1
|
Matsumoto N, Barson D, Liang L, Crair MC. Hebbian instruction of axonal connectivity by endogenous correlated spontaneous activity. Science 2024; 385:eadh7814. [PMID: 39146415 DOI: 10.1126/science.adh7814] [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: 03/17/2023] [Revised: 05/07/2024] [Accepted: 06/21/2024] [Indexed: 08/17/2024]
Abstract
Spontaneous activity refines neural connectivity prior to the onset of sensory experience, but it remains unclear how such activity instructs axonal connectivity with subcellular precision. We simultaneously measured spontaneous retinal waves and the activity of individual retinocollicular axons and tracked morphological changes in axonal arbors across hours in vivo in neonatal mice. We demonstrate that the correlation of an axon branch's activity with neighboring axons or postsynaptic neurons predicts whether the branch will be added, stabilized, or eliminated. Desynchronizing individual axons from their local networks, changing the pattern of correlated activity, or blocking N-methyl-d-aspartate receptors all significantly altered single-axon morphology. These observations provide the first direct evidence in vivo that endogenous patterns of correlated neuronal activity instruct fine-scale refinement of axonal processes.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute for Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Daniel Barson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Liang Liang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute for Neuroscience, Yale University, New Haven, CT 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
| | - Michael C Crair
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
- Kavli Institute for Neuroscience, Yale University, New Haven, CT 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
- Department of Ophthalmology & Visual Science, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
2
|
Baltar J, Miranda RM, Cabral M, Rebelo S, Grahammer F, Huber TB, Reguenga C, Monteiro FA. Neph1 is required for neurite branching and is negatively regulated by the PRRXL1 homeodomain factor in the developing spinal cord dorsal horn. Neural Dev 2024; 19:13. [PMID: 39049046 PMCID: PMC11271021 DOI: 10.1186/s13064-024-00190-6] [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: 05/16/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
The cell-adhesion molecule NEPH1 is required for maintaining the structural integrity and function of the glomerulus in the kidneys. In the nervous system of Drosophila and C. elegans, it is involved in synaptogenesis and axon branching, which are essential for establishing functional circuits. In the mammalian nervous system, the expression regulation and function of Neph1 has barely been explored. In this study, we provide a spatiotemporal characterization of Neph1 expression in mouse dorsal root ganglia (DRGs) and spinal cord. After the neurogenic phase, Neph1 is broadly expressed in the DRGs and in their putative targets at the dorsal horn of the spinal cord, comprising both GABAergic and glutamatergic neurons. Interestingly, we found that PRRXL1, a homeodomain transcription factor that is required for proper establishment of the DRG-spinal cord circuit, prevents a premature expression of Neph1 in the superficial laminae of the dorsal spinal cord at E14.5, but has no regulatory effect on the DRGs or on either structure at E16.5. By chromatin immunoprecipitation analysis of the dorsal spinal cord, we identified four PRRXL1-bound regions within the Neph1 introns, suggesting that PRRXL1 directly regulates Neph1 transcription. We also showed that Neph1 is required for branching, especially at distal neurites. Together, our work showed that Prrxl1 prevents the early expression of Neph1 in the superficial dorsal horn, suggesting that Neph1 might function as a downstream effector gene for proper assembly of the DRG-spinal nociceptive circuit.
Collapse
Affiliation(s)
- João Baltar
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Rafael Mendes Miranda
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria Cabral
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Sandra Rebelo
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Departamento de Patologia Clínica, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Florian Grahammer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos Reguenga
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Filipe Almeida Monteiro
- Unidade de Biologia Experimental, Departamento de Biomedicina, FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal.
- Pain Neurobiology, IBMC - Instituto de Biologia Celular e Molecular, Porto, Portugal.
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
3
|
van Tartwijk FW, Wunderlich LCS, Mela I, Makarchuk S, Jakobs MAH, Qamar S, Franze K, Kaminski Schierle GS, St George-Hyslop PH, Lin JQ, Holt CE, Kaminski CF. Mutation of the ALS-/FTD-Associated RNA-Binding Protein FUS Affects Axonal Development. J Neurosci 2024; 44:e2148232024. [PMID: 38692734 PMCID: PMC7616130 DOI: 10.1523/jneurosci.2148-23.2024] [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: 11/14/2023] [Revised: 02/23/2024] [Accepted: 03/29/2024] [Indexed: 05/03/2024] Open
Abstract
Aberrant condensation and localization of the RNA-binding protein (RBP) fused in sarcoma (FUS) occur in variants of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Changes in RBP function are commonly associated with changes in axonal cytoskeletal organization and branching in neurodevelopmental disorders. Here, we asked whether branching defects also occur in vivo in a model of FUS-associated disease. We use two reported Xenopus models of ALS/FTD (of either sex), the ALS-associated mutant FUS(P525L) and a mimic of hypomethylated FUS, FUS(16R). Both mutants strongly reduced axonal complexity in vivo. We also observed an axon looping defect for FUS(P525L) in the target area, which presumably arises due to errors in stop cue signaling. To assess whether the loss of axon complexity also had a cue-independent component, we assessed axonal cytoskeletal integrity in vitro. Using a novel combination of fluorescence and atomic force microscopy, we found that mutant FUS reduced actin density in the growth cone, altering its mechanical properties. Therefore, FUS mutants may induce defects during early axonal development.
Collapse
Affiliation(s)
- Francesca W van Tartwijk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Lucia C S Wunderlich
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Ioanna Mela
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Stanislaw Makarchuk
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 OAH, United Kingdom
| | - Maximilian A H Jakobs
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, United Kingdom
| | - Seema Qamar
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Kristian Franze
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, United Kingdom
| | - Gabriele S Kaminski Schierle
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| | - Peter H St George-Hyslop
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, United Kingdom
- Department of Medicine, University of Toronto and University Health Network and Tanz Centre for Research in Neurodegenerative Diseases University of Toronto, Toronto, Ontario M5T 0S8, Canada
- Department of Neurology, Taub Institute For Research on Alzheimer's Disease and the Aging Brain, Columbia University Irvine Medical Center, New York, New York 10032
| | - Julie Qiaojin Lin
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 OAH, United Kingdom
- UK Dementia Research Institute Centre and Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9NU, United Kingdom
| | - Christine E Holt
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge CB2 3DY, United Kingdom
| | - Clemens F Kaminski
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, United Kingdom
| |
Collapse
|
4
|
Lanfranchi M, Yandiev S, Meyer-Dilhet G, Ellouze S, Kerkhofs M, Dos Reis R, Garcia A, Blondet C, Amar A, Kneppers A, Polvèche H, Plassard D, Foretz M, Viollet B, Sakamoto K, Mounier R, Bourgeois CF, Raineteau O, Goillot E, Courchet J. The AMPK-related kinase NUAK1 controls cortical axons branching by locally modulating mitochondrial metabolic functions. Nat Commun 2024; 15:2487. [PMID: 38514619 PMCID: PMC10958033 DOI: 10.1038/s41467-024-46146-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
The cellular mechanisms underlying axonal morphogenesis are essential to the formation of functional neuronal networks. We previously identified the autism-linked kinase NUAK1 as a central regulator of axon branching through the control of mitochondria trafficking. However, (1) the relationship between mitochondrial position, function and axon branching and (2) the downstream effectors whereby NUAK1 regulates axon branching remain unknown. Here, we report that mitochondria recruitment to synaptic boutons supports collateral branches stabilization rather than formation in mouse cortical neurons. NUAK1 deficiency significantly impairs mitochondrial metabolism and axonal ATP concentration, and upregulation of mitochondrial function is sufficient to rescue axonal branching in NUAK1 null neurons in vitro and in vivo. Finally, we found that NUAK1 regulates axon branching through the mitochondria-targeted microprotein BRAWNIN. Our results demonstrate that NUAK1 exerts a dual function during axon branching through its ability to control mitochondrial distribution and metabolic activity.
Collapse
Affiliation(s)
- Marine Lanfranchi
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Sozerko Yandiev
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Géraldine Meyer-Dilhet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Salma Ellouze
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500, Bron, France
| | - Martijn Kerkhofs
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Raphael Dos Reis
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Audrey Garcia
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Camille Blondet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Alizée Amar
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Anita Kneppers
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Hélène Polvèche
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
- CECS/AFM, I-STEM, 28 rue Henri Desbruères, F-91100, Corbeil-Essonnes, France
| | - Damien Plassard
- CNRS UMR 7104, INSERM U1258, GenomEast Platform, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Marc Foretz
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Benoit Viollet
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Rémi Mounier
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Cyril F Bourgeois
- Laboratoire de Biologie et Modelisation de la Cellule, Ecole Normale Superieure de Lyon, CNRS, UMR 5239, Inserm, U1293, Universite Claude Bernard Lyon 1, 46 allée d'Italie F-69364, Lyon, France
| | - Olivier Raineteau
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500, Bron, France
| | - Evelyne Goillot
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Julien Courchet
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France.
| |
Collapse
|
5
|
Lan Y, Zou S, Wang W, Chen Q, Zhu Y. Progress in cancer neuroscience. MedComm (Beijing) 2023; 4:e431. [PMID: 38020711 PMCID: PMC10665600 DOI: 10.1002/mco2.431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer of the central nervous system (CNS) can crosstalk systemically and locally in the tumor microenvironment and has become a topic of attention for tumor initiation and advancement. Recently studied neuronal and cancer interaction fundamentally altered the knowledge about glioma and metastases, indicating how cancers invade complex neuronal networks. This review systematically discussed the interactions between neurons and cancers and elucidates new therapeutic avenues. We have overviewed the current understanding of direct or indirect communications of neuronal cells with cancer and the mechanisms associated with cancer invasion. Besides, tumor-associated neuronal dysfunction and the influence of cancer therapies on the CNS are highlighted. Furthermore, interactions between peripheral nervous system and various cancers have also been discussed separately. Intriguingly and importantly, it cannot be ignored that exosomes could mediate the "wireless communications" between nervous system and cancer. Finally, promising future strategies targeting neuronal-brain tumor interactions were reviewed. A great deal of work remains to be done to elucidate the neuroscience of cancer, and future more research should be directed toward clarifying the precise mechanisms of cancer neuroscience, which hold enormous promise to improve outcomes for a wide range of malignancies.
