Intriguingly, our X-ray crystal structure of GULP1 PTB-APP intracellular domain (AICD) peptide reveals that GULP1 T35 is not located at the GULP1-AICD binding interface; rather, it immediately precedes the β1-α2 loop that forms a portion of the binding groove for the APP helix αC. Phosphorylating the residue may induce an allosteric effect on the conformation of the binding groove. Our results indicate that GULP1 T35 phosphorylation is a mechanism for the regulation of GULP1-APP interaction and thereby APP processing. Moreover, the activation of atypical PKC, such as by insulin, may confer a beneficial effect on AD by lowering GULP1-mediated Aβ production.-Chau, D. D.-L., Yung, K. W.-Y., Chan, W. W.-L., An, Y., Hao, Y., Chan, H.-Y. E., Ngo, J. C.-K., Lau, K.-F. Attenuation of amyloid-β generation by atypical protein kinase C-mediated phosphorylation of engulfment adaptor PTB domain containing 1 threonine 35.
Collapse
Affiliation(s)
- Dennis Dik-Long Chau
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kristen Wing-Yu Yung
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - William Wai-Lun Chan
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ying An
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Hao
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ho-Yin Edwin Chan
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jacky Chi-Ki Ngo
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok-Fai Lau
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
25
|
Nimura T, Itoh T, Hagio H, Hayashi T, Di Donato V, Takeuchi M, Itoh T, Inoguchi F, Sato Y, Yamamoto N, Katsuyama Y, Del Bene F, Shimizu T, Hibi M. Role of Reelin in cell positioning in the cerebellum and the cerebellum-like structure in zebrafish. Dev Biol 2019; 455:393-408. [PMID: 31323192 DOI: 10.1016/j.ydbio.2019.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/05/2019] [Accepted: 07/14/2019] [Indexed: 02/07/2023]
Abstract
The cerebellum and the cerebellum-like structure in the mesencephalic tectum in zebrafish contain multiple cell types, including principal cells (i.e., Purkinje cells and type I neurons) and granule cells, that form neural circuits in which the principal cells receive and integrate inputs from granule cells and other neurons. It is largely unknown how these cells are positioned and how neural circuits form. While Reelin signaling is known to play an important role in cell positioning in the mammalian brain, its role in the formation of other vertebrate brains remains elusive. Here we found that zebrafish with mutations in Reelin or in the Reelin-signaling molecules Vldlr or Dab1a exhibited ectopic Purkinje cells, eurydendroid cells (projection neurons), and Bergmann glial cells in the cerebellum, and ectopic type I neurons in the tectum. The ectopic Purkinje cells and type I neurons received aberrant afferent fibers in these mutants. In wild-type zebrafish, reelin transcripts were detected in the internal granule cell layer, while Reelin protein was localized to the superficial layer of the cerebellum and the tectum. Laser ablation of the granule cell axons perturbed the localization of Reelin, and the mutation of both kif5aa and kif5ba, which encode major kinesin I components in the granule cells, disrupted the elongation of granule cell axons and the Reelin distribution. Our findings suggest that in zebrafish, (1) Reelin is transported from the granule cell soma to the superficial layer by axonal transport; (2) Reelin controls the migration of neurons and glial cells from the ventricular zone; and (3) Purkinje cells and type I neurons attract afferent axons during the formation of the cerebellum and the cerebellum-like structure.
Collapse
Affiliation(s)
- Takayuki Nimura
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan
| | - Tsubasa Itoh
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan
| | - Hanako Hagio
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan; Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Takuto Hayashi
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan
| | - Vincenzo Di Donato
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, UPMC Paris-Sorbonne, Paris, 75005, France
| | - Miki Takeuchi
- Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8601, Japan
| | - Takeaki Itoh
- Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Fuduki Inoguchi
- Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Yoshikatsu Sato
- Institute of Transformative Bio-Molecules, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8601, Japan
| | - Naoyuki Yamamoto
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Filippo Del Bene
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, UPMC Paris-Sorbonne, Paris, 75005, France
| | - Takashi Shimizu
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan; Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8601, Japan
| | - Masahiko Hibi
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8602, Japan; Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
26
|
A genetically encoded fluorescent sensor for in vivo imaging of GABA. Nat Methods 2019; 16:763-770. [DOI: 10.1038/s41592-019-0471-2] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/28/2019] [Indexed: 12/13/2022]
|
27
|
Lowe M. The Physiological Functions of the Golgin Vesicle Tethering Proteins. Front Cell Dev Biol 2019; 7:94. [PMID: 31316978 PMCID: PMC6611411 DOI: 10.3389/fcell.2019.00094] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/16/2019] [Indexed: 01/02/2023] Open
Abstract
The golgins comprise a family of vesicle tethering proteins that act in a selective manner to tether transport vesicles at the Golgi apparatus. Tethering is followed by membrane fusion to complete the delivery of vesicle-bound cargo to the Golgi. Different golgins are localized to different regions of the Golgi, and their ability to selectively tether transport vesicles is important for the specificity of vesicle traffic in the secretory pathway. In recent years, our mechanistic understanding of golgin-mediated tethering has greatly improved. We are also beginning to appreciate how the loss of golgin function can impact upon physiological processes through the use of animal models and the study of human disease. These approaches have revealed that loss of a golgin causes tissue-restricted phenotypes, which can vary in severity and the cell types affected. In many cases, it is possible to attribute these phenotypes to a defect in vesicular traffic, although why certain tissues are sensitive to loss of a particular golgin is still, in most cases, unclear. Here, I will summarize recent progress in our understanding of golgins, focusing on the physiological roles of these proteins, as determined from animal models and the study of disease in humans. I will describe what these in vivo analyses have taught us, as well as highlight less understood aspects, and areas for future investigations.
