1
|
Ouyang X, Sutradhar S, Trottier O, Shree S, Yu Q, Tu Y, Howard J. Neurons exploit stochastic growth to rapidly and economically build dense radially oriented dendritic arbors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639873. [PMID: 40060586 PMCID: PMC11888375 DOI: 10.1101/2025.02.24.639873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Dendrites grow by stochastic branching, elongation, and retraction. A key question is whether such a mechanism is sufficient to form highly branched dendritic morphologies. Alternatively, are signals from other cells or is the topological hierarchy of the growing network necessary for dendrite geometry? To answer these questions, we developed a mean-field model in which branch dynamics is isotropic and homogenous (i.e., no extrinsic instruction) and depends only on the average lengths and densities of branches. Branching is modeled as density-dependent nucleation so there are no tree structures and no network topology. Despite its simplicity, the model predicted several key morphological properties of class IV Drosophila sensory dendrites, including the exponential distribution of branch lengths, the parabolic scaling between dendrite number and length densities, the tight spacing of the dendritic meshwork (which required minimal total branch length), and the radial orientation of branches. Stochastic growth also accelerated the overall expansion rate of the arbor. Therefore, stochastic dynamics is an economical and rapid space-filling mechanism for building dendritic arbors without external guidance or hierarchical branching mechanisms. Our model provides a general theoretical framework for understanding how macroscopic branching patterns emerge from microscopic dynamics.
Collapse
Affiliation(s)
- Xiaoyi Ouyang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Olivier Trottier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
- Current address: Department of Chemical and Physical Sciences, University of Toronto - Mississauga, Toronto, ON M5S 1A1, Canada
| | - Sonal Shree
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Qiwei Yu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Yuhai Tu
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
- Quantitative Biology Institute, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
2
|
Ing-Esteves S, Lefebvre JL. Gamma-protocadherins regulate dendrite self-recognition and dynamics to drive self-avoidance. Curr Biol 2024; 34:4224-4239.e4. [PMID: 39214087 DOI: 10.1016/j.cub.2024.08.002] [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: 11/28/2022] [Revised: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Neurons form cell-type-specific morphologies that are shaped by cell-surface molecules and their cellular events governing dendrite growth. One growth rule is dendrite self-avoidance, whereby dendrites distribute uniformly within a neuron's territory by avoiding sibling branches. In mammalian neurons, dendrite self-avoidance is regulated by a large family of cell-recognition molecules called the clustered protocadherins (cPcdhs). Genetic and molecular studies suggest that the cPcdhs mediate homophilic recognition and repulsion between self-dendrites. However, this model has not been tested through direct investigation of self-avoidance during development. Here, we performed live imaging and four-dimensional (4D) quantifications of dendrite morphogenesis to define the dynamics and cPcdh-dependent mechanisms of self-avoidance. We focused on the mouse retinal starburst amacrine cell (SAC), which requires the gamma-Pcdhs (Pcdhgs) and self/non-self-recognition to establish a stereotypic radial morphology while permitting dendritic interactions with neighboring SACs. Through morphogenesis, SACs extend dendritic protrusions that iteratively fill the growing arbor and contact and retract from nearby self-dendrites. Compared to non-self-contacting protrusions, self-contacting events have longer lifetimes, and a subset persists as loops. In the absence of the Pcdhgs, non-self-contacting dynamics are unaffected but self-contacting retractions are significantly diminished. Self-contacting bridges accumulate, leading to the bundling of dendritic processes and disruption to the arbor shape. By tracking dendrite self-avoidance in real time, our findings establish that the γ-Pcdhs mediate self-recognition and retraction between contacting sibling dendrites. Our results also illustrate how self-avoidance shapes stochastic and space-filling dendritic outgrowth for robust pattern formation in mammalian neurons.
Collapse
Affiliation(s)
- Samantha Ing-Esteves
- Program for Neuroscience and Mental Health, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Julie L Lefebvre
- Program for Neuroscience and Mental Health, Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
3
|
Uggerud IM, Kråkenes T, Hirai H, Vedeler CA, Schubert M. Development and Optimization of a Multilayer Rat Purkinje Neuron Culture. CEREBELLUM (LONDON, ENGLAND) 2024; 23:101-111. [PMID: 36626013 PMCID: PMC10864459 DOI: 10.1007/s12311-022-01510-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/28/2022] [Indexed: 01/11/2023]
Abstract
Elucidation of the mechanisms involved in neurodegenerative diseases of the cerebellum has been hampered by the lack of robust single cell models to study Purkinje neurons and replicate at the same time in vivo features. Cerebellar Purkinje neurons are difficult to grow in dispersed cell culture, and only limited work has been done using rat cells. We developed a refined protocol for growing rat Purkinje neurons from embryonic and postnatal tissue ex vivo that results in well-developed, mature, functional, and synaptically active neurons. The rat Purkinje neurons generated are responsive to paracrine factors and genetic manipulation, allowing great experimental flexibility at the single-cell level. This ex vivo model can be used to investigate disease mechanisms that disturb Purkinje neuron morphology, function, and communication in high- and low-throughput screening formats.
Collapse
Affiliation(s)
- Ida Margrethe Uggerud
- Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine (K1), University of Bergen, 5021, Bergen, Norway
| | - Torbjørn Kråkenes
- Department of Clinical Medicine (K1), University of Bergen, 5021, Bergen, Norway
| | - Hirokazu Hirai
- Department of Neurophysiology &, Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Christian Alexander Vedeler
- Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine (K1), University of Bergen, 5021, Bergen, Norway
- Neuro-SysMed - Centre of Excellence for Experimental Therapy in Neurology, Departments of Neurology and Clinical Medicine, 5021, Bergen, Norway
| | - Manja Schubert
- Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
- Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, 01307, Dresden, Germany.
| |
Collapse
|
4
|
Liao M, Bird AD, Cuntz H, Howard J. Topology recapitulates morphogenesis of neuronal dendrites. Cell Rep 2023; 42:113268. [PMID: 38007691 PMCID: PMC10756852 DOI: 10.1016/j.celrep.2023.113268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/01/2023] [Accepted: 09/28/2023] [Indexed: 11/27/2023] Open
Abstract
Branching allows neurons to make synaptic contacts with large numbers of other neurons, facilitating the high connectivity of nervous systems. Neuronal arbors have geometric properties such as branch lengths and diameters that are optimal in that they maximize signaling speeds while minimizing construction costs. In this work, we asked whether neuronal arbors have topological properties that may also optimize their growth or function. We discovered that for a wide range of invertebrate and vertebrate neurons the distributions of their subtree sizes follow power laws, implying that they are scale invariant. The power-law exponent distinguishes different neuronal cell types. Postsynaptic spines and branchlets perturb scale invariance. Through simulations, we show that the subtree-size distribution depends on the symmetry of the branching rules governing arbor growth and that optimal morphologies are scale invariant. Thus, the subtree-size distribution is a topological property that recapitulates the functional morphology of dendrites.
Collapse
Affiliation(s)
- Maijia Liao
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Alex D Bird
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; ICAR3R-Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University, 35390 Giessen, Germany
| | - Hermann Cuntz
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; ICAR3R-Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus Liebig University, 35390 Giessen, Germany
| | - Jonathon Howard
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
5
|
Wang L, Nakazawa S, Luo W, Sato T, Mizuno H, Iwasato T. Short-Term Dendritic Dynamics of Neonatal Cortical Neurons Revealed by In Vivo Imaging with Improved Spatiotemporal Resolution. eNeuro 2023; 10:ENEURO.0142-23.2023. [PMID: 37890991 PMCID: PMC10630926 DOI: 10.1523/eneuro.0142-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Individual neurons in sensory cortices exhibit specific receptive fields based on their dendritic patterns. These dendritic morphologies are established and refined during the neonatal period through activity-dependent plasticity. This process can be visualized using two-photon in vivo time-lapse imaging, but sufficient spatiotemporal resolution is essential. We previously examined dendritic patterning from spiny stellate (SS) neurons, the major type of layer 4 (L4) neurons, in the mouse primary somatosensory cortex (barrel cortex), where mature dendrites display a strong orientation bias toward the barrel center. Longitudinal imaging at 8 h intervals revealed the long-term dynamics by which SS neurons acquire this unique dendritic pattern. However, the spatiotemporal resolution was insufficient to detect the more rapid changes in SS neuron dendrite morphology during the critical neonatal period. In the current study, we imaged neonatal L4 neurons hourly for 8 h and improved the spatial resolution by uniform cell surface labeling. The improved spatiotemporal resolution allowed detection of precise changes in dendrite morphology and revealed aspects of short-term dendritic dynamics unique to the neonatal period. Basal dendrites of barrel cortex L4 neurons were highly dynamic. In particular, both barrel-inner and barrel-outer dendrites (trees and branches) emerged/elongated and disappeared/retracted at similarly high frequencies, suggesting that SS neurons acquire biased dendrite patterns through rapid trial-and-error emergence, elongation, elimination, and retraction of dendritic trees and branches. We also found correlations between morphology and behavior (elongation/retraction) of dendritic tips. Thus, the current study revealed short-term dynamics and related features of cortical neuron dendrites during refinement.
Collapse
Affiliation(s)
- Luwei Wang
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
| | - Shingo Nakazawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima 411-8540, Japan
| | - Wenshu Luo
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima 411-8540, Japan
| | - Takuya Sato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima 411-8540, Japan
| | - Hidenobu Mizuno
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima 411-8540, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
| |
Collapse
|
6
|
Kato M, De Schutter E. Models of Purkinje cell dendritic tree selection during early cerebellar development. PLoS Comput Biol 2023; 19:e1011320. [PMID: 37486917 PMCID: PMC10399850 DOI: 10.1371/journal.pcbi.1011320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 08/03/2023] [Accepted: 06/30/2023] [Indexed: 07/26/2023] Open
Abstract
We investigate the relationship between primary dendrite selection of Purkinje cells and migration of their presynaptic partner granule cells during early cerebellar development. During postnatal development, each Purkinje cell grows more than three dendritic trees, from which a primary tree is selected for development, whereas the others completely retract. Experimental studies suggest that this selection process is coordinated by physical and synaptic interactions with granule cells, which undergo a massive migration at the same time. However, technical limitations hinder continuous experimental observation of multiple cell populations. To explore possible mechanisms underlying this selection process, we constructed a computational model using a new computational framework, NeuroDevSim. The study presents the first computational model that simultaneously simulates Purkinje cell growth and the dynamics of granule cell migrations during the first two postnatal weeks, allowing exploration of the role of physical and synaptic interactions upon dendritic selection. The model suggests that interaction with parallel fibers is important to establish the distinct planar morphology of Purkinje cell dendrites. Specific rules to select which dendritic trees to keep or retract result in larger winner trees with more synaptic contacts than using random selection. A rule based on afferent synaptic activity was less effective than rules based on dendritic size or numbers of synapses.