Collapse
Affiliation(s)
- Yu‐Long Lan
- Department of NeurosurgerySecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Shuang Zou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
| | - Wen Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical ScienceZhejiang Chinese Medical UniversityHangzhouChina
| | - Yongjian Zhu
- Department of NeurosurgerySecond Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological DiseasesHangzhouZhejiangChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
6
|
Jiang X, Zhou L, Sun Z, Xie B, Lin H, Gao X, Deng L, Yang C. MSCs overexpressing GDNF restores brain structure and neurological function in rats with intracerebral hemorrhage. MOLECULAR BIOMEDICINE 2023; 4:43. [PMID: 38008847 PMCID: PMC10678901 DOI: 10.1186/s43556-023-00159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 11/16/2023] [Indexed: 11/28/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been applied in transplantation to treat intracerebral hemorrhage (ICH) but with limited efficacy. Accumulated evidence has shown that glial cell-derived neurotrophic factor (GDNF) plays a crucial part in neuronal protection and functional recovery of the brain after ICH; however, GDNF has difficulty crossing the blood-brain barrier, which limits its application. In this study, we investigated the influences of MSCs overexpressing GDNF (MSCs/GDNF) on the brain structure as well as gait of rats after ICH and explored the possible mechanisms. We found that cell transplantation could reverse the neurological dysfunction and brain damage caused by ICH to a certain extent, and MSCs/GDNF transplantation was superior to MSCs transplantation. Moreover, Transplantation of MSCs overexpressing GDNF effectively reduced the volume of bleeding foci and increased the level of glucose uptake in rats with ICH, which could be related to improving mitochondrial quality. Furthermore, GDNF produced by transplanted MSCs/GDNF further inhibited neuroinflammation, improved mitochondrial quality and function, promoted angiogenesis and the survival of neurons and oligodendrocytes, and enhanced synaptic plasticity in ICH rats when compared with simple MSC transplantation. Overall, our data indicate that GDNF overexpression heightens the curative effect of MSC implantation in treating rats following ICH.
Collapse
Affiliation(s)
- Xiaoqian Jiang
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ling Zhou
- Clinical Skills Center, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zihuan Sun
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Heng Lin
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoqing Gao
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Li Deng
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chaoxian Yang
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Neurobiology, Preclinical Medicine Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
7
|
Xiao Y, Czopka T. Myelination-independent functions of oligodendrocyte precursor cells in health and disease. Nat Neurosci 2023; 26:1663-1669. [PMID: 37653126 DOI: 10.1038/s41593-023-01423-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/27/2023] [Indexed: 09/02/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are a population of tissue-resident glial cells found throughout the CNS, constituting approximately 5% of all CNS cells and persisting from development to adulthood and aging. The canonical role of OPCs is to give rise to myelinating oligodendrocytes. However, additional functions of OPCs beyond this traditional role as precursors have been suggested for a long time. In this Perspective, we provide an overview of the multiple myelination-independent functions that have been described for OPCs in the context of neuron development, angiogenesis, inflammatory response, axon regeneration and their recently discovered roles in neural circuit remodeling.
Collapse
Affiliation(s)
- Yan Xiao
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
8
|
Zhang H, Papadaki S, Sun X, Wang X, Drobizhev M, Yao L, Rehbock M, Köster RW, Wu L, Namikawa K, Piatkevich KD. Quantitative assessment of near-infrared fluorescent proteins. Nat Methods 2023; 20:1605-1616. [PMID: 37666982 DOI: 10.1038/s41592-023-01975-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/29/2023] [Indexed: 09/06/2023]
Abstract
Recent progress in fluorescent protein development has generated a large diversity of near-infrared fluorescent proteins (NIR FPs), which are rapidly becoming popular probes for a variety of imaging applications. However, the diversity of NIR FPs poses a challenge for end-users in choosing the optimal one for a given application. Here we conducted a systematic and quantitative assessment of intracellular brightness, photostability, oligomeric state, chemical stability and cytotoxicity of 22 NIR FPs in cultured mammalian cells and primary mouse neurons and identified a set of top-performing FPs including emiRFP670, miRFP680, miRFP713 and miRFP720, which can cover a majority of imaging applications. The top-performing proteins were further validated for in vivo imaging of neurons in Caenorhabditis elegans, zebrafish, and mice as well as in mice liver. We also assessed the applicability of the selected NIR FPs for multicolor imaging of fusions, expansion microscopy and two-photon imaging.
Collapse
Affiliation(s)
- Hanbin Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Stavrini Papadaki
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaoting Sun
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xinyue Wang
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Mikhail Drobizhev
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Luxia Yao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Michel Rehbock
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Reinhard W Köster
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Lianfeng Wu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Kazuhiko Namikawa
- Division of Cellular and Molecular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kiryl D Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
D'Elia KP, Hameedy H, Goldblatt D, Frazel P, Kriese M, Zhu Y, Hamling KR, Kawakami K, Liddelow SA, Schoppik D, Dasen JS. Determinants of motor neuron functional subtypes important for locomotor speed. Cell Rep 2023; 42:113049. [PMID: 37676768 PMCID: PMC10600875 DOI: 10.1016/j.celrep.2023.113049] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/12/2023] [Accepted: 08/11/2023] [Indexed: 09/09/2023] Open
Abstract
Locomotion requires precise control of the strength and speed of muscle contraction and is achieved by recruiting functionally distinct subtypes of motor neurons (MNs). MNs are essential to movement and differentially susceptible in disease, but little is known about how MNs acquire functional subtype-specific features during development. Using single-cell RNA profiling in embryonic and larval zebrafish, we identify novel and conserved molecular signatures for MN functional subtypes and identify genes expressed in both early post-mitotic and mature MNs. Assessing MN development in genetic mutants, we define a molecular program essential for MN functional subtype specification. Two evolutionarily conserved transcription factors, Prdm16 and Mecom, are both functional subtype-specific determinants integral for fast MN development. Loss of prdm16 or mecom causes fast MNs to develop transcriptional profiles and innervation similar to slow MNs. These results reveal the molecular diversity of vertebrate axial MNs and demonstrate that functional subtypes are specified through intrinsic transcriptional codes.
Collapse
Affiliation(s)
- Kristen P D'Elia
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Hanna Hameedy
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Dena Goldblatt
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA
| | - Paul Frazel
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Mercer Kriese
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yunlu Zhu
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Kyla R Hamling
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Shane A Liddelow
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - David Schoppik
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Jeremy S Dasen
- Department of Neuroscience & Physiology and Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Du XF, Li FN, Peng XL, Xu B, Zhang Y, Li G, Liu T, Li Y, Wang H, Yan J, Du JL. Circadian regulation of developmental synaptogenesis via the hypocretinergic system. Nat Commun 2023; 14:3195. [PMID: 37268623 DOI: 10.1038/s41467-023-38973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
The circadian clock orchestrates a wide variety of physiological and behavioral processes, enabling animals to adapt to daily environmental changes, particularly the day-night cycle. However, the circadian clock's role in the developmental processes remains unclear. Here, we employ the in vivo long-term time-lapse imaging of retinotectal synapses in the optic tectum of larval zebrafish and reveal that synaptogenesis, a fundamental developmental process for neural circuit formation, exhibits circadian rhythm. This rhythmicity arises primarily from the synapse formation rather than elimination and requires the hypocretinergic neural system. Disruption of this synaptogenic rhythm, by impairing either the circadian clock or the hypocretinergic system, affects the arrangement of the retinotectal synapses on axon arbors and the refinement of the postsynaptic tectal neuron's receptive field. Thus, our findings demonstrate that the developmental synaptogenesis is under hypocretin-dependent circadian regulation, suggesting an important role of the circadian clock in neural development.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
| | - Fu-Ning Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Xiao-Lan Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Guang Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Taole Liu
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Ying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, 215123, Suzhou, Jiangsu, China
- School of Biology and Basic Medical Sciences, Suzhou Medical College, Soochow University, 215123, Suzhou, Jiangsu, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, 200031, Shanghai, China.
- University of Chinese Academy of Sciences, 19A Yu-Quan Road, 100049, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, 200031, Shanghai, China.
| |
Collapse
|
11
|
Dutta SB, Linneweber GA, Andriatsilavo M, Hiesinger PR, Hassan BA. EGFR-dependent suppression of synaptic autophagy is required for neuronal circuit development. Curr Biol 2023; 33:517-532.e5. [PMID: 36640763 DOI: 10.1016/j.cub.2022.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/31/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
The development of neuronal connectivity requires stabilization of dynamic axonal branches at sites of synapse formation. Models that explain how axonal branching is coupled to synaptogenesis postulate molecular regulators acting in a spatiotemporally restricted fashion to ensure branching toward future synaptic partners while also stabilizing the emerging synaptic contacts between such partners. We investigated this question using neuronal circuit development in the Drosophila brain as a model system. We report that epidermal growth factor receptor (EGFR) activity is required in presynaptic axonal branches during two distinct temporal intervals to regulate circuit wiring in the developing Drosophila visual system. EGFR is required early to regulate primary axonal branching. EGFR activity is then independently required at a later stage to prevent degradation of the synaptic active zone protein Bruchpilot (Brp). Inactivation of EGFR results in a local increase of autophagy in presynaptic branches and the translocation of active zone proteins into autophagic vesicles. The protection of synaptic material during this later interval of wiring ensures the stabilization of terminal branches, circuit connectivity, and appropriate visual behavior. Phenotypes of EGFR inactivation can be rescued by increasing Brp levels or downregulating autophagy. In summary, we identify a temporally restricted molecular mechanism required for coupling axonal branching and synaptic stabilization that contributes to the emergence of neuronal wiring specificity.