Collapse
Affiliation(s)
- Martin Lowe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
28
|
Namikawa K, Dorigo A, Zagrebelsky M, Russo G, Kirmann T, Fahr W, Dübel S, Korte M, Köster RW. Modeling Neurodegenerative Spinocerebellar Ataxia Type 13 in Zebrafish Using a Purkinje Neuron Specific Tunable Coexpression System. J Neurosci 2019; 39:3948-3969. [PMID: 30862666 PMCID: PMC6520513 DOI: 10.1523/jneurosci.1862-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Purkinje cells (PCs) are primarily affected in neurodegenerative spinocerebellar ataxias (SCAs). For generating animal models for SCAs, genetic regulatory elements specifically targeting PCs are required, thereby linking pathological molecular effects with impaired function and organismic behavior. Because cerebellar anatomy and function are evolutionary conserved, zebrafish represent an excellent model to study SCAs in vivo We have isolated a 258 bp cross-species PC-specific enhancer element that can be used in a bidirectional manner for bioimaging of transgene-expressing PCs in zebrafish (both sexes) with variable copy numbers for tuning expression strength. Emerging ectopic expression at high copy numbers can be further eliminated by repurposing microRNA-mediated posttranslational mRNA regulation.Subsequently, we generated a transgenic SCA type 13 (SCA13) model, using a zebrafish-variant mimicking a human pathological SCA13R420H mutation, resulting in cell-autonomous progressive PC degeneration linked to cerebellum-driven eye-movement deficits as observed in SCA patients. This underscores that investigating PC-specific cerebellar neuropathologies in zebrafish allows for interconnecting bioimaging of disease mechanisms with behavioral analysis suitable for therapeutic compound testing.SIGNIFICANCE STATEMENT SCA13 patients carrying a KCNC3R420H allele have been shown to display mid-onset progressive cerebellar atrophy, but genetic modeling of SCA13 by expressing this pathogenic mutant in different animal models has not resulted in neuronal degeneration so far; likely because the transgene was expressed in heterologous cell types. We developed a genetic system for tunable PC-specific coexpression of several transgenes to manipulate and simultaneously monitor cerebellar PCs. We modeled a SCA13 zebrafish accessible for bioimaging to investigate disease progression, revealing robust PC degeneration, resulting in impaired eye movement. Our transgenic zebrafish mimicking both neuropathological and behavioral changes manifested in SCA-affected patients will be suitable for investigating causes of cerebellar diseases in vivo from the molecular to the behavioral level.
Collapse
Affiliation(s)
| | | | - Marta Zagrebelsky
- Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig 38106, Germany
| | - Giulio Russo
- Cellular and Molecular Neurobiology
- Biotechnology and Bioinformatics, Institute for Biochemistry, Technical University Braunschweig 38106, Germany, and
| | | | - Wieland Fahr
- Biotechnology and Bioinformatics, Institute for Biochemistry, Technical University Braunschweig 38106, Germany, and
| | - Stefan Dübel
- Biotechnology and Bioinformatics, Institute for Biochemistry, Technical University Braunschweig 38106, Germany, and
| | - Martin Korte
- Cellular Neurobiology, Zoological Institute, Technical University Braunschweig, Braunschweig 38106, Germany
- Research Group Neuroinflammation and Neurodegeneration, Helmholtz Centre for Infection Research, Braunschweig 38106, Germany
| | | |
Collapse
|
29
|
Dohaku R, Yamaguchi M, Yamamoto N, Shimizu T, Osakada F, Hibi M. Tracing of Afferent Connections in the Zebrafish Cerebellum Using Recombinant Rabies Virus. Front Neural Circuits 2019; 13:30. [PMID: 31068795 PMCID: PMC6491863 DOI: 10.3389/fncir.2019.00030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/10/2019] [Indexed: 12/31/2022] Open
Abstract
The cerebellum is involved in some forms of motor coordination and learning, and in cognitive and emotional functions. To elucidate the functions of the cerebellum, it is important to unravel the detailed connections of the cerebellar neurons. Although the cerebellar neural circuit structure is generally conserved among vertebrates, it is not clear whether the cerebellum receives and processes the same or similar information in different vertebrate species. Here, we performed monosynaptic retrograde tracing with recombinant rabies viruses (RV) to identify the afferent connections of the zebrafish cerebellar neurons. We used a G-deleted RV that expressed GFP. The virus was also pseudotyped with EnvA, an envelope protein of avian sarcoma and leucosis virus (ALSV-A). For the specific infection of cerebellar neurons, we expressed the RV glycoprotein (G) gene and the envelope protein TVA, which is the receptor for EnvA, in Purkinje cells (PCs) or granule cells (GCs), using the promoter for aldolase Ca (aldoca) or cerebellin 12 (cbln12), respectively. When the virus infected PCs in the aldoca line, GFP was detected in the PCs’ presynaptic neurons, including GCs and neurons in the inferior olivary nuclei (IOs), which send climbing fibers (CFs). These observations validated the RV tracing method in zebrafish. When the virus infected GCs in the cbln12 line, GFP was again detected in their presynaptic neurons, including neurons in the pretectal nuclei, the nucleus lateralis valvulae (NLV), the central gray (CG), the medial octavolateralis nucleus (MON), and the descending octaval nucleus (DON). GFP was not observed in these neurons when the virus infected PCs in the aldoca line. These precerebellar neurons generally agree with those reported for other teleost species and are at least partly conserved with those in mammals. Our results demonstrate that the RV system can be used for connectome analyses in zebrafish, and provide fundamental information about the cerebellar neural circuits, which will be valuable for elucidating the functions of cerebellar neural circuits in zebrafish.
Collapse
Affiliation(s)
- Ryuji Dohaku
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masahiro Yamaguchi
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Naoyuki Yamamoto
- Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Takashi Shimizu
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.,Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology, Nagoya University, Nagoya, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Masahiko Hibi
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan.,Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology, Nagoya University, Nagoya, Japan
| |
Collapse
|
30
|
Dendritic Self-Avoidance and Morphological Development of Cerebellar Purkinje Cells. THE CEREBELLUM 2018; 17:701-708. [DOI: 10.1007/s12311-018-0984-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
31
|
Hapak SM, Rothlin CV, Ghosh S. PAR3-PAR6-atypical PKC polarity complex proteins in neuronal polarization. Cell Mol Life Sci 2018; 75:2735-2761. [PMID: 29696344 PMCID: PMC11105418 DOI: 10.1007/s00018-018-2828-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/03/2018] [Accepted: 04/23/2018] [Indexed: 01/01/2023]
Abstract
Polarity is a fundamental feature of cells. Protein complexes, including the PAR3-PAR6-aPKC complex, have conserved roles in establishing polarity across a number of eukaryotic cell types. In neurons, polarity is evident as distinct axonal versus dendritic domains. The PAR3, PAR6, and aPKC proteins also play important roles in neuronal polarization. During this process, either aPKC kinase activity, the assembly of the PAR3-PAR6-aPKC complex or the localization of these proteins is regulated downstream of a number of signaling pathways. In turn, the PAR3, PAR6, and aPKC proteins control various effector molecules to establish neuronal polarity. Herein, we discuss the many signaling mechanisms and effector functions that have been linked to PAR3, PAR6, and aPKC during the establishment of neuronal polarity.
