1
|
Xu L, Sheng D, He R, Meng Y, Tian L, Luo Y, Wang Y, Aizemaiti R, An Z, Wang Y. Developmental and neurobehavioral toxicity of benzotriazole ultraviolet stabilizer UV-360 on zebrafish larvae. PLoS One 2025; 20:e0324355. [PMID: 40408449 PMCID: PMC12101659 DOI: 10.1371/journal.pone.0324355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
The presence of UV-360, a commonly utilized benzotriazole ultraviolet stabilizer, has been frequently detected in diverse environments and organisms. However, existing knowledge regarding the potential impacts of UV-360 exposure on organisms remains limited. To evaluate the influence of UV-360 exposure on zebrafish during their initial developmental phases. The study began with an assessment of the developmental impact of UV-360 on larval stages. Subsequently, the investigation focused on examining its effects on locomotor behaviors. Additionally, analyses were conducted on neuronal development, the expression of genes associated with neurotoxicity, and electrophysiological recordings. Finally, the research extended to an exploration of transcriptome-level gene expression profiles. Exposure to UV-360 exhibited significant adverse effects on larvae, evidenced by a marked reduction in hatching rate, decreased heart rate, and impaired development of total body length. Furthermore, UV-360 exposure induced notable behavioral alterations, malformations in spinal motor neuron axons, and a substantial decrease in both the area and volume of these axons. Additionally, the expression of neurotoxicity-related genes and electrophysiological spike activity were significantly altered by UV-360 exposure. Lastly, exposure to UV-360 triggered significant modifications in the transcriptomic profile of zebrafish larvae, with a considerable proportion of differentially expressed genes associated with signal transduction processes and the neuroactive ligand-receptor interaction pathway. The results of this study revealed a dose-dependent developmental and neurobehavioral toxicity associated with UV-360 exposure in zebrafish larvae. The observed modifications in neuroactive ligand-receptors and disruptions in neurotransmitter systems suggested a potential mechanism for the neurotoxicity induced by UV-360 exposure in zebrafish larvae. These findings contribute significantly to the understanding of the toxicological effects of UV-360 on zebrafish larvae and provide strong evidence to help clarify the mechanisms of UV-360-induced toxicity.
Collapse
Affiliation(s)
- Lihan Xu
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- College of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Donglai Sheng
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
| | - Rong He
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- College of Stomatology, Hangzhou Normal University, Hangzhou, China
| | - Yanlong Meng
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, China
| | - Lili Tian
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- Pharmacy of Traditional Chinese Medicine, Zhejiang Hospital, Hangzhou, China
| | - Yuhao Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Yingjia Wang
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, College of Basic Medical, Hangzhou Normal University, Hangzhou, China
| | - Rusidanmu Aizemaiti
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhou An
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuying Wang
- Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Science, Hangzhou Normal University, Hangzhou, China
- College of Stomatology, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
2
|
Henríquez JP, Bermedo-García F, Zelada D, Mella J. Integrating postsynaptic morphology and dynamics to evaluate neuromuscular synapse status: Insights from α-bungarotoxin. Toxicon 2025; 262:108404. [PMID: 40354828 DOI: 10.1016/j.toxicon.2025.108404] [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: 01/18/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The neuromuscular junction (NMJ) is a crucial peripheral synapse that controls muscle contraction. It consists of a presynaptic motor terminal, a postsynaptic muscle domain, and associated cells, such as terminal Schwann cells and kranocytes. Its larger size compared to central synapses has allowed detailed analyses of NMJ morphology that have been widely used as a reliable parameter of synaptic formation, maturation, function, and decline. Due to its high affinity for postsynaptic acetylcholine receptors (AChRs), the snake venom-derived α-bungarotoxin (BTX) has been pivotal in advancing our understanding of NMJ organization, enabling a detailed mapping of postsynaptic morphologies associated to distinct functional outcomes. Although certain morphological features are often associated with NMJ worsening, some of these cellular changes also occur in biological contexts where synaptic function remains intact. In this review, we draw on previous studies and our recent findings using BTX-based pulse-chase assays to suggest that combining morphological analyses with assessments of postsynaptic stability offers a more comprehensive understanding of NMJ function and regenerative potential. We propose that integrating diverse BTX-based tools into studies of NMJ morphology and stability will provide particularly valuable insights in contexts such as aging, injury, and neuromuscular diseases, where these combined parameters may serve as robust predictors of functional outcomes.
Collapse
Affiliation(s)
- Juan Pablo Henríquez
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile; Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, 4070386, Concepción, Chile.