Collapse
Affiliation(s)
- Suchetana B Dutta
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany
| | | | - Maheva Andriatsilavo
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany
| | | | - Bassem A Hassan
- Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié Salpêtrière, 75013 Paris, France; Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany; Einstein-BIH, Charité Universitätsmedizin, 10117 Berlin, Germany.
| |
Collapse
|
12
|
Plata ALD, Robles E. NMDA Receptor Antagonist MK801 Reduces Dendritic Spine Density and Stability in Zebrafish Pyramidal Neurons. Neuroscience 2022; 498:50-63. [PMID: 35718218 DOI: 10.1016/j.neuroscience.2022.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/02/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022]
Abstract
NMDA-type glutamate receptors play a critical role in activity-dependent neurite growth. We employed cell type-specific genetic labeling in zebrafish to examine the effects of NMDA receptor antagonism on the morphological development of tectal pyramidal neurons (PyrNs). Our data demonstrate that the NMDA receptor antagonist MK801 reduces PyrN spine density and stability without significantly altering dendritic growth and branching. However, the axons that synapse onto PyrN dendritic spines do exhibit reduced arbor growth and branching in response to MK801 treatment. Axons that synapse with PyrNs, but not on spines, are unaffected by MK801 treatment. These findings may reflect different roles for NMDARs during the development of spiny and aspiny dendrites.
Collapse
Affiliation(s)
- Amanda Lamarca Dela Plata
- Department of Biological Sciences and Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Estuardo Robles
- Department of Biological Sciences and Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
13
|
Abstract
Nervous system assembly relies on a diversity of cellular processes ranging from dramatic tissue reorganization to local, subcellular changes all driven by precise molecular programs. Combined, these processes culminate in an animal's ability to plan and execute behaviors. Animal behavior can, therefore, serve as a functional readout of nervous system development. Benefitting from an expansive and growing set of molecular and imaging tools paired with an ever-growing number of assays of diverse behaviors, the zebrafish system has emerged as an outstanding platform at the intersection of nervous system assembly, plasticity and behavior. Here, we summarize recent advancements in the field, including how developing neural circuits are refined to shape complex behaviors and plasticity.
Collapse
Affiliation(s)
- Jessica C. Nelson
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Xiao Y, Petrucco L, Hoodless LJ, Portugues R, Czopka T. Oligodendrocyte precursor cells sculpt the visual system by regulating axonal remodeling. Nat Neurosci 2022; 25:280-284. [PMID: 35241802 PMCID: PMC8904260 DOI: 10.1038/s41593-022-01023-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 01/25/2022] [Indexed: 12/29/2022]
Abstract
Many oligodendrocyte precursor cells (OPCs) do not differentiate to form myelin, suggesting additional roles of this cell population. The zebrafish optic tectum contains OPCs in regions devoid of myelin. Elimination of these OPCs impaired precise control of retinal ganglion cell axon arbor size during formation and maturation of retinotectal connectivity and degraded functional processing of visual stimuli. Therefore, OPCs fine-tune neural circuits independently of their canonical role to make myelin.
Collapse
Affiliation(s)
- Yan Xiao
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Luigi Petrucco
- Max Planck Institute of Neurobiology, Sensorimotor Control Research Group, Martinsried, Germany
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
| | - Laura J Hoodless
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ruben Portugues
- Max Planck Institute of Neurobiology, Sensorimotor Control Research Group, Martinsried, Germany
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Tim Czopka
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
15
|
Sneve MA, Piatkevich KD. Towards a Comprehensive Optical Connectome at Single Synapse Resolution via Expansion Microscopy. Front Synaptic Neurosci 2022; 13:754814. [PMID: 35115916 PMCID: PMC8803729 DOI: 10.3389/fnsyn.2021.754814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 12/17/2021] [Indexed: 12/04/2022] Open
Abstract
Mapping and determining the molecular identity of individual synapses is a crucial step towards the comprehensive reconstruction of neuronal circuits. Throughout the history of neuroscience, microscopy has been a key technology for mapping brain circuits. However, subdiffraction size and high density of synapses in brain tissue make this process extremely challenging. Electron microscopy (EM), with its nanoscale resolution, offers one approach to this challenge yet comes with many practical limitations, and to date has only been used in very small samples such as C. elegans, tadpole larvae, fruit fly brain, or very small pieces of mammalian brain tissue. Moreover, EM datasets require tedious data tracing. Light microscopy in combination with tissue expansion via physical magnification-known as expansion microscopy (ExM)-offers an alternative approach to this problem. ExM enables nanoscale imaging of large biological samples, which in combination with multicolor neuronal and synaptic labeling offers the unprecedented capability to trace and map entire neuronal circuits in fully automated mode. Recent advances in new methods for synaptic staining as well as new types of optical molecular probes with superior stability, specificity, and brightness provide new modalities for studying brain circuits. Here we review advanced methods and molecular probes for fluorescence staining of the synapses in the brain that are compatible with currently available expansion microscopy techniques. In particular, we will describe genetically encoded probes for synaptic labeling in mice, zebrafish, Drosophila fruit flies, and C. elegans, which enable the visualization of post-synaptic scaffolds and receptors, presynaptic terminals and vesicles, and even a snapshot of the synaptic activity itself. We will address current methods for applying these probes in ExM experiments, as well as appropriate vectors for the delivery of these molecular constructs. In addition, we offer experimental considerations and limitations for using each of these tools as well as our perspective on emerging tools.
Collapse
Affiliation(s)
- Madison A. Sneve
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, United States
| | - Kiryl D. Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
16
|
Hilton BJ, Husch A, Schaffran B, Lin TC, Burnside ER, Dupraz S, Schelski M, Kim J, Müller JA, Schoch S, Imig C, Brose N, Bradke F. An active vesicle priming machinery suppresses axon regeneration upon adult CNS injury. Neuron 2022; 110:51-69.e7. [PMID: 34706221 PMCID: PMC8730507 DOI: 10.1016/j.neuron.2021.10.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Axons in the adult mammalian central nervous system fail to regenerate after spinal cord injury. Neurons lose their capacity to regenerate during development, but the intracellular processes underlying this loss are unclear. We found that critical components of the presynaptic active zone prevent axon regeneration in adult mice. Transcriptomic analysis combined with live-cell imaging revealed that adult primary sensory neurons downregulate molecular constituents of the synapse as they acquire the ability to rapidly grow their axons. Pharmacogenetic reduction of neuronal excitability stimulated axon regeneration after adult spinal cord injury. Genetic gain- and loss-of-function experiments uncovered that essential synaptic vesicle priming proteins of the presynaptic active zone, but not clostridial-toxin-sensitive VAMP-family SNARE proteins, inhibit axon regeneration. Systemic administration of Baclofen reduced voltage-dependent Ca2+ influx in primary sensory neurons and promoted their regeneration after spinal cord injury. These findings indicate that functional presynaptic active zones constitute a major barrier to axon regeneration.
Collapse
Affiliation(s)
- Brett J Hilton
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Andreas Husch
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Barbara Schaffran
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Tien-Chen Lin
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Emily R Burnside
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Sebastian Dupraz
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Max Schelski
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Jisoo Kim
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Department of Stem Cell and Regenerative Biology, Center for Brain Science, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Susanne Schoch
- Institute of Neuropathology, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Cordelia Imig
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Frank Bradke
- Laboratory of Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
17
|
Spead O, Weaver CJ, Moreland T, Poulain FE. Live imaging of retinotectal mapping reveals topographic map dynamics and a previously undescribed role for Contactin 2 in map sharpening. Development 2021; 148:272618. [PMID: 34698769 DOI: 10.1242/dev.199584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022]
Abstract
Organization of neuronal connections into topographic maps is essential for processing information. Yet, our understanding of topographic mapping has remained limited by our inability to observe maps forming and refining directly in vivo. Here, we used Cre-mediated recombination of a new colorswitch reporter in zebrafish to generate the first transgenic model allowing the dynamic analysis of retinotectal mapping in vivo. We found that the antero-posterior retinotopic map forms early but remains dynamic, with nasal and temporal retinal axons expanding their projection domains over time. Nasal projections initially arborize in the anterior tectum but progressively refine their projection domain to the posterior tectum, leading to the sharpening of the retinotopic map along the antero-posterior axis. Finally, using a CRISPR-mediated mutagenesis approach, we demonstrate that the refinement of nasal retinal projections requires the adhesion molecule Contactin 2. Altogether, our study provides the first analysis of a topographic map maturing in real time in a live animal and opens new strategies for dissecting the molecular mechanisms underlying precise topographic mapping in vertebrates.
Collapse
Affiliation(s)
- Olivia Spead
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Cory J Weaver
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Trevor Moreland
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Fabienne E Poulain
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
18
|
Nunes AR, Gliksberg M, Varela SAM, Teles M, Wircer E, Blechman J, Petri G, Levkowitz G, Oliveira RF. Developmental Effects of Oxytocin Neurons on Social Affiliation and Processing of Social Information. J Neurosci 2021; 41:8742-8760. [PMID: 34470805 PMCID: PMC8528494 DOI: 10.1523/jneurosci.2939-20.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 11/21/2022] Open
Abstract
Hormones regulate behavior either through activational effects that facilitate the acute expression of specific behaviors or through organizational effects that shape the development of the nervous system thereby altering adult behavior. Much research has implicated the neuropeptide oxytocin (OXT) in acute modulation of various aspects of social behaviors across vertebrate species, and OXT signaling is associated with the developmental social deficits observed in autism spectrum disorders (ASDs); however, little is known about the role of OXT in the neurodevelopment of the social brain. We show that perturbation of OXT neurons during early zebrafish development led to a loss of dopaminergic neurons, associated with visual processing and reward, and blunted the neuronal response to social stimuli in the adult brain. Ultimately, adult fish whose OXT neurons were ablated in early life, displayed altered functional connectivity within social decision-making brain nuclei both in naive state and in response to social stimulus and became less social. We propose that OXT neurons have an organizational role, namely, to shape forebrain neuroarchitecture during development and to acquire an affiliative response toward conspecifics.SIGNIFICANCE STATEMENT Social behavior is developed over the lifetime of an organism and the neuropeptide oxytocin (OXT) modulates social behaviors across vertebrate species, and is associated with neuro-developmental social deficits such as autism. However, whether OXT plays a role in the developmental maturation of neural systems that are necessary for social behavior remains poorly explored. We show that proper behavioral and neural response to social stimuli depends on a developmental process orchestrated by OXT neurons. Animals whose OXT system is ablated in early life show blunted neuronal and behavioral responses to social stimuli as well as wide ranging disruptions in the functional connectivity of the social brain. We provide a window into the mechanisms underlying OXT-dependent developmental processes that implement adult sociality.