Collapse
Affiliation(s)
- Sophie M Hapak
- Department of Medicine, School of Medicine, University of Minnesota, 401 East River Parkway, Minneapolis, MN, 55455, USA.
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, 300 Cedar Street, New Haven, CT, 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, 300 George Street, New Haven, CT, 06511, USA
- Department of Pharmacology, School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
32
|
In Vivo Analysis of Potassium Channelopathies: Loose Patch Recording of Purkinje Cell Firing in Living, Awake Zebrafish. Methods Mol Biol 2018; 1684:237-252. [PMID: 29058196 DOI: 10.1007/978-1-4939-7362-0_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Zebrafish is a lower vertebrate model organism that facilitates integrative analysis of the in vivo effects of potassium and other ion channel mutations at the molecular, cellular, developmental, circuit, systems, and behavioral levels of analysis. Here, we describe a method for extracellular, loose patch electrophysiological recording of electrical activity in cerebellar Purkinje cells in living, awake zebrafish, with the goal of investigating pathological mechanisms underlying channelopathies or other diseases that disrupt cerebellar function. Purkinje cell excitability and a functional cerebellar circuit develop rapidly in zebrafish and show strong conservation with the mammalian cerebellum.
Collapse
|
33
|
Miller GW, Chandrasekaran V, Yaghoobi B, Lein PJ. Opportunities and challenges for using the zebrafish to study neuronal connectivity as an endpoint of developmental neurotoxicity. Neurotoxicology 2018; 67:102-111. [PMID: 29704525 PMCID: PMC6177215 DOI: 10.1016/j.neuro.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 01/28/2023]
Abstract
Chemical exposures have been implicated as environmental risk factors that interact with genetic susceptibilities to influence individual risk for complex neurodevelopmental disorders, including autism spectrum disorder, schizophrenia, attention deficit hyperactivity disorder and intellectual disabilities. Altered patterns of neuronal connectivity represent a convergent mechanism of pathogenesis for these and other neurodevelopmental disorders, and growing evidence suggests that chemicals can interfere with specific signaling pathways that regulate the development of neuronal connections. There is, therefore, a growing interest in developing screening platforms to identify chemicals that alter neuronal connectivity. Cell-cell, cell-matrix interactions and systemic influences are known to be important in defining neuronal connectivity in the developing brain, thus, a systems-based model offers significant advantages over cell-based models for screening chemicals for effects on neuronal connectivity. The embryonic zebrafish represents a vertebrate model amenable to higher throughput chemical screening that has proven useful in characterizing conserved mechanisms of neurodevelopment. Moreover, the zebrafish is readily amenable to gene editing to integrate genetic susceptibilities. Although use of the zebrafish model in toxicity testing has increased in recent years, the diverse tools available for imaging structural differences in the developing zebrafish brain have not been widely applied to studies of the influence of gene by environment interactions on neuronal connectivity in the developing zebrafish brain. Here, we discuss tools available for imaging of neuronal connectivity in the developing zebrafish, review what has been published in this regard, and suggest a path forward for applying this information to developmental neurotoxicity testing.
Collapse
Affiliation(s)
- Galen W. Miller
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary’s College of California, Moraga, CA 94575, USA
| | - Bianca Yaghoobi
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
34
|
Jia L, Wang L, Chopp M, Li C, Zhang Y, Szalad A, Zhang ZG. MiR-29c/PRKCI Regulates Axonal Growth of Dorsal Root Ganglia Neurons Under Hyperglycemia. Mol Neurobiol 2018; 55:851-858. [PMID: 28070856 PMCID: PMC5577385 DOI: 10.1007/s12035-016-0374-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022]
Abstract
Diabetes initially induces distal axonal damage of peripheral nerves, but molecular mechanisms that mediate axonal injury are not fully understood. MircoRNAs (miRNAs) regulate axonal growth. We found that diabetic db/db mice exhibited substantial upregulation of miR-29c in dorsal root ganglia (DRG) neurons, sciatic nerve, and foot pad tissues. Bioinformatic analysis revealed PRKCI, a gene that encodes a member of the protein kinase C (PKC) iota, as a putative target for miR-29c. Western blot analysis showed that diabetic mice exhibited a considerable reduction of PRKCI protein levels in sciatic nerve tissues and DRG neurons. Using dual-luciferase assay, we found that co-transfection of a plasmid containing miR-29c binding site at 3' UTR of PRKCI gene and miR-29c mimics effectively reduced luminescence activity, which was abolished when miR-29c seed sequences at 3' UTR of PRKCI gene were mutated. In vitro, high glucose substantially upregulated and reduced miR-29c and PRKCI protein levels, respectively, in DRG neurons, which were associated with significant reduction of axonal growth. Knockdown of endogenous miR-29c in DRG neurons by siRNAs overcame reduced PRKCI protein and axonal growth under high glucose condition. Moreover, knockdown of PRKCI in DRG neurons by siRNAs under regular glucose condition considerably inhibited axonal growth. Together, these findings suggest that miR-29c is a negative regulator of axonal growth of DRG neurons by targeting PRKCI under hyperglycemia.