| | - Francisca Bermedo-García
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile
| | - Diego Zelada
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, 4070386, Concepción, Chile
| | - Jessica Mella
- Neuromuscular Studies Lab (NeSt Lab), Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, 5110566, Valdivia, Chile
| |
Collapse
|
3
|
Twaddle NC, Silva CS, Beland FA, Kanungo J. Acetyl L-Carnitine Protects Zebrafish Embryos From Verapamil and Inorganic Arsenic-Induced Cardiotoxicity and Developmental Toxicity With No Effect on Supernumerary Motor Neuron Development. J Appl Toxicol 2025. [PMID: 40234040 DOI: 10.1002/jat.4788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/17/2025]
Abstract
Verapamil (a P-glycoprotein inhibitor) and inorganic arsenic cotreatment has been shown to be toxic in chick cardiomyocytes. Previously, we have shown that sodium arsenite at 200 mg/L did not cause developmental toxicity or cardiotoxicity in zebrafish embryos. Here, we investigated the effect of verapamil and sodium arsenite cotreatment on the zebrafish embryos. Embryos at 5 h post-fertilization (hpf) were exposed to sodium arsenite (100-400 mg/L; 0.77-3.08 mM) in the presence or absence of 20 μM verapamil for 67 h. At 72 hpf, all the embryos treated with sodium arsenite or verapamil alone were alive, while only ~23% and ~17% survived in the groups cotreated with 20 μM verapamil and 100 mg/L or 200 mg/L arsenite, respectively. However, 10 μM of verapamil and 200 mg/L sodium arsenite cotreatment resulted in 100% embryo survival. Inductively coupled plasma mass spectrometry analysis showed that in the verapamil and sodium arsenite cotreated group, the internal arsenic concentration was significantly higher than in the group treated with only sodium arsenite, suggesting that verapamil inhibited arsenic efflux. Surprisingly, verapamil, a calcium channel blocker, reduced sodium arsenite-induced apoptosis but caused developmental toxicity and cardiotoxicity in the sodium arsenite cotreated embryos, without affecting arsenite-induced supernumerary motor neuron development. Furthermore, acetyl L-carnitine (ALCAR) completely abolished both developmental toxicity and cardiotoxicity induced by sodium arsenite and verapamil cotreatment. We show for the first time that ALCAR prevents toxicities induced by arsenic and verapamil cotreatment in zebrafish embryos, a vertebrate model for investigating chemical toxicity.
Collapse
Affiliation(s)
- Nathan C Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Camila S Silva
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
4
|
Chakraborty G, Joshi B, Ahire K, Patra C. Tributyl phosphate inhibits neurogenesis and motor functions during embryonic development in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 279:107203. [PMID: 39667268 DOI: 10.1016/j.aquatox.2024.107203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Tributyl phosphate (TBP), an organophosphate ester (OPE), is heavily used as a solvent in chemical industries, a plasticizer, and to extract radioactive molecules. Thus, widespread uses of TBP in industrialized countries led to the release of TBP and its metabolites, dibutyl phosphate (DBP) and monobutyl phosphate (MBP), in the environment and were detected in human samples. Accumulating these OPEs over time in humans and aquatic animals may develop toxicological effects. The reports also say TBP passes through the mother-fetal transmission route and may affect embryonic development. However, the impact of TBP and its metabolites on vertebrate development has been poorly studied. Ex-utero development, high fecundity, and optical transparency make the zebrafish a preferred model for toxicological evaluation. Thus, we aim to explore the toxic effects of TBP and its metabolites on aquatic animals using zebrafish as a model organism. Embryos in the chorion were incubated in 10-60 µM test chemicals from 6 to 48 h post fertilization (hpf), and analyzed the adverse effects on embryos. Our study found that 10-20 µM TBP inhibits neural growth, resulting in decreased spontaneous movement frequency and locomotive behavior without altering the overall embryonic growth and muscle functions. In contrast, DBP-treated embryos showed increased spontaneous movement frequency without changing the motor neuron growth and locomotive behavior. Further, in higher concentrations, TBP is teratogenic, and DBP is lethal to the embryos. Altogether, we found that TBP inhibits neurogenesis and motor behavior; however, its metabolite DBP is neuroexcitatory in zebrafish embryos.
Collapse
Affiliation(s)
- Gourav Chakraborty
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India
| | - Bhagyashri Joshi
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India
| | - Kedar Ahire
- Department of Zoology, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra 411004, India.
| |
Collapse
|
5
|
Ten Martin D, Jardin N, Vougny J, Giudicelli F, Gasmi L, Berbée N, Henriot V, Lebrun L, Haumaître C, Kneussel M, Nicol X, Janke C, Magiera MM, Hazan J, Fassier C. Tubulin glutamylation regulates axon guidance via the selective tuning of microtubule-severing enzymes. EMBO J 2025; 44:107-140. [PMID: 39613968 PMCID: PMC11695996 DOI: 10.1038/s44318-024-00307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 12/01/2024] Open
Abstract
The microtubule cytoskeleton is a major driving force of neuronal circuit development. Fine-tuned remodelling of this network by selective activation of microtubule-regulating proteins, including microtubule-severing enzymes, has emerged as a central process in neuronal wiring. Tubulin posttranslational modifications control both microtubule properties and the activities of their interacting proteins. However, whether and how tubulin posttranslational modifications may contribute to neuronal connectivity has not yet been addressed. Here we show that the microtubule-severing proteins p60-katanin and spastin play specific roles in axon guidance during zebrafish embryogenesis and identify a key role for tubulin polyglutamylation in their functional specificity. Furthermore, our work reveals that polyglutamylases with undistinguishable activities in vitro, TTLL6 and TTLL11, play exclusive roles in motor circuit wiring by selectively tuning p60-katanin- and spastin-driven motor axon guidance. We confirm the selectivity of TTLL11 towards spastin regulation in mouse cortical neurons and establish its relevance in preventing axonal degeneration triggered by spastin haploinsufficiency. Our work thus provides mechanistic insight into the control of microtubule-driven neuronal development and homeostasis and opens new avenues for developing therapeutic strategies in spastin-associated hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Daniel Ten Martin
- Sorbonne Université, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France
| | - Nicolas Jardin
- Sorbonne Université, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France
| | - Juliette Vougny
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012 Paris, France
| | - François Giudicelli
- Institut de Biologie de l'École Normale Supérieure, ENS, CNRS UMR8197, INSERM U1024, Paris, France
| | - Laïla Gasmi
- Sorbonne Université, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France
| | - Naomi Berbée
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012 Paris, France
- Amsterdam University Medical Center (UMC), University of Amsterdam (UvA), Amsterdam, The Netherlands
| | - Véronique Henriot
- Institut Curie, Université PSL, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Laura Lebrun
- Institut Curie, Université PSL, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Cécile Haumaître
- Université Paris Diderot, INSERM UMR1149, ERL CNRS 8252, Paris, France
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Xavier Nicol
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012 Paris, France
| | - Carsten Janke
- Institut Curie, Université PSL, CNRS UMR3348, Orsay, France
- Université Paris-Saclay, CNRS UMR3348, Orsay, France
| | - Maria M Magiera
- Institut Curie, Université PSL, CNRS UMR3348, Orsay, France.