Collapse
Affiliation(s)
- Ana Rita Nunes
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Susana A M Varela
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- ISPA-Instituto Universitário, Lisboa 1149-041, Portugal
| | - Magda Teles
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
| | - Einav Wircer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Giovanni Petri
- Institute for Scientific Interchange (ISI) Foundation and ISI Global Science Foundation, Torino 10126, Italy
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rui F Oliveira
- Integrative Behavioural Biology Lab, Instituto Gulbenkian de Ciência, Oeiras 2780-156, Portugal
- ISPA-Instituto Universitário, Lisboa 1149-041, Portugal
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon 1400-038, Portugal
| |
Collapse
|
19
|
Correction to: Modeling Axonal Plasticity in Artificial Neural Networks. Neural Process Lett 2021. [DOI: 10.1007/s11063-021-10526-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Bodakuntla S, Nedozralova H, Basnet N, Mizuno N. Cytoskeleton and Membrane Organization at Axon Branches. Front Cell Dev Biol 2021; 9:707486. [PMID: 34540830 PMCID: PMC8440873 DOI: 10.3389/fcell.2021.707486] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Axon branching is a critical process ensuring a high degree of interconnectivity for neural network formation. As branching occurs at sites distant from the soma, it is necessary that axons have a local system to dynamically control and regulate axonal growth. This machinery depends on the orchestration of cellular functions such as cytoskeleton, subcellular transport, energy production, protein- and membrane synthesis that are adapted for branch formation. Compared to the axon shaft, branching sites show a distinct and dynamic arrangement of cytoskeleton components, endoplasmic reticulum and mitochondria. This review discusses the regulation of axon branching in the context of cytoskeleton and membrane remodeling.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hana Nedozralova
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nirakar Basnet
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
The RNA-binding protein Musashi controls axon compartment-specific synaptic connectivity through ptp69D mRNA poly(A)-tailing. Cell Rep 2021; 36:109713. [PMID: 34525368 DOI: 10.1016/j.celrep.2021.109713] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/24/2020] [Accepted: 08/24/2021] [Indexed: 10/20/2022] Open
Abstract
Synaptic targeting with subcellular specificity is essential for neural circuit assembly. Developing neurons use mechanisms to curb promiscuous synaptic connections and to direct synapse formation to defined subcellular compartments. How this selectivity is achieved molecularly remains enigmatic. Here, we discover a link between mRNA poly(A)-tailing and axon collateral branch-specific synaptic connectivity within the CNS. We reveal that the RNA-binding protein Musashi binds to the mRNA encoding the receptor protein tyrosine phosphatase Ptp69D, thereby increasing poly(A) tail length and Ptp69D protein levels. This regulation specifically promotes synaptic connectivity in one axon collateral characterized by a high degree of arborization and strong synaptogenic potential. In a different compartment of the same axon, Musashi prevents ectopic synaptogenesis, revealing antagonistic, compartment-specific functions. Moreover, Musashi-dependent Ptp69D regulation controls synaptic connectivity in the olfactory circuit. Thus, Musashi differentially shapes synaptic connectivity at the level of individual subcellular compartments and within different developmental and neuron type-specific contexts.
Collapse
|
22
|
Doshmanziari M, Shirian S, Kouchakian MR, Moniri SF, Jangnoo S, Mohammadi N, Zafari F. Mesenchymal stem cells act as stimulators of neurogenesis and synaptic function in a rat model of Alzheimer's disease. Heliyon 2021; 7:e07996. [PMID: 34589625 PMCID: PMC8461353 DOI: 10.1016/j.heliyon.2021.e07996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/17/2021] [Accepted: 09/10/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most common NDs leading to cognitive dysfunctions and dementia which are progressively worsen with age. Cell therapy is currently of particular interest in treatment of neurodegenerative disease (ND) such as AD. However, the effective treatment for AD is yet to be found. OBJECTIVE In this study, the possible roles of human umbilical mesnchymal stromal cord (hUMSCs) and adipose mesenchymal stem cells (hAD-MSCs) in neurogenesis and synaptic function were investigated using a β-amyloid 1-42 (β A42)-induced AD rat model. METHODS hUMSCs and hAD-MSCs were isolated from umbilical cord stroma and adipose tissue, respectively. The expression of Mesenchymal (CD73, CD90 and CD105) and hematopoietic (CD45 and CD133) markers of hUMSCs and hAD-MSCs were confirmed by flow cytometry. Alzheimer's rat model was created by β-amyloid 1-42 injection into the hippocampus and confirmed by Morris Water Maze and immunohistochemical staining. hUMSCs and hAD-MSCs were injected in Alzheimer's rat model, intravenously. Deposition of β-amyloid in the CA1 of hippocampus was assayed 3 months after cell administration. The expression of synaptophysin and GAP43 proteins was assessed by Western blot. Neural death was assessed by Nissl staining. RESULTS The data obtained from flow cytometry showed that surface mesenchymal and hematopoteic markers of the fibroblastic like cells isolated from adipose tissue and umbilical cord were expressed highly in hUMSCs and mostly in hAD-SCs. Transplantation of MSCs reduced β-amyloid deposition in the hippocampus of the AD rats compared to the β-amyloid group. The rate of neuronal cell death in the hippocampus of the β-amyloid-treated rats was significantly increased compared to that of the control group. The percentage of apoptotic cells in this group was 72.98 ± 1.25, which was significantly increased compared to the control group. Transplantation of either hUMSCs or hAD-SCs, respectively, resulted in a significant reduction in the apoptotic rate of the neuronal cells in the hippocampus by 39.47 ± 0.01 (p = 0.0001) and 43.23 ± 0.577 (p = 0.001) compared to the β-amyloid group. MSC transplantation resulted in a significant up-regulation in the expression levels of both synaptogenic (synaptophysin) and neurogenic markers (GAP43) by 1.289 ± 0.112 (P = 0.02) and 1.112 ± 0.106 (P = 0.005) fold in the hUMSCs-treated group and 1.174 ± 0.105 (P = 0.04) and 0.978 ± 0.167 (P = 0.008) fold in the hAD-SCs-treated group, respectively. CONCLUSION Intravenous injection of hUMSCs and hAD-MSCs is a safe approach that improves synaptic function and neurogenesis via up-regulation of synaptophysin and GAP43 protein expression levels, respectively, in Alzheimer's model. Intravenous injection of both applied SCs could improve learning and cognitive impairment induced by β A42 injection.
Collapse
Affiliation(s)
- Maryam Doshmanziari
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
- Shiraz Molecular Pathology Research Center, Dr. Daneshbod Pathology Laboratory, Shiraz, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mohammad-Reza Kouchakian
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Seyedeh Farzaneh Moniri
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | | | - Niloofar Mohammadi
- Addiction Department, School of Behavioral Sciences and Mental Health (Tehran Institute of Psychiatry), Iran University of Medical Sciences, Tehran, Iran
| | - Fariba Zafari
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
- Cellular and Molecular Research Center, Research Institute for Prevention of Non- Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
23
|
Chen W, Xie L, Yu F, Li Y, Chen C, Xie W, Huang T, Zhang Y, Zhang S, Li P. Zebrafish as a Model for In-Depth Mechanistic Study for Stroke. Transl Stroke Res 2021; 12:695-710. [PMID: 34050491 DOI: 10.1007/s12975-021-00907-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/22/2022]
Abstract
Stroke is one of the world's leading causes of death and disability, posing enormous burden to the society. However, the pathogenesis and mechanisms that underlie brain injury and brain repair remain largely unknown. There's an unmet need of in-depth mechanistic research in this field. Zebrafish (Danio rerio) is a powerful tool in brain science research mainly due to its small size and transparent body, high genome synteny with human, and similar nervous system structures. It can be used to establish both hemorrhagic and ischemic stroke models easily and effectively through different ways. After the establishment of stroke model, research methods including behavioral test, in vivo imaging, and drug screening are available to explore mechanisms that underlie the brain injury and brain repair after stroke. This review focuses on the advantages and the feasibility of zebrafish stroke model, and will also introduce the key methods available for stroke studies in zebrafish, which may drive future mechanistic studies in the pursuit of discovering novel therapeutic targets for stroke patients.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Fang Yu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Wanqing Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Tingting Huang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China
| | - Song Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine Shanghai Jiaotong University, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
24
|
Venkataramani V, Tanev DI, Kuner T, Wick W, Winkler F. Synaptic input to brain tumors: clinical implications. Neuro Oncol 2021; 23:23-33. [PMID: 32623467 DOI: 10.1093/neuonc/noaa158] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The recent discovery of synaptic connections between neurons and brain tumor cells fundamentally challenges our understanding of gliomas and brain metastases and shows how these tumors can integrate into complex neuronal circuits. Here, we provide an overview of glutamatergic neuron-to-brain tumor synaptic communication (NBTSC) and explore novel therapeutic avenues. First, we summarize current concepts of direct synaptic interactions between presynaptic neurons and postsynaptic glioma cells, and indirect perisynaptic input to metastatic breast cancer cells. We explain how these novel structures drive brain tumor growth and invasion. Second, a vicious cycle of enhanced neuronal activity, including tumor-related epilepsy, and glioma progression is described. Finally, we discuss which future avenues to target NBTSC appear most promising. All in all, further characterization of NBTSC and the exploration of NBTSC-inhibiting therapies have the potential to reveal critical vulnerabilities of yet incurable brain tumors.
Collapse
Affiliation(s)
- Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Dimitar Ivanov Tanev
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
25
|
|
26
|
Zhao S, Qian Y, Mu Y. Tracking single cells in zebrafish brain. J Neurosci Methods 2021; 353:109086. [PMID: 33508409 DOI: 10.1016/j.jneumeth.2021.109086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Changes in cell locations and morphologies shape the brain. Tracking single cells over time is a vital step to study these changes, but densely arranged brain cells impede such observation. Larval zebrafish has become a popular model animal for single-cell tracking, owing to its small, transparent brain and easy genetic manipulation. In this article, we review recent single-cell tracking studies on neurons and non-neuronal cells in the larval zebrafish brain, including soma migration, process refinement, and interactions among cell types. These findings yield new insights regarding how the translocation and morphological changes of individual cells determine brain function.