Collapse
Affiliation(s)
- Mizuki Kato
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa, Japan
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Erik De Schutter
- Computational Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa, Japan
| |
Collapse
|
7
|
Lopes MM, Paysan J, Rino J, Lopes SM, Pereira de Almeida L, Cortes L, Nobre RJ. A new protocol for whole-brain biodistribution analysis of AAVs by tissue clearing, light-sheet microscopy and semi-automated spatial quantification. Gene Ther 2022; 29:665-679. [PMID: 36316447 DOI: 10.1038/s41434-022-00372-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 12/23/2022]
Abstract
Recombinant adeno-associated virus (rAAV) has become one of the most promising gene delivery systems for both in vitro and in vivo applications. However, a key challenge is the lack of suitable imaging technologies to evaluate delivery, biodistribution and tropism of rAAVs and efficiently monitor disease amelioration promoted by AAV-based therapies at a whole-organ level with single-cell resolution. Therefore, we aimed to establish a new pipeline for the biodistribution analysis of natural and new variants of AAVs at a whole-brain level by tissue clearing and light-sheet fluorescence microscopy (LSFM). To test this platform, neonatal C57BL/6 mice were intravenously injected with rAAV9 encoding EGFP and, after sacrifice, brains were processed by standard immunohistochemistry and a recently released aqueous-based clearing procedure. This clearing technique required no dedicated equipment and rendered highly cleared brains, while simultaneously preserving endogenous fluorescence. Moreover, three-dimensional imaging by LSFM allowed the quantitative analysis of EGFP at a whole-brain level, as well as the reconstruction of Purkinje cells for the retrieval of valuable morphological information inaccessible by standard immunohistochemistry. In conclusion, the pipeline herein described takes the AAVs to a new level when coupled to LSFM, proving its worth as a bioimaging tool in tropism and gene therapy studies.
Collapse
Affiliation(s)
- Miguel M Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | | | - José Rino
- iMM - Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara M Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- ViraVector - Viral Vectors for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal.
- FFUC - Faculty of Pharmacy of the University of Coimbra, Coimbra, Portugal.
| | - Luísa Cortes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- MICC-CNC - Microscopy Imaging Center of Coimbra - CNC, University of Coimbra, Coimbra, Portugal.
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- ViraVector - Viral Vectors for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
8
|
Establishment and characterization of human pluripotent stem cells-derived brain organoids to model cerebellar diseases. Sci Rep 2022; 12:12513. [PMID: 35869235 PMCID: PMC9307606 DOI: 10.1038/s41598-022-16369-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/08/2022] [Indexed: 11/09/2022] Open
Abstract
The establishment of robust human brain organoids to model cerebellar diseases is essential to study new therapeutic strategies for cerebellum-associated disorders. Machado-Joseph disease (MJD) is a cerebellar hereditary neurodegenerative disease, without therapeutic options able to prevent the disease progression. In the present work, control and MJD induced-pluripotent stem cells were used to establish human brain organoids. These organoids were characterized regarding brain development, cell type composition, and MJD-associated neuropathology markers, to evaluate their value for cerebellar diseases modeling. Our data indicate that the organoids recapitulated, to some extent, aspects of brain development, such as astroglia emerging after neurons and the presence of ventricular-like zones surrounded by glia and neurons that are found only in primate brains. Moreover, the brain organoids presented markers of neural progenitors proliferation, neuronal differentiation, inhibitory and excitatory synapses, and firing neurons. The established brain organoids also exhibited markers of cerebellar neurons progenitors and mature cerebellar neurons. Finally, MJD brain organoids showed higher ventricular-like zone numbers, an indication of lower maturation, and an increased number of ataxin-3-positive aggregates, compared with control organoids. Altogether, our data indicate that the established organoids recapitulate important characteristics of human brain development and exhibit cerebellar features, constituting a resourceful tool for testing therapeutic approaches for cerebellar diseases.
Collapse
|
9
|
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
|
10
|
Dendritic Morphology Affects the Velocity and Amplitude of Back-propagating Action Potentials. Neurosci Bull 2022; 38:1330-1346. [PMID: 35984622 PMCID: PMC9672184 DOI: 10.1007/s12264-022-00931-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/20/2022] [Indexed: 11/30/2022] Open
Abstract
The back-propagating action potential (bpAP) is crucial for neuronal signal integration and synaptic plasticity in dendritic trees. Its properties (velocity and amplitude) can be affected by dendritic morphology. Due to limited spatial resolution, it has been difficult to explore the specific propagation process of bpAPs along dendrites and examine the influence of dendritic morphology, such as the dendrite diameter and branching pattern, using patch-clamp recording. By taking advantage of Optopatch, an all-optical electrophysiological method, we made detailed recordings of the real-time propagation of bpAPs in dendritic trees. We found that the velocity of bpAPs was not uniform in a single dendrite, and the bpAP velocity differed among distinct dendrites of the same neuron. The velocity of a bpAP was positively correlated with the diameter of the dendrite on which it propagated. In addition, when bpAPs passed through a dendritic branch point, their velocity decreased significantly. Similar to velocity, the amplitude of bpAPs was also positively correlated with dendritic diameter, and the attenuation patterns of bpAPs differed among different dendrites. Simulation results from neuron models with different dendritic morphology corresponded well with the experimental results. These findings indicate that the dendritic diameter and branching pattern significantly influence the properties of bpAPs. The diversity among the bpAPs recorded in different neurons was mainly due to differences in dendritic morphology. These results may inspire the construction of neuronal models to predict the propagation of bpAPs in dendrites with enormous variation in morphology, to further illuminate the role of bpAPs in neuronal communication.
Collapse
|
11
|
Abstract
The establishment of a functioning neuronal network is a crucial step in neural development. During this process, neurons extend neurites-axons and dendrites-to meet other neurons and interconnect. Therefore, these neurites need to migrate, grow, branch and find the correct path to their target by processing sensory cues from their environment. These processes rely on many coupled biophysical effects including elasticity, viscosity, growth, active forces, chemical signaling, adhesion and cellular transport. Mathematical models offer a direct way to test hypotheses and understand the underlying mechanisms responsible for neuron development. Here, we critically review the main models of neurite growth and morphogenesis from a mathematical viewpoint. We present different models for growth, guidance and morphogenesis, with a particular emphasis on mechanics and mechanisms, and on simple mathematical models that can be partially treated analytically.
Collapse
Affiliation(s)
- Hadrien Oliveri
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK.
| |
Collapse
|
12
|
Uçar MC, Kamenev D, Sunadome K, Fachet D, Lallemend F, Adameyko I, Hadjab S, Hannezo E. Theory of branching morphogenesis by local interactions and global guidance. Nat Commun 2021; 12:6830. [PMID: 34819507 PMCID: PMC8613190 DOI: 10.1038/s41467-021-27135-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Branching morphogenesis governs the formation of many organs such as lung, kidney, and the neurovascular system. Many studies have explored system-specific molecular and cellular regulatory mechanisms, as well as self-organizing rules underlying branching morphogenesis. However, in addition to local cues, branched tissue growth can also be influenced by global guidance. Here, we develop a theoretical framework for a stochastic self-organized branching process in the presence of external cues. Combining analytical theory with numerical simulations, we predict differential signatures of global vs. local regulatory mechanisms on the branching pattern, such as angle distributions, domain size, and space-filling efficiency. We find that branch alignment follows a generic scaling law determined by the strength of global guidance, while local interactions influence the tissue density but not its overall territory. Finally, using zebrafish innervation as a model system, we test these key features of the model experimentally. Our work thus provides quantitative predictions to disentangle the role of different types of cues in shaping branched structures across scales.
Collapse
Affiliation(s)
- Mehmet Can Uçar
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria.
| | - Dmitrii Kamenev
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Kazunori Sunadome
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Dominik Fachet
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria
- IRI Life Sciences, Humboldt-Universität zu Berlin, 10115, Berlin, Germany
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
- Ming-Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177, Stockholm, Sweden
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Edouard Hannezo
- Institute of Science and Technology Austria, Am Campus 1, 3400, Klosterneuburg, Austria.
| |
Collapse
|
13
|
Luck R, Karakatsani A, Shah B, Schermann G, Adler H, Kupke J, Tisch N, Jeong HW, Back MK, Hetsch F, D'Errico A, De Palma M, Wiedtke E, Grimm D, Acker-Palmer A, von Engelhardt J, Adams RH, Augustin HG, Ruiz de Almodóvar C. The angiopoietin-Tie2 pathway regulates Purkinje cell dendritic morphogenesis in a cell-autonomous manner. Cell Rep 2021; 36:109522. [PMID: 34407407 PMCID: PMC9110807 DOI: 10.1016/j.celrep.2021.109522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/06/2021] [Accepted: 07/22/2021] [Indexed: 01/01/2023] Open
Abstract
Neuro-vascular communication is essential to synchronize central nervous system development. Here, we identify angiopoietin/Tie2 as a neuro-vascular signaling axis involved in regulating dendritic morphogenesis of Purkinje cells (PCs). We show that in the developing cerebellum Tie2 expression is not restricted to blood vessels, but it is also present in PCs. Its ligands angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are expressed in neural cells and endothelial cells (ECs), respectively. PC-specific deletion of Tie2 results in reduced dendritic arborization, which is recapitulated in neural-specific Ang1-knockout and Ang2 full-knockout mice. Mechanistically, RNA sequencing reveals that Tie2-deficient PCs present alterations in gene expression of multiple genes involved in cytoskeleton organization, dendritic formation, growth, and branching. Functionally, mice with deletion of Tie2 in PCs present alterations in PC network functionality. Altogether, our data propose Ang/Tie2 signaling as a mediator of intercellular communication between neural cells, ECs, and PCs, required for proper PC dendritic morphogenesis and function.