- Université Paris-Saclay, CNRS UMR3348, Orsay, France.
| | - Jamilé Hazan
- Sorbonne Université, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France.
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR7241, INSERM U1050, Paris, France.
| | - Coralie Fassier
- Sorbonne Université, INSERM U1130, CNRS UMR8246, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), Paris, France.
- Sorbonne Université, CNRS, Inserm, Institut de la Vision, F-75012 Paris, France.
| |
Collapse
|
6
|
Lee BC, Tsai JC, Huang YH, Wang CC, Lee HC, Tsai HJ. The 419th Aspartic Acid of Neural Membrane Protein Enolase 2 Is a Key Residue Involved in the Axonal Growth of Motor Neurons Mediated by Interaction between Enolase 2 Receptor and Extracellular Pgk1 Ligand. Int J Mol Sci 2024; 25:10753. [PMID: 39409082 PMCID: PMC11477227 DOI: 10.3390/ijms251910753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Neuron-specific Enolase 2 (Eno2) is an isozyme primarily distributed in the central and peripheral nervous systems and neuroendocrine cells. It promotes neuronal survival, differentiation, and axonal regeneration. Recent studies have shown that Eno2 localized on the cell membrane of motor neurons acts as a receptor for extracellular phosphoglycerate kinase 1 (ePgk1), which is secreted by muscle cells and promotes the neurite outgrowth of motor neurons (NOMN). However, interaction between Eno1, another isozyme of Enolase, and ePgk1 failed to return the same result. To account for the difference, we constructed seven point-mutations of Eno2, corresponding to those of Eno1, and verified their effects on NOMN. Among the seven Eno2 mutants, eno2-siRNA-knockdown NSC34 cells transfected with plasmid encoding the 419th aspartic acid mutated into serine (Eno2-[D419S]) or Eno2-[E420K] showed a significant reduction in neurite length. Moreover, the Eno2-ePgk1-interacted synergic effect on NOMN driven by Eno2-[D419S] was more profoundly reduced than that driven by Eno2-[E420K], suggesting that D419 was the more essential residue involved in NOMN mediated by Eno2-ePgk1 interaction. Eno2-ePgk1-mediated NOMN appeared to increase the level of p-Cofilin, a growth cone collapse marker, in NSC34 cells transfected with Eno2-[D419S] and incubated with ePgk1, thereby inhibiting NOMN. Furthermore, we conducted in vivo experiments using zebrafish transgenic line Tg(mnx1:GFP), in which GFP is tagged in motor neurons. In the presence of ePgk1, the retarded growth of axons in embryos injected with eno2-specific antisense morpholino oligonucleotides (MO) could be rescued by wobble-eno2-mRNA. However, despite the addition of ePgk1, the decreased defective axons and the increased branched neurons were not significantly improved in the eno2-[D419S]-mRNA-injected embryos. Collectively, these results lead us to suggest that the 419th aspartic acid of mouse Eno2 is likely a crucial site affecting motor neuron development mediated by Eno2-ePgk1 interaction, and, hence, mutations result in a significant reduction in the degree of NOMN in vitro and axonal growth in vivo.
Collapse
Affiliation(s)
- Bing-Chang Lee
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (B.-C.L.); (Y.-H.H.); (H.-C.L.)
| | - Jui-Che Tsai
- Graduate Institute of Applied Science and Engineering, Fu Jen Catholic University, New Taipei City 242062, Taiwan;
| | - Yi-Hsin Huang
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (B.-C.L.); (Y.-H.H.); (H.-C.L.)
| | - Chun-Cheng Wang
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan;
| | - Hung-Chieh Lee
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (B.-C.L.); (Y.-H.H.); (H.-C.L.)
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan; (B.-C.L.); (Y.-H.H.); (H.-C.L.)