Collapse
Affiliation(s)
- Shan Zhao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yu Qian
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yu Mu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.
| |
Collapse
|
27
|
Kishore S, Cadoff EB, Agha MA, McLean DL. Orderly compartmental mapping of premotor inhibition in the developing zebrafish spinal cord. Science 2020; 370:431-436. [PMID: 33093104 DOI: 10.1126/science.abb4608] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
In vertebrates, faster movements involve the orderly recruitment of different types of spinal motor neurons. However, it is not known how premotor inhibitory circuits are organized to ensure alternating motor output at different movement speeds. We found that different types of commissural inhibitory interneurons in zebrafish form compartmental microcircuits during development that align inhibitory strength and recruitment order. Axonal microcircuits develop first and provide the most potent premotor inhibition during the fastest movements, followed by perisomatic microcircuits, and then dendritic microcircuits that provide the weakest inhibition during the slowest movements. The conversion of a temporal sequence of neuronal development into a spatial pattern of inhibitory connections provides an "ontogenotopic" solution to the problem of shaping spinal motor output at different speeds of movement.
Collapse
Affiliation(s)
- Sandeep Kishore
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Eli B Cadoff
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Moneeza A Agha
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - David L McLean
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
28
|
Induction of Short-Term Sensitization by an Aversive Chemical Stimulus in Zebrafish Larvae. eNeuro 2020; 7:ENEURO.0336-19.2020. [PMID: 33004417 PMCID: PMC7729299 DOI: 10.1523/eneuro.0336-19.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Larval zebrafish possess a number of molecular and genetic advantages for rigorous biological analyses of learning and memory. These advantages have motivated the search for novel forms of memory in these animals that can be exploited for understanding the cellular and molecular bases of vertebrate memory formation and consolidation. Here, we report a new form of behavioral sensitization in zebrafish larvae that is elicited by an aversive chemical stimulus [allyl isothiocyanate (AITC)] and that persists for ≥30 min. This form of sensitization is expressed as enhanced locomotion and thigmotaxis, as well as elevated heart rate. To characterize the neural basis of this nonassociative memory, we used transgenic zebrafish expressing the fluorescent calcium indicator GCaMP6 (Chen et al., 2013); because of the transparency of larval zebrafish, we could optically monitor neural activity in the brain of intact transgenic zebrafish before and after the induction of sensitization. We found a distinct brain area, previously linked to locomotion, that exhibited persistently enhanced neural activity following washout of AITC; this enhanced neural activity correlated with the behavioral sensitization. These results establish a novel form of memory in larval zebrafish and begin to unravel the neural basis of this memory.
Collapse
|
29
|
Adnan G, Rubikaite A, Khan M, Reber M, Suetterlin P, Hindges R, Drescher U. The GTPase Arl8B Plays a Principle Role in the Positioning of Interstitial Axon Branches by Spatially Controlling Autophagosome and Lysosome Location. J Neurosci 2020; 40:8103-8118. [PMID: 32917789 PMCID: PMC7574663 DOI: 10.1523/jneurosci.1759-19.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Interstitial axon branching is an essential step during the establishment of neuronal connectivity. However, the exact mechanisms on how the number and position of branches are determined are still not fully understood. Here, we investigated the role of Arl8B, an adaptor molecule between lysosomes and kinesins. In chick retinal ganglion cells (RGCs), downregulation of Arl8B reduces axon branch density and shifts their location more proximally, while Arl8B overexpression leads to increased density and more distal positions of branches. These alterations correlate with changes in the location and density of lysosomes and autophagosomes along the axon shaft. Diminishing autophagy directly by knock-down of atg7, a key autophagy gene, reduces branch density, while induction of autophagy by rapamycin increases axon branching, indicating that autophagy plays a prominent role in axon branch formation. In vivo, local inactivation of autophagy in the retina using a mouse conditional knock-out approach disturbs retino-collicular map formation which is dependent on the formation of interstitial axon branches. These data suggest that Arl8B plays a principal role in the positioning of axon branches by spatially controlling autophagy, thus directly controlling formation of neural connectivity in the brain.SIGNIFICANCE STATEMENT The formation of interstitial axonal branches plays a prominent role in numerous places of the developing brain during neural circuit establishment. We show here that the GTPase Arl8B controls density and location of interstitial axon branches, and at the same time controls also density and location of the autophagy machinery. Upregulation or downregulation of autophagy in vitro promotes or inhibits axon branching. Local disruption of autophagy in vivo disturbs retino-collicular mapping. Our data suggest that Arl8B controls axon branching by controlling locally autophagy. This work is one of the first reports showing a role of autophagy during early neural circuit development and suggests that autophagy in general plays a much more prominent role during brain development than previously anticipated.
Collapse
Affiliation(s)
- Gee Adnan
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Aine Rubikaite
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Moqadisa Khan
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Michael Reber
- Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada
| | - Philip Suetterlin
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
- Craniofacial Development and Stem Cell Biology, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Robert Hindges
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| | - Uwe Drescher
- Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| |
Collapse
|
30
|
Oprişoreanu AM, Smith HL, Arya S, Webster R, Zhong Z, Eaton-Hart C, Wehner D, Cardozo MJ, Becker T, Talbot K, Becker CG. Interaction of Axonal Chondrolectin with Collagen XIXa1 Is Necessary for Precise Neuromuscular Junction Formation. Cell Rep 2020; 29:1082-1098.e10. [PMID: 31665626 DOI: 10.1016/j.celrep.2019.09.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/13/2019] [Accepted: 09/12/2019] [Indexed: 01/05/2023] Open
Abstract
Chondrolectin (Chodl) is needed for motor axon extension in zebrafish and is dysregulated in mouse models of spinal muscular atrophy (SMA). However, the mechanistic basis of Chodl function is not known. Here, we use Chodl-deficient zebrafish and mouse mutants to show that the absence of Chodl leads to anatomical and functional defects of the neuromuscular synapse. In zebrafish, the growth of an identified motor axon beyond an "en passant" synapse and later axon branching from synaptic points are impaired, leading to functional deficits. Mechanistically, motor-neuron-autonomous Chodl function depends on its intracellular domain and on binding muscle-derived collagen XIXa1 by its extracellular C-type lectin domain. Our data support evolutionarily conserved roles of Chodl in synaptogenesis and provide evidence for a "synapse-first" scenario of motor axon growth in zebrafish.
Collapse
Affiliation(s)
- Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Hannah L Smith
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Sukrat Arya
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Richard Webster
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Zhen Zhong
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Charlotte Eaton-Hart
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Marcos J Cardozo
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK.
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
31
|
Mofatteh M. mRNA localization and local translation in neurons. AIMS Neurosci 2020; 7:299-310. [PMID: 32995487 PMCID: PMC7519968 DOI: 10.3934/neuroscience.2020016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022] Open
Abstract
The spatial and temporal regulation of gene expression in neurons is an important step in creating functional and structural neuronal networks. The complexity of neurons require differential expression of various proteins in different compartments. Highly polarised cells, such as neurons, respond rapidly to different external stimuli by changing their local protein abundance and composition. Neurons can have extensions up to a meter away from their cell body in humans, so it is easy to envisage why they need to manage the synthesis of new proteins locally and on-demand. Recent research has demonstrated that neurons can control the expression of different proteins by localising translationally silent mRNAs, followed by subsequent translation. Neurons use mRNA localization and local translation to achieve different purposes during their life cycle. While developing neurons rely on mRNA localization for axon guidance and synaptogenesis, mature neurons can use mRNA localization for maintenance of essential physiological processes. mRNA localization also plays a role in response to neuron injury to regenerate and restore neuronal connections. Recent microscopic imaging techniques such as live imaging of fluorescently tagged molecules combined with genetic and biochemical studies in neurons have illustrated evolutionarily conserved mechanisms for targeting mRNAs into their correct compartments. This review provides an overview of mRNA localization and local translation in vertebrate and invertebrate neurons and discusses the mechanism by which mRNAs are trafficked into axons. Furthermore, the role of mRNA localization in synaptic activation, as well as axonal injury is explored.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- Lincoln College, University of Oxford, Turl Street, Oxford, OX1 3DR, United Kingdom.,Merton College, University of Oxford, Merton Street, Oxford, OX1 4DJ, United Kingdom.,Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, United Kingdom
| |
Collapse
|
32
|
Hsieh JY, Ulrich BN, Issa FA, Lin MCA, Brown B, Papazian DM. Infant and adult SCA13 mutations differentially affect Purkinje cell excitability, maturation, and viability in vivo. eLife 2020; 9:57358. [PMID: 32644043 PMCID: PMC7386905 DOI: 10.7554/elife.57358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022] Open
Abstract
Mutations in KCNC3, which encodes the Kv3.3 K+ channel, cause spinocerebellar ataxia 13 (SCA13). SCA13 exists in distinct forms with onset in infancy or adulthood. Using zebrafish, we tested the hypothesis that infant- and adult-onset mutations differentially affect the excitability and viability of Purkinje cells in vivo during cerebellar development. An infant-onset mutation dramatically and transiently increased Purkinje cell excitability, stunted process extension, impaired dendritic branching and synaptogenesis, and caused rapid cell death during cerebellar development. Reducing excitability increased early Purkinje cell survival. In contrast, an adult-onset mutation did not significantly alter basal tonic firing in Purkinje cells, but reduced excitability during evoked high frequency spiking. Purkinje cells expressing the adult-onset mutation matured normally and did not degenerate during cerebellar development. Our results suggest that differential changes in the excitability of cerebellar neurons contribute to the distinct ages of onset and timing of cerebellar degeneration in infant- and adult-onset SCA13.