Collapse
Affiliation(s)
- Robert Luck
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Andromachi Karakatsani
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Bhavin Shah
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Geza Schermann
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Heike Adler
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Janina Kupke
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), University of Heidelberg, 69120 Heidelberg, Germany
| | - Nathalie Tisch
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and University of Münster, Faculty of Medicine, 48149 Münster, Germany
| | - Michaela Kerstin Back
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Florian Hetsch
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Anna D'Errico
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60323 Frankfurt, Germany
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ellen Wiedtke
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, Bioquant Center, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF), and German Center for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, 60323 Frankfurt, Germany
| | - Jakob von Engelhardt
- Institute of Pathophysiology, Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and University of Münster, Faculty of Medicine, 48149 Münster, Germany
| | - Hellmut G Augustin
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany
| | - Carmen Ruiz de Almodóvar
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| |
Collapse
|
14
|
Gao FJ, Klinedinst D, Fernandez FX, Cheng B, Savonenko A, Devenney B, Li Y, Wu D, Pomper MG, Reeves RH. Forebrain Shh overexpression improves cognitive function and locomotor hyperactivity in an aneuploid mouse model of Down syndrome and its euploid littermates. Acta Neuropathol Commun 2021; 9:137. [PMID: 34399854 PMCID: PMC8365939 DOI: 10.1186/s40478-021-01237-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/01/2021] [Indexed: 11/10/2022] Open
Abstract
Down syndrome (DS) is the leading genetic cause of intellectual disability and causes early-onset dementia and cerebellar hypoplasia. The prevalence of attention deficit hyperactivity disorder is elevated in children with DS. The aneuploid DS mouse model "Ts65Dn" shows prominent brain phenotypes, including learning and memory deficits, cerebellar hypoplasia, and locomotor hyperactivity. Previous studies indicate that impaired Sonic hedgehog (Shh) signaling contributes to neurological phenotypes associated with DS and neurodegenerative diseases. However, because of a lack of working inducible Shh knock-in mice, brain region-specific Shh overexpression and its effects on cognitive function have not been studied in vivo. Here, with Gli1-LacZ reporter mice, we demonstrated that Ts65Dn had reduced levels of Gli1, a sensitive readout of Shh signaling, in both hippocampus and cerebellum at postnatal day 6. Through site-specific transgenesis, we generated an inducible human Shh knock-in mouse, TRE-bi-hShh-Zsgreen1 (TRE-hShh), simultaneously expressing dually-lipidated Shh-Np and Zsgreen1 marker in the presence of transactivator (tTA). Double transgenic mice "Camk2a-tTA;TRE-hShh" and "Pcp2-tTA;TRE-hShh" induced Shh overexpression and activated Shh signaling in a forebrain and cerebellum, respectively, specific manner from the perinatal period. Camk2a-tTA;TRE-hShh normalized locomotor hyperactivity and improved learning and memory in 3-month-old Ts65Dn, mitigated early-onset severe cognitive impairment in 7-month-old Ts65Dn, and enhanced spatial cognition in euploid mice. Camk2a-tTA;TRE-hShh cohort maintained until 600days old showed that chronic overexpression of Shh in forebrain from the perinatal period had no effect on longevity of euploid or Ts65Dn. Pcp2-tTA;TRE-hShh did not affect cognition but mitigated the phenotype of cerebellar hypoplasia in Ts65Dn. Our study provides the first in vivo evidence that Shh overexpression from the perinatal period protects DS brain integrity and enhances learning and memory in normal mice, indicating the broad therapeutic potential of Shh ligand for other neurological conditions. Moreover, the first inducible hShh site-specific knock-in mouse could be widely used for spatiotemporal Shh signaling regulation.
Collapse
Affiliation(s)
- Feng J Gao
- Department of Physiology, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Genetic Medicine, John Hopkins University, Baltimore, MD, 21205, USA.
| | - Donna Klinedinst
- Department of Physiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
- BIO5 and McKnight Brain Research Institutes, Tucson, AZ, USA
| | - Bei Cheng
- Department of Radiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Alena Savonenko
- Department of Pathology and Neurology, John Hopkins University, Baltimore, MD, 21205, USA
| | - Benjamin Devenney
- Department of Physiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yicong Li
- Department of Physiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Dan Wu
- Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Martin G Pomper
- Department of Radiology, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Roger H Reeves
- Department of Physiology, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Genetic Medicine, John Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
15
|
Tavosanis G. Dendrite enlightenment. Curr Opin Neurobiol 2021; 69:222-230. [PMID: 34134010 DOI: 10.1016/j.conb.2021.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022]
Abstract
Neuronal dendrites acquire complex morphologies during development. These are not just the product of cell-intrinsic developmental programs; rather they are defined in close interaction with the cellular environment. Thus, to understand the molecular cascades that yield appropriate morphologies, it is essential to investigate them in vivo, in the actual complex tissue environment encountered by the differentiating neuron in the developing animal. Particularly, genetic approaches have pointed to factors controlling dendrite differentiation in vivo. These suggest that localized and transient molecular cascades might underlie the formation and stabilization of dendrite branches with neuron type-specific characteristics. Here, I highlight the need for studies of neuronal dendrite differentiation in the animal, the challenges provided by such an approach, and the promising pathways that have recently opened.
Collapse
Affiliation(s)
- Gaia Tavosanis
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, Bonn, 53127, Germany; LIMES Institute, University of Bonn, Carl-Troll-Str. 3, Bonn, 53115, Germany.
| |
Collapse
|
16
|
Bradley SS, Howe E, Bailey CDC, Vickaryous MK. The dendrite arbor of Purkinje cells is altered following to tail regeneration in the leopard gecko. Integr Comp Biol 2021; 61:370-384. [PMID: 34038505 DOI: 10.1093/icb/icab098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Purkinje cells of the cerebellum have a complex arborized arrangement of dendrites and are amongst the most distinctive cell types of the nervous system. Although the neuromorphology of Purkinje cells has been well described for some mammals and teleost fish, for most vertebrates less is known. Here we used a modified Golgi-Cox method to investigate the neuromorphology of Purkinje cells from the lizard Eublepharis macularius, the leopard gecko. Using Sholl and Branch Structure Analyses, we sought to investigate whether the neuromorphology of gecko Purkinje cells was altered is response to tail loss and regeneration. Tail loss is an evolved mechanism commonly used by geckos to escape predation. Loss of the tail represents a significant and sudden change in body length and mass, which is only partially recovered as the tail is regenerated. We predicted that tail loss and regeneration would induce a quantifiable change in Purkinje cell dendrite arborization. Post hoc comparisons of Sholl analyses data showed that geckos with regenerated tails have significant changes in dendrite diameter and the number of dendrite intersections in regions corresponding to the position of parallel fiber synapses. We propose that the neuromorphological alterations observed in gecko Purkinje cells represent a compensatory response to tail regrowth, and perhaps a role in motor learning.
Collapse
Affiliation(s)
| | - Erika Howe
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | | | | |
Collapse
|
17
|
Transcription factor encoding of neuron subtype: Strategies that specify arbor pattern. Curr Opin Neurobiol 2021; 69:149-158. [PMID: 33895620 DOI: 10.1016/j.conb.2021.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 01/01/2023]
Abstract
Dendrite and axon arbors form scaffolds that connect a neuron to its partners; they are patterned to support the specific connectivity and computational requirements of each neuron subtype. Transcription factor networks control the specification of neuron subtypes, and the consequent diversification of their stereotyped arbor patterns during differentiation. We outline how the differentiation trajectories of stereotyped arbors are shaped by hierarchical deployment of precursor cell and postmitotic transcription factors. These transcription factors exert modular control over the dendrite and axon features of a single neuron, create spatial and functional compartmentalization of an arbor, instruct implementation of developmental patterning rules, and exert operational control over the cell biological processes that construct an arbor.
Collapse
|
18
|
Fujishima K, Kurisu J, Yamada M, Kengaku M. βIII spectrin controls the planarity of Purkinje cell dendrites by modulating perpendicular axon-dendrite interactions. Development 2020; 147:226102. [PMID: 33234719 DOI: 10.1242/dev.194530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/04/2020] [Indexed: 01/14/2023]
Abstract
The mechanism underlying the geometrical patterning of axon and dendrite wiring remains elusive, despite its crucial importance in the formation of functional neural circuits. The cerebellar Purkinje cell (PC) arborizes a typical planar dendrite, which forms an orthogonal network with granule cell (GC) axons. By using electrospun nanofiber substrates, we reproduce the perpendicular contacts between PC dendrites and GC axons in culture. In the model system, PC dendrites show a preference to grow perpendicularly to aligned GC axons, which presumably contribute to the planar dendrite arborization in vivo We show that βIII spectrin, a causal protein for spinocerebellar ataxia type 5, is required for the biased growth of dendrites. βIII spectrin deficiency causes actin mislocalization and excessive microtubule invasion in dendritic protrusions, resulting in abnormally oriented branch formation. Furthermore, disease-associated mutations affect the ability of βIII spectrin to control dendrite orientation. These data indicate that βIII spectrin organizes the mouse dendritic cytoskeleton and thereby regulates the oriented growth of dendrites with respect to the afferent axons.