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei 10617, Taiwan;
| |
Collapse
|
7
|
de Sena-Tomás C, Rebola Lameira L, Rebocho da Costa M, Naique Taborda P, Laborde A, Orger M, de Oliveira S, Saúde L. Neutrophil immune profile guides spinal cord regeneration in zebrafish. Brain Behav Immun 2024; 120:514-531. [PMID: 38925414 DOI: 10.1016/j.bbi.2024.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/15/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Spinal cord injury triggers a strong innate inflammatory response in both non-regenerative mammals and regenerative zebrafish. Neutrophils are the first immune population to be recruited to the injury site. Yet, their role in the repair process, particularly in a regenerative context, remains largely unknown. Here, we show that, following rapid recruitment to the injured spinal cord, neutrophils mostly reverse migrate throughout the zebrafish body. In addition, promoting neutrophil inflammation resolution by inhibiting Cxcr4 boosts cellular and functional regeneration. Neutrophil-specific RNA-seq analysis reveals an enhanced activation state that correlates with a transient increase in tnf-α expression in macrophage/microglia populations. Conversely, blocking neutrophil recruitment through Cxcr1/2 inhibition diminishes the presence of macrophage/microglia at the injury site and impairs spinal cord regeneration. Altogether, these findings provide new insights into the role of neutrophils in spinal cord regeneration, emphasizing the significant impact of their immune profile on the outcome of the repair process.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Leonor Rebola Lameira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mariana Rebocho da Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Patrícia Naique Taborda
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Alexandre Laborde
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Michael Orger
- Champalimaud Research, Champalimaud Centre for the Unknown, 1400-038 Lisboa, Portugal
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA; Harold and Muriel Block Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Montefiore-Einstein Comprehensive Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Histologia e Biologia de Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
8
|
Miles KD, Barker CM, Russell KP, Appel BH, Doll CA. Electrical Synapses Mediate Embryonic Hyperactivity in a Zebrafish Model of Fragile X Syndrome. J Neurosci 2024; 44:e2275232024. [PMID: 38969506 PMCID: PMC11293453 DOI: 10.1523/jneurosci.2275-23.2024] [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: 12/06/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024] Open
Abstract
Although hyperactivity is associated with a wide variety of neurodevelopmental disorders, the early embryonic origins of locomotion have hindered investigation of pathogenesis of these debilitating behaviors. The earliest motor output in vertebrate animals is generated by clusters of early-born motor neurons (MNs) that occupy distinct regions of the spinal cord, innervating stereotyped muscle groups. Gap junction electrical synapses drive early spontaneous behavior in zebrafish, prior to the emergence of chemical neurotransmitter networks. We use a genetic model of hyperactivity to gain critical insight into the consequences of errors in motor circuit formation and function, finding that Fragile X syndrome model mutant zebrafish are hyperexcitable from the earliest phases of spontaneous behavior, show altered sensitivity to blockade of electrical gap junctions, and have increased expression of the gap junction protein Connexin 34/35. We further show that this hyperexcitable behavior can be rescued by pharmacological inhibition of electrical synapses. We also use functional imaging to examine MN and interneuron (IN) activity in early embryogenesis, finding genetic disruption of electrical gap junctions uncouples activity between mnx1 + MNs and INs. Taken together, our work highlights the importance of electrical synapses in motor development and suggests that the origins of hyperactivity in neurodevelopmental disorders may be established during the initial formation of locomotive circuits.
Collapse
Affiliation(s)
- Kaleb D Miles
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Chase M Barker
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristen P Russell
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Bruce H Appel
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| | - Caleb A Doll
- Section of Developmental Biology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
9
|
Lukowicz-Bedford RM, Eisen JS, Miller AC. Gap-junction-mediated bioelectric signaling required for slow muscle development and function in zebrafish. Curr Biol 2024; 34:3116-3132.e5. [PMID: 38936363 PMCID: PMC11265983 DOI: 10.1016/j.cub.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/11/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024]
Abstract
Bioelectric signaling, intercellular communication facilitated by membrane potential and electrochemical coupling, is emerging as a key regulator of animal development. Gap junction (GJ) channels can mediate bioelectric signaling by creating a fast, direct pathway between cells for the movement of ions and other small molecules. In vertebrates, GJ channels are formed by a highly conserved transmembrane protein family called the connexins. The connexin gene family is large and complex, creating challenges in identifying specific connexins that create channels within developing and mature tissues. Using the embryonic zebrafish neuromuscular system as a model, we identify a connexin conserved across vertebrate lineages, gjd4, which encodes the Cx46.8 protein, that mediates bioelectric signaling required for slow muscle development and function. Through mutant analysis and in vivo imaging, we show that gjd4/Cx46.8 creates GJ channels specifically in developing slow muscle cells. Using genetics, pharmacology, and calcium imaging, we find that spinal-cord-generated neural activity is transmitted to developing slow muscle cells, and synchronized activity spreads via gjd4/Cx46.8 GJ channels. Finally, we show that bioelectrical signal propagation within the developing neuromuscular system is required for appropriate myofiber organization and that disruption leads to defects in behavior. Our work reveals a molecular basis for GJ communication among developing muscle cells and reveals how perturbations to bioelectric signaling in the neuromuscular system may contribute to developmental myopathies. Moreover, this work underscores a critical motif of signal propagation between organ systems and highlights the pivotal role of GJ communication in coordinating bioelectric signaling during development.