Collapse
Affiliation(s)
- Jui-Yi Hsieh
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brittany N Ulrich
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Fadi A Issa
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Meng-Chin A Lin
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Brandon Brown
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States
| | - Diane M Papazian
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Interdepartmental PhD Program in Molecular, Cellular, and Integrative Physiology, David Geffen School of Medicine at UCLA, Los Angeles, United States.,Brain Research Institute, UCLA, Los Angeles, United States.,Molecular Biology Institute, UCLA, Los Angeles, United States
| |
Collapse
|
33
|
Gallop J. Filopodia and their links with membrane traffic and cell adhesion. Semin Cell Dev Biol 2020; 102:81-89. [DOI: 10.1016/j.semcdb.2019.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 01/24/2023]
|
34
|
Pende M, Vadiwala K, Schmidbaur H, Stockinger AW, Murawala P, Saghafi S, Dekens MPS, Becker K, Revilla-i-Domingo R, Papadopoulos SC, Zurl M, Pasierbek P, Simakov O, Tanaka EM, Raible F, Dodt HU. A versatile depigmentation, clearing, and labeling method for exploring nervous system diversity. SCIENCE ADVANCES 2020; 6:eaba0365. [PMID: 32523996 PMCID: PMC7259959 DOI: 10.1126/sciadv.aba0365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/27/2020] [Indexed: 06/11/2023]
Abstract
Tissue clearing combined with deep imaging has emerged as a powerful alternative to classical histological techniques. Whereas current techniques have been optimized for imaging selected nonpigmented organs such as the mammalian brain, natural pigmentation remains challenging for most other biological specimens of larger volume. We have developed a fast DEpigmEntation-Plus-Clearing method (DEEP-Clear) that is easily incorporated in existing workflows and combines whole system labeling with a spectrum of detection techniques, ranging from immunohistochemistry to RNA in situ hybridization, labeling of proliferative cells (EdU labeling) and visualization of transgenic markers. With light-sheet imaging of whole animals and detailed confocal studies on pigmented organs, we provide unprecedented insight into eyes, whole nervous systems, and subcellular structures in animal models ranging from worms and squids to axolotls and zebrafish. DEEP-Clear thus paves the way for the exploration of species-rich clades and developmental stages that are largely inaccessible by regular imaging approaches.
Collapse
Affiliation(s)
- Marko Pende
- Department for Bioelectronics, FKE, Vienna University of Technology, Gußhausstraße 25-25A, building CH, 1040 Vienna, Austria
- Section for Bioelectronics, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Karim Vadiwala
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Hannah Schmidbaur
- Department of Neuroscience and Development, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Alexander W. Stockinger
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Prayag Murawala
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Saiedeh Saghafi
- Department for Bioelectronics, FKE, Vienna University of Technology, Gußhausstraße 25-25A, building CH, 1040 Vienna, Austria
| | - Marcus P. S. Dekens
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Klaus Becker
- Department for Bioelectronics, FKE, Vienna University of Technology, Gußhausstraße 25-25A, building CH, 1040 Vienna, Austria
- Section for Bioelectronics, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Roger Revilla-i-Domingo
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Sofia-Christina Papadopoulos
- Department for Bioelectronics, FKE, Vienna University of Technology, Gußhausstraße 25-25A, building CH, 1040 Vienna, Austria
| | - Martin Zurl
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Pawel Pasierbek
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Oleg Simakov
- Department of Neuroscience and Development, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Elly M. Tanaka
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Florian Raible
- Max Perutz Labs and Research Platform “Rhythms of Life”, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9/4, 1030 Vienna, Austria
| | - Hans-Ulrich Dodt
- Department for Bioelectronics, FKE, Vienna University of Technology, Gußhausstraße 25-25A, building CH, 1040 Vienna, Austria
- Section for Bioelectronics, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| |
Collapse
|
35
|
Sasaki Y. Local Translation in Growth Cones and Presynapses, Two Axonal Compartments for Local Neuronal Functions. Biomolecules 2020; 10:biom10050668. [PMID: 32344905 PMCID: PMC7277458 DOI: 10.3390/biom10050668] [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: 01/13/2020] [Revised: 04/08/2020] [Accepted: 04/21/2020] [Indexed: 01/02/2023] Open
Abstract
During neural development, growth cones, very motile compartments of tips of axons, lead axonal extension to the correct targets. Subsequently, presynapses, another axonal compartment with vigorous trafficking of synaptic vesicles, emerge to form functional synapses with postsynapses. In response to extracellular stimuli, the immediate supply of proteins by local translation within these two axonal compartments far from cell bodies confers high motility of growth cones and active vesicle trafficking in presynapses. Although local translation in growth cones and presynapses occurs at a very low level compared with cell bodies and even dendrites, recent progress in omics and visualization techniques with subcellular fractionation of these compartments has revealed the actual situation of local translation within these two axonal compartments. Here, the increasing evidence for local protein synthesis in growth cones and presynapses for axonal and synaptic functions has been reviewed. Furthermore, the mechanisms regulating local translation in these two compartments and pathophysiological conditions caused by dysregulated local translation are highlighted.
Collapse
Affiliation(s)
- Yukio Sasaki
- Functional Structure Biology Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
36
|
Jaimes LF, Mansk LMZ, Almeida-Santos AF, Pereira GS. Maturation of newborn neurons predicts social memory persistence in mice. Neuropharmacology 2020; 171:108102. [PMID: 32302616 DOI: 10.1016/j.neuropharm.2020.108102] [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: 11/11/2019] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 10/24/2022]
Abstract
Memory transience is essential to gain cognitive flexibility. Recently, hippocampal neurogenesis is emerging as one of the mechanisms involved in the balance between persistence and forgetting. Social recognition memory (SRM) has its duration prolonged by neurogenesis. However, it is still to be determined whether boosting neurogenesis in distinct phases of SRM may favor forgetting over persistence. In the present study, we used enriched environment (EE) and memantine (MEM) to increase neurogenesis. SRM was ubiquitously prolonged by both, while EE after the memory acquisition did not favor forgetting. Interestingly, the proportion of newborn neurons with mature morphology in the dorsal hippocampus was higher in animals where persistence prevailed. Finally, one of the main factors for dendritic growth is the formation of cytoskeleton. We found that Latrunculin A, an inhibitor of actin polymerization, blunted the promnesic effect of EE. Altogether, our results indicate that the mechanisms triggered by EE to improve SRM are not limited to increasing the number of newborn neurons.
Collapse
Affiliation(s)
- Laura F Jaimes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas, Gerais, Brazil
| | - Lara M Z Mansk
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas, Gerais, Brazil
| | - Ana F Almeida-Santos
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas, Gerais, Brazil
| | - Grace S Pereira
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas, Gerais, Brazil.
| |
Collapse
|
37
|
Functionally distinct subgroups of oligodendrocyte precursor cells integrate neural activity and execute myelin formation. Nat Neurosci 2020; 23:363-374. [PMID: 32066987 PMCID: PMC7292734 DOI: 10.1038/s41593-019-0581-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/20/2019] [Indexed: 01/06/2023]
Abstract
Recent reports have revealed oligodendrocyte precursor cell (OPC)
heterogeneity. It remains unclear if such heterogeneity reflects different
subtypes of cells with distinct functions, or rather transiently acquired states
of cells with the same function. By integrating lineage formation of individual
OPC clones, single-cell transcriptomics, calcium imaging and neural activity
manipulation, we show that OPCs in the zebrafish spinal cord can be divided into
two functionally distinct groups. One subgroup forms elaborate networks of
processes and exhibits a high degree of calcium signalling, but infrequently
differentiates, despite contact with permissive axons. Instead, these OPCs
divide in an activity and calcium dependent manner to produce another subgroup
with higher process motility and less calcium signaling, which readily
differentiates. Our data show that OPC subgroups are functionally diverse in
responding to neurons and reveal that activity regulates proliferation of a
subset of OPCs that is distinct from the cells that generate differentiated
oligodendrocytes.
Collapse
|
38
|
Martin-Peña A, Ferrus A. CCB is Involved in Actin-Based Axonal Transport of Selected Synaptic Proteins. J Neurosci 2020; 40:542-556. [PMID: 31754011 PMCID: PMC6961990 DOI: 10.1523/jneurosci.0915-18.2019] [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: 04/15/2018] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 11/21/2022] Open
Abstract
Synapse formation, maturation, and turnover require a finely regulated transport system that delivers selected cargos to specific synapses. However, the supporting mechanisms of this process are not fully understood. The present study unravels a new molecular system for vesicle-based axonal transport of proteins in male and female flies (Drosophila melanogaster). Here, we identify the gene CG14579 as the transcription unit corresponding to the regulatory mutations known as central complex broad (ccb). These mutations were previously isolated for their morphological phenotype in R-neurons of the ellipsoid body, a component of the central complex. Mutant axons from R-neurons fail to cross the midline, which is indicative of an aberrant composition of the growth cone. However, the molecular mechanism remained to be deciphered. In this manuscript, we show that CCB is involved in axonal trafficking of FasII and synaptobrevin, but not syntaxin. These results suggest that axonal transport of certain proteins is required for the correct pathfinding of R-neurons. We further investigated the molecular network supporting the CCB system and found that CCB colocalizes and coimmunoprecipitates with Rab11. Epistasis studies indicated that Rab11 is positioned downstream of CCB within this axonal transport system. Interestingly, ccb also interacts with actin and the actin nucleator spire The data revealed that this interaction plays a key role in the development of axonal connections within the ellipsoid body. We propose that the CCB/Rab11/SPIRE system regulates axonal trafficking of synaptic proteins required for proper connectivity and synaptic function.SIGNIFICANCE STATEMENT Proper function of the nervous system requires the establishment of mature, functional synapses. Differential protein composition in the synapse enables optimal performance of cognitive tasks. Therefore, it is critical to have a finely regulated transport system to deliver selected synaptic proteins to synapses. Remarkably, impairments in cytoskeleton-based protein-transport systems often underlie cognitive deficits, such as those associated with aging and neurodegenerative diseases. This study reveals that CCB is part of a novel transport system that delivers certain synaptic proteins via the actin cytoskeleton within the Rab11-related domain of slow recycling endosomes.