Collapse
Affiliation(s)
- Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Junko Kurisu
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Midori Yamada
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Mineko Kengaku
- Institute for Integrated Cell-Material Sciences (KUIAS-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
19
|
Ariyani W, Miyazaki W, Amano I, Hanamura K, Shirao T, Koibuchi N. Soy Isoflavones Accelerate Glial Cell Migration via GPER-Mediated Signal Transduction Pathway. Front Endocrinol (Lausanne) 2020; 11:554941. [PMID: 33250856 PMCID: PMC7672195 DOI: 10.3389/fendo.2020.554941] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/12/2020] [Indexed: 01/04/2023] Open
Abstract
Soybean isoflavones, such as genistein, daidzein, and its metabolite, S-equol, are widely known as phytoestrogens. Their biological actions are thought to be exerted via the estrogen signal transduction pathway. Estrogens, such as 17β-estradiol (E2), play a crucial role in the development and functional maintenance of the central nervous system. E2 bind to the nuclear estrogen receptor (ER) and regulates morphogenesis, migration, functional maturation, and intracellular metabolism of neurons and glial cells. In addition to binding to nuclear ER, E2 also binds to the G-protein-coupled estrogen receptor (GPER) and activates the nongenomic estrogen signaling pathway. Soybean isoflavones also bind to the ER and GPER. However, the effect of soybean isoflavone on brain development, particularly glial cell function, remains unclear. We examined the effects of soybean isoflavones using an astrocyte-enriched culture and astrocyte-derived C6 clonal cells. Isoflavones increased glial cell migration. This augmentation was suppressed by co-exposure with G15, a selective GPER antagonist, or knockdown of GPER expression using RNA interference. Isoflavones also activated actin cytoskeleton arrangement via increased actin polymerization and cortical actin, resulting in an increased number and length of filopodia. Isoflavones exposure increased the phosphorylation levels of FAK (Tyr397 and Tyr576/577), ERK1/2 (Thr202/Tyr204), Akt (Ser473), and Rac1/cdc42 (Ser71), and the expression levels of cortactin, paxillin and ERα. These effects were suppressed by knockdown of the GPER. Co-exposure of isoflavones to the selective RhoA inhibitor, rhosin, selective Cdc42 inhibitor, casin, or Rac1/Cdc42 inhibitor, ML-141, decreased the effects of isoflavones on cell migration. These findings indicate that soybean isoflavones exert their action via the GPER to activate the PI3K/FAK/Akt/RhoA/Rac1/Cdc42 signaling pathway, resulting in increased glial cell migration. Furthermore, in silico molecular docking studies to examine the binding mode of isoflavones to the GPER revealed the possibility that isoflavones bind directly to the GPER at the same position as E2, further confirming that the effects of the isoflavones are at least in part exerted via the GPER signal transduction pathway. The findings of the present study indicate that isoflavones may be an effective supplement to promote astrocyte migration in developing and/or injured adult brains.
Collapse
Affiliation(s)
- Winda Ariyani
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Wataru Miyazaki
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
- Department of Bioscience and Laboratory Medicine, Graduate School of Health Science, Hirosaki University, Hirosaki, Japan
| | - Izuki Amano
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Kenji Hanamura
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Noriyuki Koibuchi
- Department of Integrative Physiology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
20
|
Iwasato T. In vivo imaging of neural circuit formation in the neonatal mouse barrel cortex. Dev Growth Differ 2020; 62:476-486. [DOI: 10.1111/dgd.12693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/13/2020] [Accepted: 09/27/2020] [Indexed: 01/21/2023]
Affiliation(s)
- Takuji Iwasato
- Laboratory of Mammalian Neural Circuits National Institute of Genetics Mishima Japan
- Department of Genetics SOKENDAI Mishima Japan
| |
Collapse
|
21
|
Multiscale dynamics of branching morphogenesis. Curr Opin Cell Biol 2019; 60:99-105. [DOI: 10.1016/j.ceb.2019.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/09/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022]
|
22
|
Cha HL, Choi JM, Oh HH, Bashyal N, Kim SS, Birnbaumer L, Suh-Kim H. Deletion of the α subunit of the heterotrimeric Go protein impairs cerebellar cortical development in mice. Mol Brain 2019; 12:57. [PMID: 31221179 PMCID: PMC6585000 DOI: 10.1186/s13041-019-0477-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/29/2019] [Indexed: 02/02/2023] Open
Abstract
Go is a member of the pertussis toxin-sensitive Gi/o family. Despite its abundance in the central nervous system, the precise role of Go remains largely unknown compared to other G proteins. In the present study, we explored the functions of Go in the developing cerebellar cortex by deleting its gene, Gnao. We performed a histological analysis with cerebellar sections of adult mice by cresyl violet- and immunostaining. Global deletion of Gnao induced cerebellar hypoplasia, reduced arborization of Purkinje cell dendrites, and atrophied Purkinje cell dendritic spines and the terminal boutons of climbing fibers from the inferior olivary nucleus. These results indicate that Go-mediated signaling pathway regulates maturation of presynaptic parallel fibers from granule cells and climbing fibers during the cerebellar cortical development.
Collapse
Affiliation(s)
- Hye Lim Cha
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Jung-Mi Choi
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Huy-Hyen Oh
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Narayan Bashyal
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
- Departments of Biomedical Sciences, The Graduate School, Ajou University School of Medicine, World cup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Sung-Soo Kim
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, 27709 NC USA
- Institute of Biomedical Research (BIOMED), School of Medical Sciences, Catholic University of Argentina, Av. Alicia Moreau de Justo 1300, Edificio San Jose Piso 3, C1107AAZ Buenos Aires, Argentina
| | - Haeyoung Suh-Kim
- Departments of Anatomy, Ajou University School of Medicine, Woldcup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
- Departments of Biomedical Sciences, The Graduate School, Ajou University School of Medicine, World cup-ro 164, Yeongtong-gu, Suwon, 16499 South Korea
| |
Collapse
|
23
|
Kanaoka Y, Skibbe H, Hayashi Y, Uemura T, Hattori Y. DeTerm: Software for automatic detection of neuronal dendritic branch terminals via an artificial neural network. Genes Cells 2019; 24:464-472. [PMID: 31095815 DOI: 10.1111/gtc.12700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/11/2019] [Accepted: 05/11/2019] [Indexed: 02/06/2023]
Abstract
Dendrites of neurons receive and process synaptic or sensory inputs. The Drosophila class IV dendritic arborization (da) neuron is an established model system to explore molecular mechanisms of dendrite morphogenesis. The total number of dendritic branch terminals is one of the frequently employed parameters to characterize dendritic arborization complexity of class IV neurons. This parameter gives a useful phenotypic readout of arborization during neurogenesis, and it is typically determined by laborious manual analyses of numerous images. Ideally, an automated analysis would greatly reduce the workload; however, it is challenging to automatically discriminate dendritic branch terminals from signals of surrounding tissues in whole-mount live larvae. Here, we describe our newly developed software, called DeTerm, which automatically recognizes and quantifies dendrite branch terminals via an artificial neural network. Once we input an image file of a neuronal dendritic arbor and its region of interest information, DeTerm is capable of labeling terminals of larval class IV neurons with high precision, and it also provides positional data of individual terminals. We further show that DeTerm is applicable to other types of neurons, including mouse cerebellar Purkinje cells. DeTerm is freely available on the web and was successfully tested on Mac, Windows and Linux.
Collapse
Affiliation(s)
| | - Henrik Skibbe
- Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Yusaku Hayashi
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tadashi Uemura
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.,Research Center for Dynamic Living Systems, Kyoto University, Kyoto, Japan.,AMED-CREST, AMED, Tokyo, Japan
| | - Yukako Hattori
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Hannezo E, Simons BD. Statistical theory of branching morphogenesis. Dev Growth Differ 2018; 60:512-521. [PMID: 30357803 PMCID: PMC6334508 DOI: 10.1111/dgd.12570] [Citation(s) in RCA: 7] [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: 08/13/2018] [Revised: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 12/11/2022]
Abstract
Branching morphogenesis remains a subject of abiding interest. Although much is known about the gene regulatory programs and signaling pathways that operate at the cellular scale, it has remained unclear how the macroscopic features of branched organs, including their size, network topology and spatial patterning, are encoded. Lately, it has been proposed that, these features can be explained quantitatively in several organs within a single unifying framework. Based on large-scale organ reconstructions and cell lineage tracing, it has been argued that morphogenesis follows from the collective dynamics of sublineage-restricted self-renewing progenitor cells, localized at ductal tips, that act cooperatively to drive a serial process of ductal elongation and stochastic tip bifurcation. By correlating differentiation or cell cycle exit with proximity to maturing ducts, this dynamic results in the specification of a complex network of defined density and statistical organization. These results suggest that, for several mammalian tissues, branched epithelial structures develop as a self-organized process, reliant upon a strikingly simple, but generic, set of local rules, without recourse to a rigid and deterministic sequence of genetically programmed events. Here, we review the basis of these findings and discuss their implications.
Collapse
Affiliation(s)
| | - Benjamin D. Simons
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUK
- Wellcome Trust Centre for Stem Cell ResearchUniversity of CambridgeCambridgeUK
- Cavendish LaboratoryDepartment of PhysicsUniversity of CambridgeCambridgeUK
| |
Collapse
|
25
|
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]
|
26
|
Differential dynamics of cortical neuron dendritic trees revealed by long-term in vivo imaging in neonates. Nat Commun 2018; 9:3106. [PMID: 30082783 PMCID: PMC6078955 DOI: 10.1038/s41467-018-05563-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 07/10/2018] [Indexed: 12/13/2022] Open
Abstract
Proper neuronal circuit function relies on precise dendritic projection, which is established through activity-dependent refinement during early postnatal development. Here we revealed dynamics of dendritic refinement in the mammalian brain by conducting long-term imaging of the neonatal mouse barrel cortex. By “retrospective” analyses, we identified “prospective” barrel-edge spiny stellate (SS) neurons in early neonates, which had an apical dendrite and primitive basal dendrites (BDs). These neurons retracted the apical dendrite gradually and established strong BD orientation bias through continuous “dendritic tree” turnover. A greater chance of survival was given to BD trees emerged in the barrel-center side, where thalamocortical axons (TCAs) cluster. When the spatial bias of TCA inputs to SS neurons was lost, BD tree turnover was suppressed, and most BD trees became stable and elaborated mildly. Thus, barrel-edge SS neurons could establish the characteristic BD projection pattern through differential dynamics of dendritic trees induced by spatially biased inputs. Layer 4 stellate neurons in barrel cortex have a characteristic dendritic pattern. Here, the authors conduct long-term imaging from postnatal day 3–6 to show that an orientation bias is established through dendritic tree turnover and selective elaboration, which may be induced by biased thalamocortical inputs.