Collapse
Affiliation(s)
| | - Judith S Eisen
- University of Oregon, Institute of Neuroscience, Eugene, OR 97405, USA
| | - Adam C Miller
- University of Oregon, Institute of Neuroscience, Eugene, OR 97405, USA.
| |
Collapse
|
10
|
Couturier N, Hörner SJ, Nürnberg E, Joazeiro C, Hafner M, Rudolf R. Aberrant evoked calcium signaling and nAChR cluster morphology in a SOD1 D90A hiPSC-derived neuromuscular model. Front Cell Dev Biol 2024; 12:1429759. [PMID: 38966427 PMCID: PMC11222430 DOI: 10.3389/fcell.2024.1429759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Familial amyotrophic lateral sclerosis (ALS) is a progressive neuromuscular disorder that is due to mutations in one of several target genes, including SOD1. So far, clinical records, rodent studies, and in vitro models have yielded arguments for either a primary motor neuron disease, or a pleiotropic pathogenesis of ALS. While mouse models lack the human origin, in vitro models using human induced pluripotent stem cells (hiPSC) have been recently developed for addressing ALS pathogenesis. In spite of improvements regarding the generation of muscle cells from hiPSC, the degree of maturation of muscle cells resulting from these protocols has remained limited. To fill these shortcomings, we here present a new protocol for an enhanced myotube differentiation from hiPSC with the option of further maturation upon coculture with hiPSC-derived motor neurons. The described model is the first to yield a combination of key myogenic maturation features that are consistent sarcomeric organization in association with complex nAChR clusters in myotubes derived from control hiPSC. In this model, myotubes derived from hiPSC carrying the SOD1 D90A mutation had reduced expression of myogenic markers, lack of sarcomeres, morphologically different nAChR clusters, and an altered nAChR-dependent Ca2+ response compared to control myotubes. Notably, trophic support provided by control hiPSC-derived motor neurons reduced nAChR cluster differences between control and SOD1 D90A myotubes. In summary, a novel hiPSC-derived neuromuscular model yields evidence for both muscle-intrinsic and nerve-dependent aspects of neuromuscular dysfunction in SOD1-based ALS.
Collapse
Affiliation(s)
- Nathalie Couturier
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Sarah Janice Hörner
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Elina Nürnberg
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Claudio Joazeiro
- Center for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, Mannheim, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, Mannheim, Germany
| |
Collapse
|
11
|
Xie W, Chen J, Cao X, Zhang J, Luo J, Wang Y. Roxithromycin exposure induces motoneuron malformation and behavioral deficits of zebrafish by interfering with the differentiation of motor neuron progenitor cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116327. [PMID: 38626605 DOI: 10.1016/j.ecoenv.2024.116327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
Roxithromycin (ROX), a commonly used macrolide antibiotic, is extensively employed in human medicine and livestock industries. Due to its structural stability and resistance to biological degradation, ROX persists as a resilient environmental contaminant, detectable in aquatic ecosystems and food products. However, our understanding of the potential health risks to humans from continuous ROX exposure remains limited. In this study, we used the zebrafish as a vertebrate model to explore the potential developmental toxicity of early ROX exposure, particularly focusing on its effects on locomotor functionality and CaP motoneuron development. Early exposure to ROX induces marked developmental toxicity in zebrafish embryos, significantly reducing hatching rates (n=100), body lengths (n=100), and increased malformation rates (n=100). The zebrafish embryos treated with a corresponding volume of DMSO (0.1%, v/v) served as vehicle controls (veh). Moreover, ROX exposure adversely affected the locomotive capacity of zebrafish embryos, and observations in transgenic zebrafish Tg(hb9:eGFP) revealed axonal loss in motor neurons, evident through reduced or irregular axonal lengths (n=80). Concurrently, abnormal apoptosis in ROX-exposed zebrafish embryos intensified alongside the upregulation of apoptosis-related genes (bax, bcl2, caspase-3a). Single-cell sequencing further disclosed substantial effects of ROX on genes involved in the differentiation of motor neuron progenitor cells (ngn1, olig2), axon development (cd82a, mbpa, plp1b, sema5a), and neuroimmunity (aplnrb, aplnra) in zebrafish larvae (n=30). Furthermore, the CaP motor neuron defects and behavioral deficits induced by ROX can be rescued by administering ngn1 agonist (n=80). In summary, ROX exposure leads to early-life abnormalities in zebrafish motor neurons and locomotor behavior by hindering the differentiation of motor neuron progenitor cells and inducing abnormal apoptosis.