Collapse
Affiliation(s)
- Alfonso Martin-Peña
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Madrid 28002, Spain,
- Department of Neurology, and
- Center for Smell and Taste, McKnight Brain Institute, University of Florida, Gainesville, Florida 32611
| | - Alberto Ferrus
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Madrid 28002, Spain,
| |
Collapse
|
39
|
Cheresiz SV, Volgin AD, Kokorina Evsyukova A, Bashirzade AAO, Demin KA, de Abreu MS, Amstislavskaya TG, Kalueff AV. Understanding neurobehavioral genetics of zebrafish. J Neurogenet 2020; 34:203-215. [PMID: 31902276 DOI: 10.1080/01677063.2019.1698565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Due to its fully sequenced genome, high genetic homology to humans, external fertilization, fast development, transparency of embryos, low cost and active reproduction, the zebrafish (Danio rerio) has become a novel promising model organism in biomedicine. Zebrafish are a useful tool in genetic and neuroscience research, including linking various genetic mutations to brain mechanisms using forward and reverse genetics. These approaches have produced novel models of rare genetic CNS disorders and common brain illnesses, such as addiction, aggression, anxiety and depression. Genetically modified zebrafish also foster neuroanatomical studies, manipulating neural circuits and linking them to different behaviors. Here, we discuss recent advances in neurogenetics of zebrafish, and evaluate their unique strengths, inherent limitations and the rapidly growing potential for elucidating the conserved roles of genes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sergey V Cheresiz
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.,Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.,Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Alexandra Kokorina Evsyukova
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.,Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Alim A O Bashirzade
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.,Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil.,The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia.,Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia.,The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China.,Ural Federal University, Ekaterinburg, Russia.,Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.,Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| |
Collapse
|
40
|
An interhemispheric neural circuit allowing binocular integration in the optic tectum. Nat Commun 2019; 10:5471. [PMID: 31784529 PMCID: PMC6884480 DOI: 10.1038/s41467-019-13484-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 10/30/2019] [Indexed: 11/16/2022] Open
Abstract
Binocular stereopsis requires the convergence of visual information from corresponding points in visual space seen by two different lines of sight. This may be achieved by superposition of retinal input from each eye onto the same downstream neurons via ipsi- and contralaterally projecting optic nerve fibers. Zebrafish larvae can perceive binocular cues during prey hunting but have exclusively contralateral retinotectal projections. Here we report brain activity in the tectal neuropil ipsilateral to the visually stimulated eye, despite the absence of ipsilateral retinotectal projections. This activity colocalizes with arbors of commissural neurons, termed intertectal neurons (ITNs), that connect the tectal hemispheres. ITNs are GABAergic, establish tectal synapses bilaterally and respond to small moving stimuli. ITN-ablation impairs capture swim initiation when prey is positioned in the binocular strike zone. We propose an intertectal circuit that controls execution of the prey-capture motor program following binocular localization of prey, without requiring ipsilateral retinotectal projections. Zebrafish larvae can binocularly detect prey objects in order to strike but lack ipsilateral retinotectal fibers for binocular superposition of visual information. Here the authors describe commissural intertectal neurons and show that they are required for the initiation of capture strikes.
Collapse
|
41
|
DeMarco E, Xu N, Baier H, Robles E. Neuron types in the zebrafish optic tectum labeled by an id2b transgene. J Comp Neurol 2019; 528:1173-1188. [PMID: 31725916 DOI: 10.1002/cne.24815] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/31/2019] [Accepted: 11/06/2019] [Indexed: 01/30/2023]
Abstract
The larval zebrafish optic tectum has emerged as a prominent model for understanding how neural circuits control visually guided behaviors. Further advances in this area will require tools to monitor and manipulate tectal neurons with cell type specificity. Here, we characterize the morphology and neurotransmitter phenotype of tectal neurons labeled by an id2b:gal4 transgene. Whole-brain imaging of stable transgenic id2b:gal4 larvae revealed labeling in a subset of neurons in optic tectum, cerebellum, and hindbrain. Genetic mosaic labeling of single neurons within the id2b:gal4 expression pattern enabled us to characterize three tectal neuron types with distinct morphologies and connectivities. The first is a neuron type previously identified in the optic tectum of other teleost fish: the tectal pyramidal neuron (PyrN). PyrNs are local interneurons that form two stratified dendritic arbors and one stratified axonal arbor in the tectal neuropil. The second tectal neuron type labeled by the id2b:gal4 transgene is a projection neuron that forms a stratified dendritic arbor in the tectal neuropil and an axon that exits tectum to form a topographic projection to torus longitudinalis (TL). A third neuron type labeled is a projection neuron with a nonstratified dendritic arbor and a descending axonal projection to tegmentum. These findings establish the id2b:gal4 transgenic as a useful tool for future studies aimed at elucidating the functional role of tectum, TL, and tegmentum in visually guided behaviors.
Collapse
Affiliation(s)
- Elisabeth DeMarco
- Department of Biological Sciences and Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Nina Xu
- Department of Biological Sciences and Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Herwig Baier
- Max Planck Institute for Neurobiology, Martinsried, Germany
| | - Estuardo Robles
- Department of Biological Sciences and Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| |
Collapse
|
42
|
Kaneyama T, Shirasaki R. Post-crossing segment of dI1 commissural axons forms collateral branches to motor neurons in the developing spinal cord. J Comp Neurol 2019; 526:1943-1961. [PMID: 29752714 DOI: 10.1002/cne.24464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/30/2018] [Accepted: 05/03/2018] [Indexed: 11/09/2022]
Abstract
The dI1 commissural axons in the developing spinal cord, upon crossing the midline through the floor plate, make a sharp turn to grow rostrally. These post-crossing axons initially just extend adjacent to the floor plate without entering nearby motor columns. However, it remains poorly characterized how these post-crossing dI1 axons behave subsequently to this process. In the present study, to address this issue, we examined in detail the behavior of post-crossing dI1 axons in mice, using the Atoh1 enhancer-based conditional expression system that enables selective and sparse labeling of individual dI1 axons, together with Hb9 and ChAT immunohistochemistry for precise identification of spinal motor neurons (MNs). We found unexpectedly that the post-crossing segment of dI1 axons later gave off collateral branches that extended laterally to invade motor columns. Interestingly, these collateral branches emerged at around the time when their primary growth cones initiated invasion into motor columns. In addition, although the length of the laterally growing collateral branches increased with age, the majority of them remained within motor columns. Strikingly, these collateral branches further gave rise to multiple secondary branches in the region of MNs that innervate muscles close to the body axis. Moreover, these axonal branches formed presynaptic terminals on MNs. These observations demonstrate that dI1 commissural neurons develop axonal projection to spinal MNs via collateral branches arising later from the post-crossing segment of these axons. Our findings thus reveal a previously unrecognized projection of dI1 commissural axons that may contribute directly to generating proper motor output.
Collapse
Affiliation(s)
- Takeshi Kaneyama
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Shirasaki
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
43
|
Devine MJ, Kittler JT. Mitochondria at the neuronal presynapse in health and disease. Nat Rev Neurosci 2019; 19:63-80. [PMID: 29348666 DOI: 10.1038/nrn.2017.170] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Synapses enable neurons to communicate with each other and are therefore a prerequisite for normal brain function. Presynaptically, this communication requires energy and generates large fluctuations in calcium concentrations. Mitochondria are optimized for supplying energy and buffering calcium, and they are actively recruited to presynapses. However, not all presynapses contain mitochondria; thus, how might synapses with and without mitochondria differ? Mitochondria are also increasingly recognized to serve additional functions at the presynapse. Here, we discuss the importance of presynaptic mitochondria in maintaining neuronal homeostasis and how dysfunctional presynaptic mitochondria might contribute to the development of disease.
Collapse
Affiliation(s)
- Michael J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
44
|
Özel MN, Kulkarni A, Hasan A, Brummer J, Moldenhauer M, Daumann IM, Wolfenberg H, Dercksen VJ, Kiral FR, Weiser M, Prohaska S, von Kleist M, Hiesinger PR. Serial Synapse Formation through Filopodial Competition for Synaptic Seeding Factors. Dev Cell 2019; 50:447-461.e8. [PMID: 31353313 DOI: 10.1016/j.devcel.2019.06.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/15/2019] [Accepted: 06/21/2019] [Indexed: 11/15/2022]
Abstract
Following axon pathfinding, growth cones transition from stochastic filopodial exploration to the formation of a limited number of synapses. How the interplay of filopodia and synapse assembly ensures robust connectivity in the brain has remained a challenging problem. Here, we developed a new 4D analysis method for filopodial dynamics and a data-driven computational model of synapse formation for R7 photoreceptor axons in developing Drosophila brains. Our live data support a "serial synapse formation" model, where at any time point only 1-2 "synaptogenic" filopodia suppress the synaptic competence of other filopodia through competition for synaptic seeding factors. Loss of the synaptic seeding factors Syd-1 and Liprin-α leads to a loss of this suppression, filopodial destabilization, and reduced synapse formation. The failure to form synapses can cause the destabilization and secondary retraction of axon terminals. Our model provides a filopodial "winner-takes-all" mechanism that ensures the formation of an appropriate number of synapses.