Collapse
|
27
|
Kawabata Galbraith K, Fujishima K, Mizuno H, Lee SJ, Uemura T, Sakimura K, Mishina M, Watanabe N, Kengaku M. MTSS1 Regulation of Actin-Nucleating Formin DAAM1 in Dendritic Filopodia Determines Final Dendritic Configuration of Purkinje Cells. Cell Rep 2018; 24:95-106.e9. [DOI: 10.1016/j.celrep.2018.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/01/2018] [Accepted: 06/01/2018] [Indexed: 10/28/2022] Open
|
28
|
Cacao E, Parihar VK, Limoli CL, Cucinotta FA. Stochastic Modeling of Radiation-induced Dendritic Damage on in silico Mouse Hippocampal Neurons. Sci Rep 2018; 8:5494. [PMID: 29615729 PMCID: PMC5882641 DOI: 10.1038/s41598-018-23855-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022] Open
Abstract
Cognitive dysfunction associated with radiotherapy for cancer treatment has been correlated to several factors, one of which is changes to the dendritic morphology of neuronal cells. Alterations in dendritic geometry and branching patterns are often accompanied by deficits that impact learning and memory. The purpose of this study is to develop a novel predictive model of neuronal dendritic damages caused by exposure to low linear energy transfer (LET) radiation, such as X-rays, γ-rays and high-energy protons. We established in silico representations of mouse hippocampal dentate granule cell layer (GCL) and CA1 pyramidal neurons, which are frequently examined in radiation-induced cognitive decrements. The in silico representations are used in a stochastic model that describes time dependent dendritic damage induced by exposure to low LET radiation. Changes in morphometric parameters, such as total dendritic length, number of branch points and branch number, including the Sholl analysis for single neurons are described by the model. Our model based predictions for different patterns of morphological changes based on energy deposition in dendritic segments (EDDS) will serve as a useful basis to compare specific patterns of morphological alterations caused by EDDS mechanisms.
Collapse
Affiliation(s)
- Eliedonna Cacao
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, United States of America
| | - Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada, Las Vegas, NV, United States of America.
| |
Collapse
|
29
|
Ing-Esteves S, Kostadinov D, Marocha J, Sing AD, Joseph KS, Laboulaye MA, Sanes JR, Lefebvre JL. Combinatorial Effects of Alpha- and Gamma-Protocadherins on Neuronal Survival and Dendritic Self-Avoidance. J Neurosci 2018; 38:2713-2729. [PMID: 29439167 PMCID: PMC5852656 DOI: 10.1523/jneurosci.3035-17.2018] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/12/2018] [Accepted: 01/29/2018] [Indexed: 12/16/2022] Open
Abstract
The clustered protocadherins (Pcdhs) comprise 58 cadherin-related proteins encoded by three tandemly arrayed gene clusters, Pcdh-α, Pcdh-β, and Pcdh-γ (Pcdha, Pcdhb, and Pcdhg, respectively). Pcdh isoforms from different clusters are combinatorially expressed in neurons. They form multimers that interact homophilically and mediate a variety of developmental processes, including neuronal survival, synaptic maintenance, axonal tiling, and dendritic self-avoidance. Most studies have analyzed clusters individually. Here, we assessed functional interactions between Pcdha and Pcdhg clusters. To circumvent neonatal lethality associated with deletion of Pcdhgs, we used Crispr-Cas9 genome editing in mice to combine a constitutive Pcdha mutant allele with a conditional Pcdhg allele. We analyzed roles of Pcdhas and Pcdhgs in the retina and cerebellum from mice (both sexes) lacking one or both clusters. In retina, Pcdhgs are essential for survival of inner retinal neurons and dendritic self-avoidance of starburst amacrine cells, whereas Pcdhas are dispensable for both processes. Deletion of both Pcdha and Pcdhg clusters led to far more dramatic defects in survival and self-avoidance than Pcdhg deletion alone. Comparisons of an allelic series of mutants support the conclusion that Pcdhas and Pcdhgs function together in a dose-dependent and cell-type-specific manner to provide a critical threshold of Pcdh activity. In the cerebellum, Pcdhas and Pcdhgs also cooperate to mediate self-avoidance of Purkinje cell dendrites, with modest but significant defects in either single mutant and dramatic defects in the double mutant. Together, our results demonstrate complex patterns of redundancy between Pcdh clusters and the importance of Pcdh cluster diversity in postnatal CNS development.SIGNIFICANCE STATEMENT The formation of neural circuits requires diversification and combinatorial actions of cell surface proteins. Prominent among them are the clustered protocadherins (Pcdhs), a family of ∼60 neuronal recognition molecules. Pcdhs are encoded by three closely linked gene clusters called Pcdh-α, Pcdh-β, and Pcdh-γ. The Pcdhs mediate a variety of developmental processes, including neuronal survival, synaptic maintenance, and spatial patterning of axons and dendrites. Most studies to date have been limited to single clusters. Here, we used genome editing to assess interactions between Pcdh-α and Pcdh-γ gene clusters. We examined two regions of the CNS, the retina and cerebellum and show that the 14 α-Pcdhs and 22 γ-Pcdhs act synergistically to mediate neuronal survival and dendrite patterning.
Collapse
Affiliation(s)
- Samantha Ing-Esteves
- Program for Neurosciences and Mental Health, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada and
| | - Dimitar Kostadinov
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Julie Marocha
- Program for Neurosciences and Mental Health, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada and
| | - Anson D Sing
- Program for Neurosciences and Mental Health, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada and
| | - Kezia S Joseph
- Program for Neurosciences and Mental Health, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada and
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Julie L Lefebvre
- Program for Neurosciences and Mental Health, Hospital for Sick Children, Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 0A4, Canada and
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| |
Collapse
|
30
|
Muñoz-Castañeda R, Díaz D, Peris L, Andrieux A, Bosc C, Muñoz-Castañeda JM, Janke C, Alonso JR, Moutin MJ, Weruaga E. Cytoskeleton stability is essential for the integrity of the cerebellum and its motor- and affective-related behaviors. Sci Rep 2018; 8:3072. [PMID: 29449678 PMCID: PMC5814431 DOI: 10.1038/s41598-018-21470-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/29/2018] [Indexed: 01/06/2023] Open
Abstract
The cerebellum plays a key role in motor tasks, but its involvement in cognition is still being considered. Although there is an association of different psychiatric and cognitive disorders with cerebellar impairments, the lack of time-course studies has hindered the understanding of the involvement of cerebellum in cognitive and non-motor functions. Such association was here studied using the Purkinje Cell Degeneration mutant mouse, a model of selective and progressive cerebellar degeneration that lacks the cytosolic carboxypeptidase 1 (CCP1). The effects of the absence of this enzyme on the cerebellum of mutant mice were analyzed both in vitro and in vivo. These analyses were carried out longitudinally (throughout both the pre-neurodegenerative and neurodegenerative stages) and different motor and non-motor tests were performed. We demonstrate that the lack of CCP1 affects microtubule dynamics and flexibility, defects that contribute to the morphological alterations of the Purkinje cells (PCs), and to progressive cerebellar breakdown. Moreover, this degeneration led not only to motor defects but also to gradual cognitive impairments, directly related to the progression of cellular damage. Our findings confirm the cerebellar implication in non-motor tasks, where the formation of the healthy, typical PCs structure is necessary for normal cognitive and affective behavior.
Collapse
Affiliation(s)
- Rodrigo Muñoz-Castañeda
- Laboratory of Neural Plasticity and Neurorepair. Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, E-37007, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), E-37007, Salamanca, Spain
| | - David Díaz
- Laboratory of Neural Plasticity and Neurorepair. Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, E-37007, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), E-37007, Salamanca, Spain
| | - Leticia Peris
- Inserm, U1216, F-38000, Grenoble, France.,Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France
| | - Annie Andrieux
- Inserm, U1216, F-38000, Grenoble, France.,Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France.,CEA, BIG-GPC, F-38000, Grenoble, France
| | - Christophe Bosc
- Inserm, U1216, F-38000, Grenoble, France.,Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France
| | - José M Muñoz-Castañeda
- Physics Department, Aeronautics Engineering School, Polytechnic University of Madrid, E-28040, Madrid, Spain
| | - Carsten Janke
- Institut Curie, F-91405, Orsay, France.,Paris Sciences et Lettres Research University, F-75005, Paris, France.,Centre National de la Recherche Scientifique, UMR3348, F-91405, Orsay, France
| | - José R Alonso
- Laboratory of Neural Plasticity and Neurorepair. Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, E-37007, Salamanca, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), E-37007, Salamanca, Spain.,Institute for Higher Research, University of Tarapaca, Arica, Chile
| | - Marie-Jo Moutin
- Inserm, U1216, F-38000, Grenoble, France.,Université Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, F-38000, Grenoble, France
| | - Eduardo Weruaga
- Laboratory of Neural Plasticity and Neurorepair. Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, E-37007, Salamanca, Spain. .,Institute for Biomedical Research of Salamanca (IBSAL), E-37007, Salamanca, Spain.
| |
Collapse
|
31
|
Dendritic space-filling requires a neuronal type-specific extracellular permissive signal in Drosophila. Proc Natl Acad Sci U S A 2017; 114:E8062-E8071. [PMID: 28874572 DOI: 10.1073/pnas.1707467114] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurons sometimes completely fill available space in their receptive fields with evenly spaced dendrites to uniformly sample sensory or synaptic information. The mechanisms that enable neurons to sense and innervate all space in their target tissues are poorly understood. Using Drosophila somatosensory neurons as a model, we show that heparan sulfate proteoglycans (HSPGs) Dally and Syndecan on the surface of epidermal cells act as local permissive signals for the dendritic growth and maintenance of space-filling nociceptive C4da neurons, allowing them to innervate the entire skin. Using long-term time-lapse imaging with intact Drosophila larvae, we found that dendrites grow into HSPG-deficient areas but fail to stay there. HSPGs are necessary to stabilize microtubules in newly formed high-order dendrites. In contrast to C4da neurons, non-space-filling sensory neurons that develop in the same microenvironment do not rely on HSPGs for their dendritic growth. Furthermore, HSPGs do not act by transporting extracellular diffusible ligands or require leukocyte antigen-related (Lar), a receptor protein tyrosine phosphatase (RPTP) and the only known Drosophila HSPG receptor, for promoting dendritic growth of space-filling neurons. Interestingly, another RPTP, Ptp69D, promotes dendritic growth of C4da neurons in parallel to HSPGs. Together, our data reveal an HSPG-dependent pathway that specifically allows dendrites of space-filling neurons to innervate all target tissues in Drosophila.