Collapse
Affiliation(s)
- Longfei Jia
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Lei Wang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
- Department of Physics Oakland University, Rochester, MI, 48309, USA
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| |
Collapse
|
35
|
Repositioning of Somatic Golgi Apparatus Is Essential for the Dendritic Establishment of Adult-Born Hippocampal Neurons. J Neurosci 2017; 38:631-647. [PMID: 29217690 DOI: 10.1523/jneurosci.1217-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/02/2017] [Accepted: 10/29/2017] [Indexed: 01/22/2023] Open
Abstract
New dentate granule cells (DGCs) are continuously generated, and integrate into the preexisting hippocampal network in the adult brain. How an adult-born neuron with initially simple spindle-like morphology develops into a DGC, consisting of a single apical dendrite with further branches, remains largely unknown. Here, using retroviruses to birth date and manipulate newborn neurons, we examined initial dendritic formation and possible underlying mechanisms. We found that GFP-expressing newborn cells began to establish a DGC-like morphology at ∼7 d after birth, with a primary dendrite pointing to the molecular layer, but at this stage, with several neurites in the neurogenic zone. Interestingly, the Golgi apparatus, an essential organelle for neurite growth and maintenance, was dynamically repositioning in the soma of newborn cells during this initial integration stage. Two weeks after birth, by which time most neurites in the neurogenic zone were eliminated, a compact Golgi apparatus was positioned exclusively at the base of the primary dendrite. We analyzed the presence of Golgi-associated genes using single-cell transcriptomes of newborn DGCs, and among Golgi-related genes, found the presence of STK25 and STRAD, regulators of embryonic neuronal development. When we knocked down either of these two proteins, we found Golgi mislocalization and extensive aberrant dendrite formation. Furthermore, overexpression of a mutated form of STRAD, underlying the disorder polyhydramnios, megalencephaly, and symptomatic epilepsy, characterized by abnormal brain development and intractable epilepsy, caused similar defects in Golgi localization and dendrite formation in adult-born neurons. Together, our findings reveal a role for Golgi repositioning in regulating the initial integration of adult-born DGCs.SIGNIFICANCE STATEMENT Since the discovery of the continuous generation of new neurons in the adult hippocampus, extensive effort was directed toward understanding the functional contribution of these newborn neurons to the existing hippocampal circuit and associated behaviors, while the molecular mechanisms controlling their early morphological integration are less well understood. Dentate granule cells (DGCs) have a single, complex, apical dendrite. The events leading adult-born DGCs' to transition from simple spindle-like morphology to mature dendrite morphology are largely unknown. We studied establishment of newborn DGCs dendritic pattern and found it was mediated by a signaling pathway regulating precise localization of the Golgi apparatus. Furthermore, this Golgi-associated mechanism for dendrite establishment might be impaired in a human genetic epilepsy syndrome, polyhydramnios, megalencephaly, and symptomatic epilepsy.
Collapse
|
36
|
Takeuchi M, Inoue C, Goshima A, Nagao Y, Shimizu K, Miyamoto H, Shimizu T, Hashimoto H, Yonemura S, Kawahara A, Hirata Y, Yoshida M, Hibi M. Medaka and zebrafishcontactin1mutants as a model for understanding neural circuits for motor coordination. Genes Cells 2017. [DOI: 10.1111/gtc.12509] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Miki Takeuchi
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
| | - Chikako Inoue
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
| | - Akiko Goshima
- Division of Biological Science; Graduate School of Science; Nagoya University; Furo Chikusa Nagoya Aichi 464-8602 Japan
| | - Yusuke Nagao
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
| | - Koichi Shimizu
- Division of Biological Science; Graduate School of Science; Nagoya University; Furo Chikusa Nagoya Aichi 464-8602 Japan
| | - Hiroki Miyamoto
- Department of Computer Science; Chubu University; 1200 Matsumoto Kasugai Aichi 485-8501 Japan
| | - Takashi Shimizu
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
- Division of Biological Science; Graduate School of Science; Nagoya University; Furo Chikusa Nagoya Aichi 464-8602 Japan
| | - Hisashi Hashimoto
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
- Division of Biological Science; Graduate School of Science; Nagoya University; Furo Chikusa Nagoya Aichi 464-8602 Japan
| | - Shigenobu Yonemura
- Department of Cell Biology; Graduate School of Medical Science; Tokushima University; 3-18-15 Kuramoto Tokushima Tokushima 770-8503 Japan
| | - Atsuo Kawahara
- Laboratory for Developmental Biology; Center for Medical Education and Sciences; Graduate School of Medical Science; University of Yamanashi; 1110 Shimokato, Chuo; Yamanashi 409-3898 Japan
| | - Yutaka Hirata
- Department of Computer Science; Chubu University; 1200 Matsumoto Kasugai Aichi 485-8501 Japan
| | - Masayuki Yoshida
- Graduate School of Biosphere Sciences; Hiroshima University; 1-4-4 Kagamiyama Higashihiroshima Hiroshima 739-8528 Japan
| | - Masahiko Hibi
- Laboratory of Organogenesis and Organ Function; Bioscience and Biotechnology Center; Nagoya University; Furo Chikusa Nagoya Aichi 464-8601 Japan
- Division of Biological Science; Graduate School of Science; Nagoya University; Furo Chikusa Nagoya Aichi 464-8602 Japan
| |
Collapse
|
37
|
Takeuchi M, Yamaguchi S, Sakakibara Y, Hayashi T, Matsuda K, Hara Y, Tanegashima C, Shimizu T, Kuraku S, Hibi M. Gene expression profiling of granule cells and Purkinje cells in the zebrafish cerebellum. J Comp Neurol 2016; 525:1558-1585. [DOI: 10.1002/cne.24114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/03/2016] [Accepted: 09/04/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Miki Takeuchi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology; Nagoya University; Nagoya Aichi 464-8601 Japan
| | - Shingo Yamaguchi
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Yoshimasa Sakakibara
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Takuto Hayashi
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Koji Matsuda
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology; Nagoya University; Nagoya Aichi 464-8601 Japan
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Yuichiro Hara
- Phyloinformatics Unit, RIKEN Center for Life Science Technologies; Kobe Hyogo 650-0047 Japan
| | - Chiharu Tanegashima
- Phyloinformatics Unit, RIKEN Center for Life Science Technologies; Kobe Hyogo 650-0047 Japan
| | - Takashi Shimizu
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology; Nagoya University; Nagoya Aichi 464-8601 Japan
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| | - Shigehiro Kuraku