Collapse
Affiliation(s)
- Wenjie Xie
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China; Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, China
| | - Juntao Chen
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China; Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, China
| | - Xiaoqian Cao
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Jiannan Zhang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China
| | - Juanjuan Luo
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, China.
| | - Yajun Wang
- Key Laboratory of Bioresources and Eco-environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Lee JJ, Wang T, Wiggins K, Lu PN, Underwood C, Ochenkowska K, Samarut E, Pollard LM, Flanagan-Steet H, Steet R. Dysregulated lysosomal exocytosis drives protease-mediated cartilage pathogenesis in multiple lysosomal disorders. iScience 2024; 27:109293. [PMID: 38495824 PMCID: PMC10940929 DOI: 10.1016/j.isci.2024.109293] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
The classic view of the lysosome as a static recycling center has been replaced with one of a dynamic and mobile hub of metabolic regulation. This revised view raises new questions about how dysfunction of this organelle causes pathology in inherited lysosomal disorders. Here we provide evidence for increased lysosomal exocytosis in the developing cartilage of three lysosomal disease zebrafish models with distinct etiologies. Dysregulated exocytosis was linked to altered cartilage development, increased activity of multiple cathepsin proteases, and cathepsin- and TGFβ-mediated pathogenesis in these models. Moreover, inhibition of cathepsin activity or direct blockade of exocytosis with small molecule modulators improved the cartilage phenotypes, reinforcing a connection between excessive extracellular protease activity and cartilage pathogenesis. This study highlights the pathogenic consequences in early cartilage development arising from uncontrolled release of lysosomal enzymes via exocytosis, and suggests that pharmacological enhancement of this process could be detrimental during tissue development.
Collapse
Affiliation(s)
- Jen-Jie Lee
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Tong Wang
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Kali Wiggins
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Po Nien Lu
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Christina Underwood
- Biochemical Genetics Laboratory, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | - Katarzyna Ochenkowska
- Research Center, Centre hospitalier de l’Université de Montréal (CHUM), Montreal, Canada
- Department of Neuroscience, Université de Montréal, Montréal, Canada
| | - Eric Samarut
- Research Center, Centre hospitalier de l’Université de Montréal (CHUM), Montreal, Canada
- Department of Neuroscience, Université de Montréal, Montréal, Canada
| | - Laura M. Pollard
- Biochemical Genetics Laboratory, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | - Richard Steet
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| |
Collapse
|
13
|
Wang X, Yue M, Cheung JPY, Cheung PWH, Fan Y, Wu M, Wang X, Zhao S, Khanshour AM, Rios JJ, Chen Z, Wang X, Tu W, Chan D, Yuan Q, Qin D, Qiu G, Wu Z, Zhang TJ, Ikegawa S, Wu N, Wise CA, Hu Y, Luk KDK, Song YQ, Gao B. Impaired glycine neurotransmission causes adolescent idiopathic scoliosis. J Clin Invest 2024; 134:e168783. [PMID: 37962965 PMCID: PMC10786698 DOI: 10.1172/jci168783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/08/2023] [Indexed: 11/16/2023] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is the most common form of spinal deformity, affecting millions of adolescents worldwide, but it lacks a defined theory of etiopathogenesis. Because of this, treatment of AIS is limited to bracing and/or invasive surgery after onset. Preonset diagnosis or preventive treatment remains unavailable. Here, we performed a genetic analysis of a large multicenter AIS cohort and identified disease-causing and predisposing variants of SLC6A9 in multigeneration families, trios, and sporadic patients. Variants of SLC6A9, which encodes glycine transporter 1 (GLYT1), reduced glycine-uptake activity in cells, leading to increased extracellular glycine levels and aberrant glycinergic neurotransmission. Slc6a9 mutant zebrafish exhibited discoordination of spinal neural activities and pronounced lateral spinal curvature, a phenotype resembling human patients. The penetrance and severity of curvature were sensitive to the dosage of functional glyt1. Administration of a glycine receptor antagonist or a clinically used glycine neutralizer (sodium benzoate) partially rescued the phenotype. Our results indicate a neuropathic origin for "idiopathic" scoliosis, involving the dysfunction of synaptic neurotransmission and central pattern generators (CPGs), potentially a common cause of AIS. Our work further suggests avenues for early diagnosis and intervention of AIS in preadolescents.
Collapse
Affiliation(s)
- Xiaolu Wang
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Ming Yue
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
| | - Prudence Wing Hang Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yanhui Fan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Meicheng Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiaojun Wang
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sen Zhao
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Anas M. Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
| | - Jonathan J. Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
- Eugene McDermott Center for Human Growth and Development, Departments of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zheyi Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Xiwei Wang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Tai Po, Hong Kong, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Tai Po, Hong Kong, China
| | - Guixing Qiu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Terry Jianguo Zhang
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Nan Wu
- Department of Orthopaedic Surgery, Department of Medical Research Center, Key Laboratory of Big Data for Spinal Deformities, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Peking Union Medical College Hospital (PUMCH) and Chinese Academy of Medical Sciences, Beijing, China
| | - Carol A. Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children (SRC), Dallas, Texas, USA
- Eugene McDermott Center for Human Growth and Development, Departments of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yong Hu
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
| | - Keith Dip Kei Luk
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
| | - You-Qiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Medicine, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Brain and Cognitive Sciences, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Bo Gao
- School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong, China
- Department of Orthopaedics and Traumatology, University of Hong Kong–Shenzhen Hospital, Shenzhen, China
- Centre for Translational Stem Cell Biology, Tai Po, Hong Kong, China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
14
|
Nemoz-Billet L, Brocard J, Ruggiero F, Bretaud S. Quantitative Image Analysis of Axonal Morphology in In Vivo Model. Methods Protoc 2023; 6:116. [PMID: 38133136 PMCID: PMC10745806 DOI: 10.3390/mps6060116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023] Open
Abstract
Quantifying axonal branching is crucial for understanding neural circuit function, developmental and regeneration processes and disease mechanisms. Factors that regulate patterns of axonal arborization and tune neuronal circuits are investigated for their implication in various disorders in brain connectivity. The lack of a reliable and user-friendly method makes the quantitative analysis of axon morphology difficult. Specifically, methods to visualize and quantify the complex axon arborization are challenging to implement and apply practically. Our study was aimed at developing a robust but simple method of quantification that used ImageJ 2D analysis and compared it with Imaris visualization and analysis of 3D images. We used zebrafish fluorescent transgenic lines to perform in vivo imaging of developing motor neuron axons that adequately reflected the complexity of axonal networks. Our new method, developed on ImageJ, is easy and fast, giving access to new information such as collateral distribution along the axonal shaft. This study describes step-by-step procedures that can be easily applied to a variety of organisms and in vitro systems. Our study provides a basis for further exploration of neural circuits to gain new insights into neuronal disorders and potential therapeutic interventions.