Collapse
|
32
|
Lefebvre JL. Neuronal territory formation by the atypical cadherins and clustered protocadherins. Semin Cell Dev Biol 2017; 69:111-121. [DOI: 10.1016/j.semcdb.2017.07.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/04/2023]
|
33
|
Sherkhane P, Kapfhammer JP. Chronic pharmacological blockade of the Na + /Ca 2+ exchanger modulates the growth and development of the Purkinje cell dendritic arbor in mouse cerebellar slice cultures. Eur J Neurosci 2017; 46:2108-2120. [PMID: 28715135 DOI: 10.1111/ejn.13649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 07/11/2017] [Accepted: 07/11/2017] [Indexed: 01/22/2023]
Abstract
The Na+ /Ca2+ exchanger (NCX) is a bidirectional plasma membrane antiporter involved in Ca2+ homeostasis in eukaryotes. NCX has three isoforms, NCX1-3, and all of them are expressed in the cerebellum. Immunostaining on cerebellar slice cultures indicates that NCX is widely expressed in the cerebellum, including expression in Purkinje cells. The pharmacological blockade of the forward mode of NCX (Ca2+ efflux mode) by bepridil moderately inhibited growth and development of Purkinje cell dendritic arbor in cerebellar slice cultures. However, the blockade of the reverse mode (Ca2+ influx mode) by KB-R7943 severely reduced the dendritic arbor and induced a morphological change with thickened distal dendrites. The effect of KB-R7943 on dendritic growth was unrelated to the activity of voltage-gated calcium channels and was also apparent in the absence of bioelectrical activity indicating that it was mediated by NCX expressed in Purkinje cells. We have used additional NCX inhibitors including CB-DMB, ORM-10103, SEA0400, YM-244769, and SN-6 which have higher specificity for NCX isoforms and target either the forward, reverse, or both modes. These inhibitors caused a strong dendritic reduction similar to that seen with KB-R7943, but did not elicit thickening of distal dendrites. Our findings indicate that disturbance of the NCX-dependent calcium transport in Purkinje cells induces a reduction of dendritic arbor, which is presumably caused by changes in the calcium handling, and underline the importance of the calcium equilibrium for the dendritic development in cerebellar Purkinje cells.
Collapse
Affiliation(s)
- Pradeep Sherkhane
- Department of Biomedicine, Anatomical Institute, University of Basel, Pestalozzistrasse 20, CH-4056, Basel, Switzerland
| | - Josef P Kapfhammer
- Department of Biomedicine, Anatomical Institute, University of Basel, Pestalozzistrasse 20, CH-4056, Basel, Switzerland
| |
Collapse
|
34
|
Surface chemistry for cytosolic gene delivery and photothermal transgene expression by gold nanorods. Sci Rep 2017; 7:4694. [PMID: 28680130 PMCID: PMC5498644 DOI: 10.1038/s41598-017-04912-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/22/2017] [Indexed: 11/09/2022] Open
Abstract
Light-inducible gene regulation has great potential for remote and noninvasive control of the fate and function of target cells. One method to achieve such control is delivery of heat shock protein (HSP) promoter-driven protein expression vectors and photothermal heaters into the cells, followed by activation by illumination. In this study, we show that gold nanorods (AuNRs) functionalized with two conventional lipids, oleate and 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), are capable of efficient transfection and quick photoactivation of the HSP promoter. Use of our AuNRs (DOTAP-AuNRs) was comparable to Lipofectamine 2000 in terms of transfection efficiency, while lower in cytotoxicity. Subsequent near-infrared laser (NIR) illumination of the cells transfected by DOTAP-AuNRs for 10 s induced time- and site-specific transgene expression without significant phototoxicity, to a degree similar to that of heating the entire culture dish for 30 min. Our mechanistic studies suggest that efficient transfection and quick photoactivation of the HSP promoter (HSP70b’) are due to the promoted endosomal escape of DOTAP-AuNRs. We propose a novel protocol for NIR-inducible, site-directed gene expression using an unprecedented complex of the three conventional components capable of both transfection and photothermal heating.
Collapse
|
35
|
Hatsukano T, Kurisu J, Fukumitsu K, Fujishima K, Kengaku M. Thyroid Hormone Induces PGC-1α during Dendritic Outgrowth in Mouse Cerebellar Purkinje Cells. Front Cell Neurosci 2017; 11:133. [PMID: 28536504 PMCID: PMC5422430 DOI: 10.3389/fncel.2017.00133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/20/2017] [Indexed: 11/26/2022] Open
Abstract
Thyroid hormone 3,3′,5-Triiodo-L-thyronine (T3) is essential for proper brain development. Perinatal loss of T3 causes severe growth defects in neurons and glia, including strong inhibition of dendrite formation in Purkinje cells in the cerebellar cortex. Here we show that T3 promotes dendritic outgrowth of Purkinje cells through induction of peroxisome proliferator-activated receptor gamma (PPARγ) co-activator 1α (PGC-1α), a master regulator of mitochondrial biogenesis. PGC-1α expression in Purkinje cells is upregulated during dendritic outgrowth in normal mice, while it is significantly retarded in hypothyroid mice or in cultures depleted of T3. In cultured Purkinje cells, PGC-1α knockdown or molecular perturbation of PGC-1α signaling inhibits enhanced dendritic outgrowth and mitochondrial generation and activation caused by T3 treatment. In contrast, PGC-1α overexpression promotes dendrite extension even in the absence of T3. PGC-1α knockdown also downregulates dendrite formation in Purkinje cells in vivo. Our findings suggest that the growth-promoting activity of T3 is partly mediated by PGC-1α signaling in developing Purkinje cells.
Collapse
Affiliation(s)
- Tetsu Hatsukano
- Kengaku Group, Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto UniversityKyoto, Japan.,Kengaku Group, Graduate School of Biostudies, Kyoto UniversityKyoto, Japan
| | - Junko Kurisu
- Kengaku Group, Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto UniversityKyoto, Japan
| | - Kansai Fukumitsu
- Kengaku Group, Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto UniversityKyoto, Japan.,Kengaku Group, Graduate School of Biostudies, Kyoto UniversityKyoto, Japan
| | - Kazuto Fujishima
- Kengaku Group, Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto UniversityKyoto, Japan
| | - Mineko Kengaku
- Kengaku Group, Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto UniversityKyoto, Japan.,Kengaku Group, Graduate School of Biostudies, Kyoto UniversityKyoto, Japan
| |
Collapse
|
36
|
The Dendritic Differentiation of Purkinje Neurons: Unsolved Mystery in Formation of Unique Dendrites. THE CEREBELLUM 2016; 14:227-30. [PMID: 25015299 DOI: 10.1007/s12311-014-0585-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
37
|
Hassan BA, Hiesinger PR. Beyond Molecular Codes: Simple Rules to Wire Complex Brains. Cell 2016; 163:285-91. [PMID: 26451480 DOI: 10.1016/j.cell.2015.09.031] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Indexed: 11/30/2022]
Abstract
Molecular codes, like postal zip codes, are generally considered a robust way to ensure the specificity of neuronal target selection. However, a code capable of unambiguously generating complex neural circuits is difficult to conceive. Here, we re-examine the notion of molecular codes in the light of developmental algorithms. We explore how molecules and mechanisms that have been considered part of a code may alternatively implement simple pattern formation rules sufficient to ensure wiring specificity in neural circuits. This analysis delineates a pattern-based framework for circuit construction that may contribute to our understanding of brain wiring.
Collapse
Affiliation(s)
- Bassem A Hassan
- Center for the Biology of Disease, VIB, 3000 Leuven, Belgium; Center for Human Genetics, University of Leuven School of Medicine, 3000 Leuven, Belgium.
| | - P Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charite Universitätsmedizin Berlin, 10117 Berlin, Germany.
| |
Collapse
|
38
|
Abstract
The establishment of cell-type-specific dendritic arbors is fundamental for proper neural circuit formation. Here, using temporal- and cell-specific knock-down, knock-out, and overexpression approaches, we show that multiple aspects of the dendritic organization of cerebellar Purkinje cells (PCs) are controlled by a single transcriptional factor, retinoic acid-related orphan receptor-alpha (RORα), a gene defective in staggerer mutant mice. As reported earlier, RORα was required for regression of primitive dendrites before postnatal day 4 (P4). RORα was also necessary for PCs to form a single Purkinje layer from P0 to P4. The knock-down of RORα from P4 impaired the elimination of perisomatic dendrites and maturation of single stem dendrites in PCs at P8. Filopodia and spines were also absent in these PCs. The knock-down of RORα from P8 impaired the formation and maintenance of terminal dendritic branches of PCs at P14. Finally, even after dendrite formation was completed at P21, RORα was required for PCs to maintain dendritic complexity and functional synapses, but their mature innervation pattern by single climbing fibers was unaffected. Interestingly, overexpression of RORα in PCs at various developmental stages did not facilitate dendrite development, but had specific detrimental effects on PCs. Because RORα deficiency during development is closely related to the severity of spinocerebellar ataxia type 1, delineating the specific roles of RORα in PCs in vivo at different time windows during development and throughout adulthood would facilitate our understanding of the pathogenesis of cerebellar disorders. Significance statement: The genetic programs by which each neuron subtype develops and maintains dendritic arbors have remained largely unclear. This is partly because dendrite development is modulated dynamically by neuronal activities and interactions with local environmental cues in vivo. In addition, dendrites are formed and maintained by the balance between their growth and regression; the effects caused by the disruption of transcription factors during the early developmental stages could be masked by dendritic growth or regression in the later stages. Here, using temporal- and cell-specific knock-down, knock-out, and overexpression approaches in vivo, we show that multiple aspects of the dendritic organization of cerebellar Purkinje cells are controlled by a single transcriptional factor, retinoic acid-related orphan receptor alpha.