- Phyloinformatics Unit, RIKEN Center for Life Science Technologies; Kobe Hyogo 650-0047 Japan
| | - Masahiko Hibi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology; Nagoya University; Nagoya Aichi 464-8601 Japan
- Division of Biological Science, Graduate School of Science; Nagoya University; Nagoya Aichi 464-8602 Japan
| |
Collapse
|
38
|
Scalise K, Shimizu T, Hibi M, Sawtell NB. Responses of cerebellar Purkinje cells during fictive optomotor behavior in larval zebrafish. J Neurophysiol 2016; 116:2067-2080. [PMID: 27512018 DOI: 10.1152/jn.00042.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 08/03/2016] [Indexed: 12/17/2022] Open
Abstract
Although most studies of the cerebellum have been conducted in mammals, cerebellar circuitry is highly conserved across vertebrates, suggesting that studies of simpler systems may be useful for understanding cerebellar function. The larval zebrafish is particularly promising in this regard because of its accessibility to optical monitoring and manipulations of neural activity. Although several studies suggest that the cerebellum plays a role in behavior at larval stages, little is known about the signals conveyed by particular classes of cerebellar neurons. Here we use electrophysiological recordings to characterize subthreshold, simple spike, and climbing fiber responses in larval zebrafish Purkinje cells in the context of the fictive optomotor response (OMR)-a paradigm in which fish adjust motor output to stabilize their virtual position relative to a visual stimulus. Although visual responses were prominent in Purkinje cells, they lacked the direction or velocity sensitivity that would be expected for controlling the OMR. On the other hand, Purkinje cells exhibited strong responses during fictive swim bouts. Temporal characteristics of these responses are suggestive of a general role for the larval zebrafish cerebellum in controlling swimming. Climbing fibers encoded both visual and motor signals but did not appear to encode signals that could be used to adjust OMR gain, such as retinal slip. Finally, the observation of diverse relationships between simple spikes and climbing fiber responses in individual Purkinje cells highlights the importance of distinguishing between these two types of activity in calcium imaging experiments.
Collapse
Affiliation(s)
- Karina Scalise
- Department of Neuroscience, Columbia University, New York, New York; and
| | - Takashi Shimizu
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, Aichi, Japan
| | - Masahiko Hibi
- Bioscience and Biotechnology Center, Nagoya University, Nagoya, Aichi, Japan
| | | |
Collapse
|
39
|
Engerer P, Plucinska G, Thong R, Trovò L, Paquet D, Godinho L. Imaging Subcellular Structures in the Living Zebrafish Embryo. J Vis Exp 2016:e53456. [PMID: 27078038 DOI: 10.3791/53456] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In vivo imaging provides unprecedented access to the dynamic behavior of cellular and subcellular structures in their natural context. Performing such imaging experiments in higher vertebrates such as mammals generally requires surgical access to the system under study. The optical accessibility of embryonic and larval zebrafish allows such invasive procedures to be circumvented and permits imaging in the intact organism. Indeed the zebrafish is now a well-established model to visualize dynamic cellular behaviors using in vivo microscopy in a wide range of developmental contexts from proliferation to migration and differentiation. A more recent development is the increasing use of zebrafish to study subcellular events including mitochondrial trafficking and centrosome dynamics. The relative ease with which these subcellular structures can be genetically labeled by fluorescent proteins and the use of light microscopy techniques to image them is transforming the zebrafish into an in vivo model of cell biology. Here we describe methods to generate genetic constructs that fluorescently label organelles, highlighting mitochondria and centrosomes as specific examples. We use the bipartite Gal4-UAS system in multiple configurations to restrict expression to specific cell-types and provide protocols to generate transiently expressing and stable transgenic fish. Finally, we provide guidelines for choosing light microscopy methods that are most suitable for imaging subcellular dynamics.
Collapse
Affiliation(s)
- Peter Engerer
- Institute of Neuronal Cell Biology, Technische Universität München;
| | - Gabriela Plucinska
- Institute of Neuronal Cell Biology, Technische Universität München; Cell Biology, Department of Biology, Faculty of Science, Utrecht University
| | - Rachel Thong
- Institute of Neuronal Cell Biology, Technische Universität München; Faculty of Biology, Ludwig-Maximilians-Universität-München
| | - Laura Trovò
- Institute of Neuronal Cell Biology, Technische Universität München
| | - Dominik Paquet
- Adolf-Butenandt-Institute, Biochemistry, Ludwig-Maximilians-Universität-München; German Center for Neurodegenerative Diseases; Laboratory of Brain Development and Repair, The Rockefeller University
| | - Leanne Godinho
- Institute of Neuronal Cell Biology, Technische Universität München;
| |
Collapse
|
40
|
Park JY, Hughes LJ, Moon UY, Park R, Kim SB, Tran K, Lee JS, Cho SH, Kim S. The apical complex protein Pals1 is required to maintain cerebellar progenitor cells in a proliferative state. Development 2015; 143:133-46. [PMID: 26657772 DOI: 10.1242/dev.124180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022]
Abstract
Through their biased localization and function within the cell, polarity complex proteins are necessary to establish the cellular asymmetry required for tissue organization. Well-characterized germinal zones, mitogenic signals and cell types make the cerebellum an excellent model for addressing the crucial function of polarity complex proteins in the generation and organization of neural tissues. Deletion of the apical polarity complex protein Pals1 in the developing cerebellum results in a remarkably undersized cerebellum with disrupted layers in poorly formed folia and strikingly reduced granule cell production. We demonstrate that Pals1 is not only essential for cerebellum organogenesis, but also for preventing premature differentiation and thus maintaining progenitor pools in cerebellar germinal zones, including cerebellar granule neuron precursors in the external granule layer. In the Pals1 mouse mutants, the expression of genes that regulate the cell cycle was diminished, correlating with the loss of the proliferating cell population of germinal zones. Furthermore, enhanced Shh signaling through activated Smo cannot overcome impaired cerebellar cell generation, arguing for an epistatic role of Pals1 in proliferation capacity. Our study identifies Pals1 as a novel intrinsic factor that regulates the generation of cerebellar cells and Pals1 deficiency as a potential inhibitor of overactive mitogenic signaling.