Collapse
Affiliation(s)
- Laurie Nemoz-Billet
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon-1, 69364 Lyon, France; (L.N.-B.); (F.R.)
| | - Jacques Brocard
- PLATIM, SFR Biosciences, ENS de Lyon, Inserm US8, CNRS UMS3444, Université Claude Bernard-Lyon-1, 69364 Lyon, France;
| | - Florence Ruggiero
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon-1, 69364 Lyon, France; (L.N.-B.); (F.R.)
| | - Sandrine Bretaud
- Institut de Génomique Fonctionnelle de Lyon, Ecole Normale Supérieure de Lyon, UMR5242 CNRS, Université Claude Bernard-Lyon-1, 69364 Lyon, France; (L.N.-B.); (F.R.)
| |
Collapse
|
15
|
Kelly JJ, Wen H, Brehm P. Single-cell RNAseq analysis of spinal locomotor circuitry in larval zebrafish. eLife 2023; 12:RP89338. [PMID: 37975797 PMCID: PMC10656102 DOI: 10.7554/elife.89338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Identification of the neuronal types that form the specialized circuits controlling distinct behaviors has benefited greatly from the simplicity offered by zebrafish. Electrophysiological studies have shown that in addition to connectivity, understanding of circuitry requires identification of functional specializations among individual circuit components, such as those that regulate levels of transmitter release and neuronal excitability. In this study, we use single-cell RNA sequencing (scRNAseq) to identify the molecular bases for functional distinctions between motoneuron types that are causal to their differential roles in swimming. The primary motoneuron, in particular, expresses high levels of a unique combination of voltage-dependent ion channel types and synaptic proteins termed functional 'cassettes.' The ion channel types are specialized for promoting high-frequency firing of action potentials and augmented transmitter release at the neuromuscular junction, both contributing to greater power generation. Our transcriptional profiling of spinal neurons further assigns expression of this cassette to specific interneuron types also involved in the central circuitry controlling high-speed swimming and escape behaviors. Our analysis highlights the utility of scRNAseq in functional characterization of neuronal circuitry, in addition to providing a gene expression resource for studying cell type diversity.
Collapse
Affiliation(s)
- Jimmy J Kelly
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Hua Wen
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Paul Brehm
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
16
|
Kelly JJ, Wen H, Brehm P. Single cell RNA-seq analysis of spinal locomotor circuitry in larval zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543939. [PMID: 37333232 PMCID: PMC10274715 DOI: 10.1101/2023.06.06.543939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Identification of the neuronal types that form the specialized circuits controlling distinct behaviors has benefited greatly from the simplicity offered by zebrafish. Electrophysiological studies have shown that additional to connectivity, understanding of circuitry requires identification of functional specializations among individual circuit components, such as those that regulate levels of transmitter release and neuronal excitability. In this study we use single cell RNA sequencing (scRNAseq) to identify the molecular bases for functional distinctions between motoneuron types that are causal to their differential roles in swimming. The primary motoneuron (PMn) in particular, expresses high levels of a unique combination of voltage-dependent ion channel types and synaptic proteins termed functional 'cassettes'. The ion channel types are specialized for promoting high frequency firing of action potentials and augmented transmitter release at the neuromuscular junction, both contributing to greater power generation. Our transcriptional profiling of spinal neurons further assigns expression of this cassette to specific interneuron types also involved in the central circuitry controlling high speed swimming and escape behaviors. Our analysis highlights the utility of scRNAseq in functional characterization of neuronal circuitry, in addition to providing a gene expression resource for studying cell type diversity.