Collapse
|
39
|
Mitochondrial fission protein Drp1 regulates mitochondrial transport and dendritic arborization in cerebellar Purkinje cells. Mol Cell Neurosci 2015; 71:56-65. [PMID: 26689905 DOI: 10.1016/j.mcn.2015.12.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/16/2015] [Accepted: 12/09/2015] [Indexed: 01/08/2023] Open
Abstract
Mitochondria dynamically change their shape by repeated fission and fusion in response to physiological and pathological conditions. Recent studies have uncovered significant roles of mitochondrial fission and fusion in neuronal functions, such as neurotransmission and spine formation. However, the contribution of mitochondrial fission to the development of dendrites remains controversial. We analyzed the function of the mitochondrial fission GTPase Drp1 in dendritic arborization in cerebellar Purkinje cells. Overexpression of a dominant-negative mutant of Drp1 in postmitotic Purkinje cells enlarged and clustered mitochondria, which failed to exit from the soma into the dendrites. The emerging dendrites lacking mitochondrial transport remained short and unstable in culture and in vivo. The dominant-negative Drp1 affected neither the basal respiratory function of mitochondria nor the survival of Purkinje cells. Enhanced ATP supply by creatine treatment, but not reduced ROS production by antioxidant treatment, restored the hypomorphic dendrites caused by inhibition of Drp1 function. Collectively, our results suggest that Drp1 is required for dendritic distribution of mitochondria and thereby regulates energy supply in growing dendritic branches in developing Purkinje cells.
Collapse
|
40
|
Synergistic action of dendritic mitochondria and creatine kinase maintains ATP homeostasis and actin dynamics in growing neuronal dendrites. J Neurosci 2015; 35:5707-23. [PMID: 25855183 DOI: 10.1523/jneurosci.4115-14.2015] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The distribution of mitochondria within mature, differentiated neurons is clearly adapted to their regional physiological needs and can be perturbed under various pathological conditions, but the function of mitochondria in developing neurons has been less well studied. We have studied mitochondrial distribution within developing mouse cerebellar Purkinje cells and have found that active delivery of mitochondria into their dendrites is a prerequisite for proper dendritic outgrowth. Even when mitochondria in the Purkinje cell bodies are functioning normally, interrupting the transport of mitochondria into their dendrites severely disturbs dendritic growth. Additionally, we find that the growth of atrophic dendrites lacking mitochondria can be rescued by activating ATP-phosphocreatine exchange mediated by creatine kinase (CK). Conversely, inhibiting cytosolic CKs decreases dendritic ATP levels and also disrupts dendrite development. Mechanistically, this energy depletion appears to perturb normal actin dynamics and enhance the aggregation of cofilin within growing dendrites, reminiscent of what occurs in neurons overexpressing the dephosphorylated form of cofilin. These results suggest that local ATP synthesis by dendritic mitochondria and ATP-phosphocreatine exchange act synergistically to sustain the cytoskeletal dynamics necessary for dendritic development.
Collapse
|
41
|
Dulneva A, Lee S, Oliver PL, Di Gleria K, Kessler BM, Davies KE, Becker EBE. The mutant Moonwalker TRPC3 channel links calcium signaling to lipid metabolism in the developing cerebellum. Hum Mol Genet 2015; 24:4114-25. [PMID: 25908616 PMCID: PMC4476454 DOI: 10.1093/hmg/ddv150] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/20/2015] [Indexed: 12/22/2022] Open
Abstract
The Moonwalker (Mwk) mouse is a model of dominantly inherited cerebellar ataxia caused by a gain-of-function mutation in the transient receptor potential (TRP) channel TRPC3. Here, we report impairments in dendritic growth and synapse formation early on during Purkinje cell development in the Mwk cerebellum that are accompanied by alterations in calcium signaling. To elucidate the molecular effector pathways that regulate Purkinje cell dendritic arborization downstream of mutant TRPC3, we employed transcriptomic analysis of developing Purkinje cells isolated by laser-capture microdissection. We identified significant gene and protein expression changes in molecules involved in lipid metabolism. Consistently, lipid homeostasis in the Mwk cerebellum was found to be disturbed, and treatment of organotypic cerebellar slices with ceramide significantly improved dendritic outgrowth of Mwk Purkinje cells. These findings provide the first mechanistic insights into the TRPC3-dependent mechanisms, by which activated calcium signaling is coupled to lipid metabolism and the regulation of Purkinje cell development in the Mwk cerebellum.
Collapse
Affiliation(s)
- Anna Dulneva
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK and
| | - Sheena Lee
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK and
| | - Peter L Oliver
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK and
| | - Katalin Di Gleria
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Kay E Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK and
| | - Esther B E Becker
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK and
| |
Collapse
|
42
|
Hayashi S, Takeichi M. Emerging roles of protocadherins: from self-avoidance to enhancement of motility. J Cell Sci 2015; 128:1455-64. [PMID: 25749861 DOI: 10.1242/jcs.166306] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Protocadherins are a group of transmembrane proteins belonging to the cadherin superfamily that are subgrouped into 'clustered' and 'non-clustered' protocadherins. Although cadherin superfamily members are known to regulate various forms of cell-cell interactions, including cell-cell adhesion, the functions of protocadherins have long been elusive. Recent studies are, however, uncovering their unique roles. The clustered protocadherins regulate neuronal survival, as well as dendrite self-avoidance. Combinatorial expression of clustered protocadherin isoforms creates a great diversity of adhesive specificity for cells, and this process is likely to underlie the dendritic self-avoidance. Non-clustered protocadherins promote cell motility rather than the stabilization of cell adhesion, unlike the classic cadherins, and mediate dynamic cellular processes, such as growth cone migration. Protocadherin dysfunction in humans is implicated in neurological disorders, such as epilepsy and mental retardation. This Commentary provides an overview of recent findings regarding protocadherin functions, as well as a discussion of the molecular basis underlying these functions.
Collapse
Affiliation(s)
- Shuichi Hayashi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Masatoshi Takeichi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| |
Collapse
|
43
|
Wu YK, Fujishima K, Kengaku M. Differentiation of apical and basal dendrites in pyramidal cells and granule cells in dissociated hippocampal cultures. PLoS One 2015; 10:e0118482. [PMID: 25705877 PMCID: PMC4338060 DOI: 10.1371/journal.pone.0118482] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/18/2015] [Indexed: 11/21/2022] Open
Abstract
Hippocampal pyramidal cells and dentate granule cells develop morphologically distinct dendritic arbors, yet also share some common features. Both cell types form a long apical dendrite which extends from the apex of the cell soma, while short basal dendrites are developed only in pyramidal cells. Using quantitative morphometric analyses of mouse hippocampal cultures, we evaluated the differences in dendritic arborization patterns between pyramidal and granule cells. Furthermore, we observed and described the final apical dendrite determination during dendritic polarization by time-lapse imaging. Pyramidal and granule cells in culture exhibited similar dendritic patterns with a single principal dendrite and several minor dendrites so that the cell types were not readily distinguished by appearance. While basal dendrites in granule cells are normally degraded by adulthood in vivo, cultured granule cells retained their minor dendrites. Asymmetric growth of a single principal dendrite harboring the Golgi was observed in both cell types soon after the onset of dendritic growth. Time-lapse imaging revealed that up until the second week in culture, final principal dendrite designation was not stabilized, but was frequently replaced by other minor dendrites. Before dendritic polarity was stabilized, the Golgi moved dynamically within the soma and was repeatedly repositioned at newly emerging principal dendrites. Our results suggest that polarized growth of the apical dendrite is regulated by cell intrinsic programs, while regression of basal dendrites requires cue(s) from the extracellular environment in the dentate gyrus. The apical dendrite designation is determined from among multiple growing dendrites of young developing neurons.
Collapse
Affiliation(s)
- You Kure Wu
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | - Mineko Kengaku
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
44
|
Lee J, Peng Y, Lin WY, Parrish JZ. Coordinate control of terminal dendrite patterning and dynamics by the membrane protein Raw. Development 2014; 142:162-73. [PMID: 25480915 DOI: 10.1242/dev.113423] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The directional flow of information in neurons depends on compartmentalization: dendrites receive inputs whereas axons transmit them. Axons and dendrites likewise contain structurally and functionally distinct subcompartments. Axon/dendrite compartmentalization can be attributed to neuronal polarization, but the developmental origin of subcompartments in axons and dendrites is less well understood. To identify the developmental bases for compartment-specific patterning in dendrites, we screened for mutations that affect discrete dendritic domains in Drosophila sensory neurons. From this screen, we identified mutations that affected distinct aspects of terminal dendrite development with little or no effect on major dendrite patterning. Mutation of one gene, raw, affected multiple aspects of terminal dendrite patterning, suggesting that Raw might coordinate multiple signaling pathways to shape terminal dendrite growth. Consistent with this notion, Raw localizes to branch-points and promotes dendrite stabilization together with the Tricornered (Trc) kinase via effects on cell adhesion. Raw independently influences terminal dendrite elongation through a mechanism that involves modulation of the cytoskeleton, and this pathway is likely to involve the RNA-binding protein Argonaute 1 (AGO1), as raw and AGO1 genetically interact to promote terminal dendrite growth but not adhesion. Thus, Raw defines a potential point of convergence in distinct pathways shaping terminal dendrite patterning.