Collapse
Affiliation(s)
- Jun Young Park
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lucinda J Hughes
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA Graduate Program of Biomedical Sciences, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Uk Yeol Moon
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Raehee Park
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Sang-Bae Kim
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Khoi Tran
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Seo-Hee Cho
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Seonhee Kim
- Shriners Hospitals Pediatric Research Center, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
41
|
Sengupta M, Thirumalai V. AMPA receptor mediated synaptic excitation drives state-dependent bursting in Purkinje neurons of zebrafish larvae. eLife 2015; 4. [PMID: 26416140 PMCID: PMC4584246 DOI: 10.7554/elife.09158] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/29/2015] [Indexed: 11/13/2022] Open
Abstract
Purkinje neurons are central to cerebellar function and show membrane bistability when recorded in vitro or in vivo under anesthesia. The existence of bistability in vivo in awake animals is disputed. Here, by recording intracellularly from Purkinje neurons in unanesthetized larval zebrafish (Danio rerio), we unequivocally demonstrate bistability in these neurons. Tonic firing was seen in depolarized regimes and bursting at hyperpolarized membrane potentials. In addition, Purkinje neurons could switch from one state to another spontaneously or with current injection. While GABAAR or NMDAR were not required for bursting, activation of AMPARs by climbing fibers (CFs) was sufficient to trigger bursts. Further, by recording Purkinje neuron membrane potential intracellularly, and motor neuron spikes extracellularly, we show that initiation of motor neuron spiking is correlated with increased incidence of CF EPSPs and membrane depolarization. Developmentally, bistability was observed soon after Purkinje neuron specification and persists at least until late larval stages.
Collapse
|
42
|
Wang JY, Yu IS, Huang CC, Chen CY, Wang WP, Lin SW, Jeang KT, Chi YH. Sun1 deficiency leads to cerebellar ataxia in mice. Dis Model Mech 2015; 8:957-67. [PMID: 26035387 PMCID: PMC4527285 DOI: 10.1242/dmm.019240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/23/2015] [Indexed: 01/22/2023] Open
Abstract
Migration and organization of the nucleus are essential for the proliferation and differentiation of cells, including neurons. However, the relationship between the positioning of the nucleus and cellular morphogenesis remains poorly understood. Inherited recessive cerebellar ataxia has been attributed to mutations in SYNE1, a component of the linker of nucleoskeleton and cytoskeleton (LINC) complex. Regardless, Syne1-mutant mice present with normal cerebellar development. The Sad1-Unc-84 homology (SUN)-domain proteins are located at the inner nuclear membrane and recruit Syne proteins through the KASH domain to the outer nuclear membrane. Here, we report an unrecognized contribution of Sun1 and Sun2 to the postnatal development of murine cerebellum. Mice depleted of Sun1 showed a marked reduction in the cerebellar volume, and this phenotype is exacerbated with additional loss of a Sun2 allele. Consistent with these histological changes, Sun1(-/-) and Sun1(-/-)Sun2(+/-) mice exhibited defective motor coordination. Results of immunohistochemical analyses suggested that Sun1 is highly expressed in Purkinje cells and recruits Syne2 to the periphery of the nucleus. Approximately 33% of Purkinje cells in Sun1(-/-) mice and 66% of Purkinje cells in Sun1(-/-)Sun2(+/-) mice were absent from the surface of the internal granule layer (IGL), whereas the proliferation and migration of granule neurons were unaffected. Furthermore, the Sun1(-/-)Sun2(+/-) Purkinje cells exhibited retarded primary dendrite specification, reduced dendritic complexity and aberrant patterning of synapses. Our findings reveal a cell-type-specific role for Sun1 and Sun2 in nucleokinesis during cerebellar development, and we propose the use of Sun-deficient mice as a model for studying cerebellar ataxia that is associated with mutation of human SYNE genes or loss of Purkinje cells.
Collapse
Affiliation(s)
- Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - I-Shing Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei 10048, Taiwan Center of Genomic Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10048, Taiwan
| | - Chien-Chi Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Chia-Yen Chen
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wan-Ping Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei 10048, Taiwan Center of Genomic Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 10048, Taiwan Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan
| | - Kuan-Teh Jeang
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
43
|
Hamling KR, Tobias ZJ, Weissman TA. Mapping the development of cerebellar Purkinje cells in zebrafish. Dev Neurobiol 2015; 75:1174-88. [DOI: 10.1002/dneu.22275] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/06/2015] [Accepted: 01/07/2015] [Indexed: 01/04/2023]
Affiliation(s)
- Kyla R. Hamling
- Department of Biology; Lewis & Clark College; Portland Oregon 97219
| | | | | |
Collapse
|
44
|
Hsieh JY, Ulrich B, Issa FA, Wan J, Papazian DM. Rapid development of Purkinje cell excitability, functional cerebellar circuit, and afferent sensory input to cerebellum in zebrafish. Front Neural Circuits 2014; 8:147. [PMID: 25565973 PMCID: PMC4271617 DOI: 10.3389/fncir.2014.00147] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/02/2014] [Indexed: 11/13/2022] Open
Abstract
The zebrafish has significant advantages for studying the morphological development of the brain. However, little is known about the functional development of the zebrafish brain. We used patch clamp electrophysiology in live animals to investigate the emergence of excitability in cerebellar Purkinje cells, functional maturation of the cerebellar circuit, and establishment of sensory input to the cerebellum. Purkinje cells are born at 3 days post-fertilization (dpf). By 4 dpf, Purkinje cells spontaneously fired action potentials in an irregular pattern. By 5 dpf, the frequency and regularity of tonic firing had increased significantly and most cells fired complex spikes in response to climbing fiber activation. Our data suggest that, as in mammals, Purkinje cells are initially innervated by multiple climbing fibers that are winnowed to a single input. To probe the development of functional sensory input to the cerebellum, we investigated the response of Purkinje cells to a visual stimulus consisting of a rapid change in light intensity. At 4 dpf, sudden darkness increased the rate of tonic firing, suggesting that afferent pathways carrying visual information are already active by this stage. By 5 dpf, visual stimuli also activated climbing fibers, increasing the frequency of complex spiking. Our results indicate that the electrical properties of zebrafish and mammalian Purkinje cells are highly conserved and suggest that the same ion channels, Nav1.6 and Kv3.3, underlie spontaneous pacemaking activity. Interestingly, functional development of the cerebellum is temporally correlated with the emergence of complex, visually-guided behaviors such as prey capture. Because of the rapid formation of an electrically-active cerebellum, optical transparency, and ease of genetic manipulation, the zebrafish has great potential for functionally mapping cerebellar afferent and efferent pathways and for investigating cerebellar control of motor behavior.