Collapse
Affiliation(s)
- M Neset Özel
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany; Neuroscience Graduate Program, UT Southwestern Medical Center Dallas, Dallas, TX 75390, USA
| | - Abhishek Kulkarni
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Amr Hasan
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Josephine Brummer
- Department of Visual Data Analysis, Zuse Institute Berlin, 14195 Berlin, Germany
| | - Marian Moldenhauer
- Computational Medicine and Numerical Mathematics, Zuse Institute Berlin, 14195 Berlin, Germany; Department of Mathematics and Informatics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Ilsa-Maria Daumann
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Heike Wolfenberg
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Vincent J Dercksen
- Department of Visual Data Analysis, Zuse Institute Berlin, 14195 Berlin, Germany
| | - F Ridvan Kiral
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Martin Weiser
- Computational Medicine and Numerical Mathematics, Zuse Institute Berlin, 14195 Berlin, Germany
| | - Steffen Prohaska
- Department of Visual Data Analysis, Zuse Institute Berlin, 14195 Berlin, Germany
| | - Max von Kleist
- Department of Mathematics and Informatics, Freie Universität Berlin, 14195 Berlin, Germany.
| | - P Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany.
| |
Collapse
|
45
|
Bercier V, Hubbard JM, Fidelin K, Duroure K, Auer TO, Revenu C, Wyart C, Del Bene F. Dynactin1 depletion leads to neuromuscular synapse instability and functional abnormalities. Mol Neurodegener 2019; 14:27. [PMID: 31291987 PMCID: PMC6617949 DOI: 10.1186/s13024-019-0327-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dynactin subunit 1 is the largest subunit of the dynactin complex, an activator of the molecular motor protein complex dynein. Reduced levels of DCTN1 mRNA and protein have been found in sporadic amyotrophic lateral sclerosis (ALS) patients, and mutations have been associated with disease, but the role of this protein in disease pathogenesis is still unknown. METHODS We characterized a Dynactin1a depletion model in the zebrafish embryo and combined in vivo molecular analysis of primary motor neuron development with live in vivo axonal transport assays in single cells to investigate ALS-related defects. To probe neuromuscular junction (NMJ) function and organization we performed paired motor neuron-muscle electrophysiological recordings and GCaMP calcium imaging in live, intact larvae, and the synapse structure was investigated by electron microscopy. RESULTS Here we show that Dynactin1a depletion is sufficient to induce defects in the development of spinal cord motor neurons and in the function of the NMJ. We observe synapse instability, impaired growth of primary motor neurons, and higher failure rates of action potentials at the NMJ. In addition, the embryos display locomotion defects consistent with NMJ dysfunction. Rescue of the observed phenotype by overexpression of wild-type human DCTN1-GFP indicates a cell-autonomous mechanism. Synaptic accumulation of DCTN1-GFP, as well as ultrastructural analysis of NMJ synapses exhibiting wider synaptic clefts, support a local role for Dynactin1a in synaptic function. Furthermore, live in vivo analysis of axonal transport and cytoskeleton dynamics in primary motor neurons show that the phenotype reported here is independent of modulation of these processes. CONCLUSIONS Our study reveals a novel role for Dynactin1 in ALS pathogenesis, where it acts cell-autonomously to promote motor neuron synapse stability independently of dynein-mediated axonal transport.
Collapse
Affiliation(s)
- Valérie Bercier
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
- Present Address: VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
| | - Jeffrey M. Hubbard
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
| | - Kevin Fidelin
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
- Present Address: Zuckerman Mind Brain Behavior Institute, Columbia University, New York, USA
| | - Karine Duroure
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| | - Thomas O. Auer
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
- Present Address: Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Céline Revenu
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| | - Claire Wyart
- Sorbonne Université, Inserm, CNRS, AP-HP, Institut du Cerveau et de la Moelle Épinière, ICM, F-75013 Paris, France
| | - Filippo Del Bene
- Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, Sorbonne Université, F-75005 Paris, France
| |
Collapse
|
46
|
Kramer A, Wu Y, Baier H, Kubo F. Neuronal Architecture of a Visual Center that Processes Optic Flow. Neuron 2019; 103:118-132.e7. [PMID: 31147153 DOI: 10.1016/j.neuron.2019.04.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/17/2019] [Accepted: 04/10/2019] [Indexed: 01/23/2023]
Abstract
Animals use global image motion cues to actively stabilize their position by compensatory movements. Neurons in the zebrafish pretectum distinguish different optic flow patterns, e.g., rotation and translation, to drive appropriate behaviors. Combining functional imaging and morphological reconstruction of single cells, we revealed critical neuroanatomical features of this sensorimotor transformation. Terminals of direction-selective retinal ganglion cells (DS-RGCs) are located within the pretectal retinal arborization field 5 (AF5), where they meet dendrites of pretectal neurons with simple tuning to monocular optic flow. Translation-selective neurons, which respond selectively to optic flow in the same direction for both eyes, are intermingled with these simple cells but do not receive inputs from DS-RGCs. Mutually exclusive populations of pretectal projection neurons innervate either the reticular formation or the cerebellum, which in turn control motor responses. We posit that local computations in a defined pretectal circuit transform optic flow signals into neural commands driving optomotor behavior. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Anna Kramer
- Department Genes - Circuits - Behavior, Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Yunmin Wu
- Department Genes - Circuits - Behavior, Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Herwig Baier
- Department Genes - Circuits - Behavior, Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Fumi Kubo
- Department Genes - Circuits - Behavior, Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany; Center for Frontier Research, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan.
| |
Collapse
|
47
|
Urwyler O, Izadifar A, Vandenbogaerde S, Sachse S, Misbaer A, Schmucker D. Branch-restricted localization of phosphatase Prl-1 specifies axonal synaptogenesis domains. Science 2019; 364:364/6439/eaau9952. [DOI: 10.1126/science.aau9952] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/29/2019] [Indexed: 01/21/2023]
Abstract
Central nervous system (CNS) circuit development requires subcellular control of synapse formation and patterning of synapse abundance. We identified the Drosophila membrane-anchored phosphatase of regenerating liver (Prl-1) as an axon-intrinsic factor that promotes synapse formation in a spatially restricted fashion. The loss of Prl-1 in mechanosensory neurons reduced the number of CNS presynapses localized on a single axon collateral and organized as a terminal arbor. Flies lacking all Prl-1 protein had locomotor defects. The overexpression of Prl-1 induced ectopic synapses. In mechanosensory neurons, Prl-1 modulates the insulin receptor (InR) signaling pathway within a single contralateral axon compartment, thereby affecting the number of synapses. The axon branch–specific localization and function of Prl-1 depend on untranslated regions of the prl-1 messenger RNA (mRNA). Therefore, compartmentalized restriction of Prl-1 serves as a specificity factor for the subcellular control of axonal synaptogenesis.
Collapse
|
48
|
Bello-Rojas S, Istrate AE, Kishore S, McLean DL. Central and peripheral innervation patterns of defined axial motor units in larval zebrafish. J Comp Neurol 2019; 527:2557-2572. [PMID: 30919953 DOI: 10.1002/cne.24689] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Spinal motor neurons and the peripheral muscle fibers they innervate form discrete motor units that execute movements of varying force and speed. Subsets of spinal motor neurons also exhibit axon collaterals that influence motor output centrally. Here, we have used in vivo imaging to anatomically characterize the central and peripheral innervation patterns of axial motor units in larval zebrafish. Using early born "primary" motor neurons and their division of epaxial and hypaxial muscle into four distinct quadrants as a reference, we define three distinct types of later born "secondary" motor units. The largest is "m-type" units, which innervate deeper fast-twitch muscle fibers via medial nerves. Next in size are "ms-type" secondaries, which innervate superficial fast-twitch and slow fibers via medial and septal nerves, followed by "s-type" units, which exclusively innervate superficial slow muscle fibers via septal nerves. All types of secondaries innervate up to four axial quadrants. Central axon collaterals are found in subsets of primaries based on soma position and predominantly in secondary fast-twitch units (m, ms) with increasing likelihood based on number of quadrants innervated. Collaterals are labeled by synaptophysin-tagged fluorescent proteins, but not PSD95, consistent with their output function. Also, PSD95 dendrite labeling reveals that larger motor units receive more excitatory synaptic input. Collaterals are largely restricted to the neuropil, however, perisomatic connections are observed between motor units. These observations suggest that recurrent interactions are dominated by motor neurons recruited during stronger movements and set the stage for functional investigations of recurrent motor circuitry in larval zebrafish.
Collapse
Affiliation(s)
- Saul Bello-Rojas
- Interdepartmental Neuroscience Postbaccalaureate Research Education Program, Northwestern University, Evanston, Illinois
| | - Ana E Istrate
- Masters Program in Neurobiology, Northwestern University, Evanston, Illinois
| | - Sandeep Kishore
- Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - David L McLean
- Interdepartmental Neuroscience Postbaccalaureate Research Education Program, Northwestern University, Evanston, Illinois.,Masters Program in Neurobiology, Northwestern University, Evanston, Illinois.,Department of Neurobiology, Northwestern University, Evanston, Illinois
| |
Collapse
|
49
|
Tanimura A, Du Y, Kondapalli J, Wokosin DL, Surmeier DJ. Cholinergic Interneurons Amplify Thalamostriatal Excitation of Striatal Indirect Pathway Neurons in Parkinson’s Disease Models. Neuron 2019; 101:444-458.e6. [DOI: 10.1016/j.neuron.2018.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 09/13/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022]
|
50
|
Razetti A, Medioni C, Malandain G, Besse F, Descombes X. A stochastic framework to model axon interactions within growing neuronal populations. PLoS Comput Biol 2018; 14:e1006627. [PMID: 30507939 PMCID: PMC6292646 DOI: 10.1371/journal.pcbi.1006627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/13/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
The confined and crowded environment of developing brains imposes spatial constraints on neuronal cells that have evolved individual and collective strategies to optimize their growth. These include organizing neurons into populations extending their axons to common target territories. How individual axons interact with each other within such populations to optimize innervation is currently unclear and difficult to analyze experimentally in vivo. Here, we developed a stochastic model of 3D axon growth that takes into account spatial environmental constraints, physical interactions between neighboring axons, and branch formation. This general, predictive and robust model, when fed with parameters estimated on real neurons from the Drosophila brain, enabled the study of the mechanistic principles underlying the growth of axonal populations. First, it provided a novel explanation for the diversity of growth and branching patterns observed in vivo within populations of genetically identical neurons. Second, it uncovered that axon branching could be a strategy optimizing the overall growth of axons competing with others in contexts of high axonal density. The flexibility of this framework will make it possible to investigate the rules underlying axon growth and regeneration in the context of various neuronal populations. Understanding how neuronal cells establish complex circuits with specific functions within a developing brain is a major current challenge. Over the last past years, enormous progress has been done to precisely resolve brain anatomy and to dissect the mechanisms controlling the establishment of precise neuronal networks. However, due to the extreme complexity of the brain, it is still experimentally difficult to investigate in vivo how neurons interact with each other and with their physical environments to innervate target territories during development. Here, we have developed a framework that integrates a dynamic 3D mathematical model of single axonal growth with parameters estimated from neurons grown in vivo and simulations of entire populations of growing axons. The emergent properties of our model enable the study of the mechanistic principles underlying the growth of axonal population in developing brains. Specifically, our results highlight the impact of mechanical interactions on both individual and collective axon growth, and uncover how branching regulate this process.
Collapse
|