Collapse
Affiliation(s)
- Jiae Lee
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Yun Peng
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Wen-Yang Lin
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
45
|
Paraneoplastic CDR2 and CDR2L antibodies affect Purkinje cell calcium homeostasis. Acta Neuropathol 2014; 128:835-52. [PMID: 25341622 PMCID: PMC4231287 DOI: 10.1007/s00401-014-1351-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Paraneoplastic cerebellar degeneration (PCD) is characterized by loss of Purkinje cells (PCs) associated with progressive pancerebellar dysfunction in the presence of onconeural Yo antibodies. These antibodies recognize the cerebellar degeneration-related antigens CDR2 and CDR2L. Response to PCD therapy is disappointing due to limited understanding of the neuropathological mechanisms. Here, we report the pathological role of CDR antibodies on the calcium homeostasis in PCs. We developed an antibody-mediated PCD model based on co-incubation of cerebellar organotypic slice culture with human patient serum or rabbit CDR2 and CDR2L antibodies. The CDR antibody-induced pathology was investigated by high-resolution multiphoton imaging and biochemical analysis. Both human and rabbit CDR antibodies were rapidly internalized by PCs and led to reduced immunoreactivity of calbindin D28K (CB) and L7/Pcp-2 as well as reduced dendritic arborizations in the remaining PCs. Washout of the CDR antibodies partially recovered CB immunoreactivity, suggesting a transient structural change in CB calcium-binding site. We discovered that CDR2 and CB co-immunoprecipitate. Furthermore, the expression levels of voltage-gated calcium channel Cav2.1, protein kinase C gamma and calcium-dependent protease, calpain-2, were increased after CDR antibody internalization. Inhibition of these signaling pathways prevented or attenuated CDR antibody-induced CB and L7/Pcp-2 immunoreactivity loss, morphological changes and increased protein expression. These results signify that CDR antibody internalization causes dysregulation of cell calcium homeostasis. Hence, drugs that modulate these events may represent novel neuroprotective therapies that limit the damaging effects of CDR antibodies and prevent PC neurodegeneration.
Collapse
|
46
|
Gibson DA, Tymanskyj S, Yuan RC, Leung HC, Lefebvre JL, Sanes JR, Chédotal A, Ma L. Dendrite self-avoidance requires cell-autonomous slit/robo signaling in cerebellar purkinje cells. Neuron 2014; 81:1040-1056. [PMID: 24607227 DOI: 10.1016/j.neuron.2014.01.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
Dendrites from the same neuron usually develop nonoverlapping patterns by self-avoidance, a process requiring contact-dependent recognition and repulsion. Recent studies have implicated homophilic interactions of cell surface molecules, including Dscams and Pcdhgs, in self-recognition, but repulsive molecular mechanisms remain obscure. Here, we report a role for the secreted molecule Slit2 and its receptor Robo2 in self-avoidance of cerebellar Purkinje cells (PCs). Both molecules are highly expressed by PCs, and their deletion leads to excessive dendrite self-crossing without affecting arbor size and shape. This cell-autonomous function is supported by the boundary-establishing activity of Slit in culture and the phenotype rescue by membrane-associated Slit2 activities. Furthermore, genetic studies show that they act independently from Pcdhg-mediated recognition. Finally, PC-specific deletion of Robo2 is associated with motor behavior alterations. Thus, our study uncovers a local repulsive mechanism required for self-avoidance and demonstrates the molecular complexity at the cell surface in dendritic patterning.
Collapse
Affiliation(s)
- Daniel A Gibson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Stephen Tymanskyj
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Rachel C Yuan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Haiwen C Leung
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Julie L Lefebvre
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alain Chédotal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMR_S968, CNRS_UMR7210, Institut de la Vision, 750012, Paris, France
| | - Le Ma
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
47
|
Toyoda S, Kawaguchi M, Kobayashi T, Tarusawa E, Toyama T, Okano M, Oda M, Nakauchi H, Yoshimura Y, Sanbo M, Hirabayashi M, Hirayama T, Hirabayashi T, Yagi T. Developmental epigenetic modification regulates stochastic expression of clustered protocadherin genes, generating single neuron diversity. Neuron 2014; 82:94-108. [PMID: 24698270 DOI: 10.1016/j.neuron.2014.02.005] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2014] [Indexed: 01/08/2023]
Abstract
In the brain, enormous numbers of neurons have functional individuality and distinct circuit specificities. Clustered Protocadherins (Pcdhs), diversified cell-surface proteins, are stochastically expressed by alternative promoter choice and affect dendritic arborization in individual neurons. Here we found that the Pcdh promoters are differentially methylated by the de novo DNA methyltransferase Dnmt3b during early embryogenesis. To determine this methylation's role in neurons, we produced chimeric mice from Dnmt3b-deficient induced pluripotent stem cells (iPSCs). Single-cell expression analysis revealed that individual Dnmt3b-deficient Purkinje cells expressed increased numbers of Pcdh isoforms; in vivo, they exhibited abnormal dendritic arborization. These results indicate that DNA methylation by Dnmt3b at early embryonic stages regulates the probability of expression for the stochastically expressed Pcdh isoforms. They also suggest a mechanism for a rare human recessive disease, the ICF (Immunodeficiency, Centromere instability, and Facial anomalies) syndrome, which is caused by Dnmt3b mutations.
Collapse
Affiliation(s)
- Shunsuke Toyoda
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahumi Kawaguchi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihiro Kobayashi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Japan Science Technology Agency, ERATO, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Etsuko Tarusawa
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Division of Developmental Neurophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Tomoko Toyama
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masaki Okano
- Laboratory for Mammalian Epigenetic Studies, RIKEN Center for Developmental Biology, Minatojima-minamimachi 2-2-3, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masaaki Oda
- Laboratory for Mammalian Epigenetic Studies, RIKEN Center for Developmental Biology, Minatojima-minamimachi 2-2-3, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan; Japan Science Technology Agency, ERATO, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yumiko Yoshimura
- Division of Developmental Neurophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Aichi 444-8585, Japan
| | - Makoto Sanbo
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Masumi Hirabayashi
- Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Aichi 444-8585, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Teruyoshi Hirayama
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takahiro Hirabayashi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeshi Yagi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan; Japan Science and Technology Agency-Core Research for Evolutional Science and Technology, CREST, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
48
|
Shimono K, Fujishima K, Nomura T, Ohashi M, Usui T, Kengaku M, Toyoda A, Uemura T. An evolutionarily conserved protein CHORD regulates scaling of dendritic arbors with body size. Sci Rep 2014; 4:4415. [PMID: 24643112 PMCID: PMC3958717 DOI: 10.1038/srep04415] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/04/2014] [Indexed: 12/17/2022] Open
Abstract
Most organs scale proportionally with body size through regulation of individual cell size and/or cell number. Here we addressed how postmitotic and morphologically complex cells such as neurons scale with the body size by using the dendritic arbor of one Drosophila sensory neuron as an assay system. In small adults eclosed under a limited-nutrition condition, the wild-type neuron preserved the branching complexity of the arbor, but scaled down the entire arbor, making a “miniature”. In contrast, mutant neurons for the Insulin/IGF signaling (IIS) or TORC1 pathway exhibited “undergrowth”, which was characterized by decreases in both the branching complexity and the arbor size, despite a normal diet. These contrasting phenotypes hinted that a novel regulatory mechanism contributes to the dendritic scaling in wild-type neurons. Indeed, we isolated a mutation in the gene CHORD/morgana that uncoupled the neuron size and the body size: CHORD mutant neurons generated miniature dendritic arbors regardless of the body size. CHORD encodes an evolutionarily conserved co-chaperone of HSP90. Our results support the notion that dendritic growth and branching are controlled by partly separate mechanisms. The IIS/TORC1 pathways control both growth and branching to avert underdevelopment, whereas CHORD together with TORC2 realizes proportional scaling of the entire arbor.
Collapse
Affiliation(s)
- Kohei Shimono
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University Kyoto 606-8501, Japan
| | - Takafumi Nomura
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Masayoshi Ohashi
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Tadao Usui
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Mineko Kengaku
- 1] Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan [2] Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University Kyoto 606-8501, Japan
| | - Atsushi Toyoda
- Center for Information Biology, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Tadashi Uemura
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
49
|
Baker MW, Macagno ER. Control of neuronal morphology and connectivity: Emerging developmental roles for gap junctional proteins. FEBS Lett 2014; 588:1470-9. [DOI: 10.1016/j.febslet.2014.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/10/2014] [Accepted: 02/12/2014] [Indexed: 11/25/2022]
|
50
|
The plasma membrane Ca2+-ATPase2 (PMCA2) is involved in the regulation of Purkinje cell dendritic growth in cerebellar organotypic slice cultures. Neural Plast 2013; 2013:321685. [PMID: 24288624 PMCID: PMC3830849 DOI: 10.1155/2013/321685] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/30/2013] [Accepted: 09/03/2013] [Indexed: 11/17/2022] Open
Abstract
Purkinje cells are the principal neurons of the cerebellar cortex and have an extensive and elaborate dendritic tree. Chronic activation of type I metabotropic glutamate receptors inhibits Purkinje cell dendritic growth in organotypic cerebellar slice cultures. This effect is mediated by calcium influx through P/Q-type and T-type Ca2+ channels. We have now studied the role of the plasma membrane Ca2+-ATPase2 (PMCA2), a major calcium extrusion pump, for Purkinje cell dendritic development. We found that PMCA2 is strongly expressed in the plasma membrane and dendritic spines of Purkinje cells in organotypic slice cultures compatible with a role for controlling the local dendritic calcium equilibrium. Inhibition of PMCA2 activity by carboxyeosin resulted in a moderate reduction of Purkinje cell dendritic tree size indicating that the extrusion of calcium by PMCA2 is important for maintaining the dendritic calcium concentration and controlling dendritic growth. When inhibition of PMCA2 was combined with stimulation of type I metabotropic glutamate receptors, it partially rescued dendritic morphology. This protection can be explained by a compensatory inactivation of voltage-gated calcium channels in Purkinje cells after PMCA2 inhibition. Our results demonstrate that PMCA2 activity is an important regulator of the dendritic calcium equilibrium controlling Purkinje cell dendritic growth.
Collapse
|