Collapse
Affiliation(s)
- Jui-Yi Hsieh
- Department of Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA ; Interdepartmental Ph.D. Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA
| | - Brittany Ulrich
- Department of Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA ; Interdepartmental Ph.D. Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA
| | - Fadi A Issa
- Department of Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA
| | - Jijun Wan
- Department of Neurology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA ; Interdepartmental Ph.D. Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA ; Molecular Biology Institute, David Geffen School of Medicine at University of California Los Angeles Los Angeles, CA, USA
| |
Collapse
|
45
|
Takeuchi M, Matsuda K, Yamaguchi S, Asakawa K, Miyasaka N, Lal P, Yoshihara Y, Koga A, Kawakami K, Shimizu T, Hibi M. Establishment of Gal4 transgenic zebrafish lines for analysis of development of cerebellar neural circuitry. Dev Biol 2014; 397:1-17. [PMID: 25300581 DOI: 10.1016/j.ydbio.2014.09.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/03/2014] [Accepted: 09/26/2014] [Indexed: 02/02/2023]
Abstract
The cerebellum is involved in some forms of motor coordination and motor learning. Here we isolated transgenic (Tg) zebrafish lines that express a modified version of Gal4-VP16 (GFF) in the cerebellar neural circuits: granule, Purkinje, or eurydendroid cells, Bergmann glia, or the neurons in the inferior olive nuclei (IO) which send climbing fibers to Purkinje cells, with the transposon Tol2 system. By combining GFF lines with Tg lines carrying a reporter gene located downstream of Gal4 binding sequences (upstream activating sequence: UAS), we investigated the anatomy and developmental processes of the cerebellar neural circuitry. Combining an IO-specific Gal4 line with a UAS reporter line expressing the photoconvertible fluorescent protein Kaede demonstrated the contralateral projections of climbing fibers. Combining a granule cell-specific Gal4 line with a UAS reporter line expressing wheat germ agglutinin (WGA) confirmed direct and/or indirect connections of granule cells with Purkinje cells, eurydendroid cells, and IO neurons in zebrafish. Time-lapse analysis of a granule cell-specific Gal4 line revealed initial random movements and ventral migration of granule cell nuclei. Transgenesis of a reporter gene with another transposon Tol1 system visualized neuronal structure at a single cell resolution. Our findings indicate the usefulness of these zebrafish Gal4 Tg lines for studying the development and function of cerebellar neural circuits.
Collapse
Affiliation(s)
- Miki Takeuchi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Koji Matsuda
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Shingo Yamaguchi
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Kazuhide Asakawa
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | | | - Pradeep Lal
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | | | - Akihiko Koga
- Primate Research Institute, Kyoto University, Inuyama 464-8506, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Takashi Shimizu
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Nagoya, Aichi 464-8601, Japan; Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Masahiko Hibi
- Laboratory of Organogenesis and Organ Function, Bioscience and Biotechnology Center, Nagoya University, Nagoya, Aichi 464-8601, Japan; Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan.
| |
Collapse
|
46
|
Yamanaka T, Tosaki A, Kurosawa M, Akimoto K, Hirose T, Ohno S, Hattori N, Nukina N. Loss of aPKCλ in differentiated neurons disrupts the polarity complex but does not induce obvious neuronal loss or disorientation in mouse brains. PLoS One 2013; 8:e84036. [PMID: 24391875 PMCID: PMC3877147 DOI: 10.1371/journal.pone.0084036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 11/11/2013] [Indexed: 11/19/2022] Open
Abstract
Cell polarity plays a critical role in neuronal differentiation during development of the central nervous system (CNS). Recent studies have established the significance of atypical protein kinase C (aPKC) and its interacting partners, which include PAR-3, PAR-6 and Lgl, in regulating cell polarization during neuronal differentiation. However, their roles in neuronal maintenance after CNS development remain unclear. Here we performed conditional deletion of aPKCλ, a major aPKC isoform in the brain, in differentiated neurons of mice by camk2a-cre or synapsinI-cre mediated gene targeting. We found significant reduction of aPKCλ and total aPKCs in the adult mouse brains. The aPKCλ deletion also reduced PAR-6β, possibly by its destabilization, whereas expression of other related proteins such as PAR-3 and Lgl-1 was unaffected. Biochemical analyses suggested that a significant fraction of aPKCλ formed a protein complex with PAR-6β and Lgl-1 in the brain lysates, which was disrupted by the aPKCλ deletion. Notably, the aPKCλ deletion mice did not show apparent cell loss/degeneration in the brain. In addition, neuronal orientation/distribution seemed to be unaffected. Thus, despite the polarity complex disruption, neuronal deletion of aPKCλ does not induce obvious cell loss or disorientation in mouse brains after cell differentiation.
Collapse
Affiliation(s)
- Tomoyuki Yamanaka
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Saitama, Japan
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Asako Tosaki
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Saitama, Japan
| | - Masaru Kurosawa
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Saitama, Japan
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazunori Akimoto
- Department of Molecular Medical Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tomonori Hirose
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Yokohama City University Graduate School of Medical Science, Yokohama, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuyuki Nukina
- Laboratory for Structural Neuropathology, RIKEN Brain Science Institute, Saitama, Japan
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Core Research for Evolutionary Science and Technology, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
47
|
Hocking JC, Distel M, Köster RW. Studying cellular and subcellular dynamics in the developing zebrafish nervous system. Exp Neurol 2013; 242:1-10. [DOI: 10.1016/j.expneurol.2012.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 11/22/2011] [Accepted: 03/15/2012] [Indexed: 12/23/2022]
|
48
|
Greer YE, Fields AP, Brown AMC, Rubin JS. Atypical protein kinase Cι is required for Wnt3a-dependent neurite outgrowth and binds to phosphorylated dishevelled 2. J Biol Chem 2013; 288:9438-46. [PMID: 23396968 DOI: 10.1074/jbc.m112.448282] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously we reported that Wnt3a-dependent neurite outgrowth in Ewing sarcoma family tumor cell lines was mediated by Frizzled3, Dishevelled (Dvl), and c-Jun N-terminal kinase (Endo, Y., Beauchamp, E., Woods, D., Taylor, W. G., Toretsky, J. A., Uren, A., and Rubin, J. S. (2008) Mol. Cell. Biol. 28, 2368-2379). Subsequently, we observed that Dvl2/3 phosphorylation correlated with neurite outgrowth and that casein kinase 1δ, one of the enzymes that mediate Wnt3a-dependent Dvl phosphorylation, was required for neurite extension (Greer, Y. E., and Rubin, J. S. (2011) J. Cell Biol. 192, 993-1004). However, the functional relevance of Dvl phosphorylation in neurite outgrowth was not established. Dvl1 has been shown by others to be important for axon specification in hippocampal neurons via an interaction with atypical PKCζ, but the role of Dvl phosphorylation was not evaluated. Here we report that Ewing sarcoma family tumor cells express PKCι but not PKCζ. Wnt3a stimulated PKCι activation and caused a punctate distribution of pPKCι in the neurites and cytoplasm, with a particularly intense signal at the centrosome. Knockdown of PKCι expression with siRNA reagents blocked neurite formation in response to Wnt3a. Aurothiomalate, a specific inhibitor of PKCι/Par6 binding, also suppressed neurite extension. Wnt3a enhanced the co-immunoprecipitation of endogenous PKCι and Dvl2. Although FLAG-tagged wild-type Dvl2 immunoprecipitated with PKCι, a phosphorylation-deficient Dvl2 derivative did not. This derivative also was unable to rescue neurite outgrowth when endogenous Dvl2/3 was suppressed by siRNA (González-Sancho, J. M., Greer, Y. E., Abrahams, C. L., Takigawa, Y., Baljinnyam, B., Lee, K. H., Lee, K. S., Rubin, J. S., and Brown, A. M. (2013) J. Biol. Chem. 288, 9428-9437). Taken together, these results suggest that site-specific Dvl2 phosphorylation is required for Dvl2 association with PKCι. This interaction is likely to be one of the mechanisms essential for Wnt3a-dependent neurite outgrowth.
Collapse
Affiliation(s)
- Yoshimi Endo Greer
- From the Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
49
|
Ethanol-induced disruption of Golgi apparatus morphology, primary neurite number and cellular orientation in developing cortical neurons. Alcohol 2012; 46:619-27. [PMID: 22840816 DOI: 10.1016/j.alcohol.2012.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 07/10/2012] [Accepted: 07/12/2012] [Indexed: 01/28/2023]
Abstract
Prenatal ethanol exposure disrupts cortical neurite initiation and outgrowth, but prior studies have reported both ethanol-dependent growth promotion and inhibition. To resolve this ambiguity and better approximate in vivo conditions, we quantitatively analyzed neuronal morphology using a new, whole hemisphere explant model. In this model, Layer 6 (L6) cortical neurons migrate, laminate and extend neurites in an organotypic fashion. To selectively label L6 neurons, we performed ex utero electroporation of a GFP expression construct at embryonic day 13 and allowed the explants to develop for 2 days in vitro. Explants were exposed to (400 mg/dL) ethanol for either 4 or 24 h prior to fixation. Complete 3-D reconstructions were made of >80 GFP-positive neurons in each experimental condition. Acute responses to ethanol exposure included compaction of the Golgi apparatus accompanied by elaboration of supernumerary primary apical neurites, as well as a modest (∼15%) increase in higher order apical neurite length. With longer exposure time, ethanol exposure leads to a consistent, significant disorientation of the cell (cell body, primary apical neurite, and Golgi) with respect to the pial surface. The effects on cellular orientation were accompanied by decreased expression of cytoskeletal elements, microtubule-associated protein 2 and F-actin. These findings indicate that upon exposure to ethanol, developing L6 neurons manifest disruptions in Golgi apparatus and cytoskeletal elements which may in turn trigger selective and significant perturbations to primary neurite formation and neuronal polarity.
Collapse
|
50
|
Fujishima K, Horie R, Mochizuki A, Kengaku M. Principles of branch dynamics governing shape characteristics of cerebellar Purkinje cell dendrites. Development 2012; 139:3442-55. [PMID: 22912417 PMCID: PMC3491647 DOI: 10.1242/dev.081315] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neurons develop dendritic arbors in cell type-specific patterns. Using growing Purkinje cells in culture as a model, we performed a long-term time-lapse observation of dendrite branch dynamics to understand the rules that govern the characteristic space-filling dendrites. We found that dendrite architecture was sculpted by a combination of reproducible dynamic processes, including constant tip elongation, stochastic terminal branching, and retraction triggered by contacts between growing dendrites. Inhibition of protein kinase C/protein kinase D signaling prevented branch retraction and significantly altered the characteristic morphology of long proximal segments. A computer simulation of dendrite branch dynamics using simple parameters from experimental measurements reproduced the time-dependent changes in the dendrite configuration in live Purkinje cells. Furthermore, perturbation analysis to parameters in silico validated the important contribution of dendritic retraction in the formation of the characteristic morphology. We present an approach using live imaging and computer simulations to clarify the fundamental mechanisms of dendrite patterning in the developing brain.
Collapse
Affiliation(s)
- Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Yoshida Honmachi, Kyoto 606-8501, Japan.
| | | | | | | |
Collapse
|