Collapse
Affiliation(s)
- Jimmy J Kelly
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Hua Wen
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Paul Brehm
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
17
|
Fu CY, Chen HY, Lin CY, Chen SJ, Sheu JC, Tsai HJ. Extracellular Pgk1 interacts neural membrane protein enolase-2 to improve the neurite outgrowth of motor neurons. Commun Biol 2023; 6:849. [PMID: 37582937 PMCID: PMC10427645 DOI: 10.1038/s42003-023-05223-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 08/07/2023] [Indexed: 08/17/2023] Open
Abstract
Understanding the molecular interaction between ligand and receptor is important for providing the basis for the development of regenerative drugs. Although it has been reported that extracellular phosphoglycerate kinase 1 (Pgk1) can promote the neurite outgrowth of motoneurons, the Pgk1-interacting neural receptor remains unknown. Here we show that neural membranous Enolase-2 exhibits strong affinity with recombinant Pgk1-Flag, which is also evidently demonstrated by immunoelectron microscopy. The 325th-417th domain of Pgk1 interacts with the 405th-431st domain of Enolase-2, but neither Enolase-1 nor Enolase-3, promoting neurite outgrowth. Combining Pgk1 incubation and Enolase-2 overexpression, we demonstrate a highly significant enhancement of neurite outgrowth of motoneurons through a reduced p-P38-T180/p-Limk1-S323/p-Cofilin signaling. Collectively, extracellular Pgk1 interacts neural membrane receptor Enolase-2 to reduce the P38/Limk1/Cofilin signaling which results in promoting neurite outgrowth. The extracellular Pgk1-specific neural receptor found in this study should provide a material for screening potential small molecule drugs that promote motor nerve regeneration.
Collapse
Affiliation(s)
- Chuan-Yang Fu
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hong-Yu Chen
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | - Cheng-Yung Lin
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
| | - Shiang-Jiuun Chen
- Department of Life Science and Institute of Ecology and Evolutionary Biology, National Taiwan University, Taipei, Taiwan
- TechCommon-5, Bioimage Tool, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Jin-Chuan Sheu
- Liver Disease Prevention and Treatment Research Foundation, Taipei, Taiwan
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan.
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan.
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, City, Taiwan.
| |
Collapse
|
18
|
Walker LJ, Guevara C, Kawakami K, Granato M. Target-selective vertebrate motor axon regeneration depends on interaction with glial cells at a peripheral nerve plexus. PLoS Biol 2023; 21:e3002223. [PMID: 37590333 PMCID: PMC10464982 DOI: 10.1371/journal.pbio.3002223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/29/2023] [Accepted: 06/28/2023] [Indexed: 08/19/2023] Open
Abstract
A critical step for functional recovery from peripheral nerve injury is for regenerating axons to connect with their pre-injury targets. Reestablishing pre-injury target specificity is particularly challenging for limb-innervating axons as they encounter a plexus, a network where peripheral nerves converge, axons from different nerves intermingle, and then re-sort into target-specific bundles. Here, we examine this process at a plexus located at the base of the zebrafish pectoral fin, equivalent to tetrapod forelimbs. Using live cell imaging and sparse axon labeling, we find that regenerating motor axons from 3 nerves coalesce into the plexus. There, they intermingle and sort into distinct branches, and then navigate to their original muscle domains with high fidelity that restores functionality. We demonstrate that this regeneration process includes selective retraction of mistargeted axons, suggesting active correction mechanisms. Moreover, we find that Schwann cells are enriched and associate with axons at the plexus, and that Schwann cell ablation during regeneration causes profound axonal mistargeting. Our data provide the first real-time account of regenerating vertebrate motor axons navigating a nerve plexus and reveal a previously unappreciated role for Schwann cells to promote axon sorting at a plexus during regeneration.
Collapse
Affiliation(s)
- Lauren J. Walker
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Camilo Guevara
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka, Japan
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
19
|
Kim Y, Kim SS, Park BH, Hwang KS, Bae MA, Cho SH, Kim S, Park HC. Mechanism of Bisphenol F Affecting Motor System and Motor Activity in Zebrafish. TOXICS 2023; 11:477. [PMID: 37368577 DOI: 10.3390/toxics11060477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023]
Abstract
Bisphenol F (BPF; 4,4'-dihydroxydiphenylmethane) is one of the most frequently used compounds in the manufacture of plastics and epoxy resins. Previous studies have demonstrated that BPF affects locomotor behavior, oxidative stress, and neurodevelopment in zebrafish. However, its neurotoxic effects are controversial, and the underlying mechanisms are unclear. In order to determine whether BPF affects the motor system, we exposed zebrafish embryos to BPF and assessed behavioral, histological, and neurochemical changes. Spontaneous locomotor behavior and startle response were significantly decreased in BPF-treated zebrafish larvae compared with control larvae. BPF induced motor degeneration and myelination defects in zebrafish larvae. In addition, embryonic exposure to BPF resulted in altered metabolic profiles of neurochemicals, including neurotransmitters and neurosteroids, which may impact locomotion and motor function. In conclusion, exposure to BPF has the potential to affect survival, motor axon length, locomotor activity, myelination, and neurochemical levels of zebrafish larvae.
Collapse
Affiliation(s)
- Yeonhwa Kim
- Zebrafish Translational Medical Research Center, Korea University, Ansan 15588, Republic of Korea
| | - Seong Soon Kim
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34141, Republic of Korea
| | - Byeong Heon Park
- Medical Science Research Center, Ansan Hospital, Korea University, Ansan 15588, Republic of Korea
| | - Kyu-Seok Hwang
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34141, Republic of Korea
| | - Myung Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34141, Republic of Korea
| | - Sung-Hee Cho
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Suhyun Kim
- Zebrafish Translational Medical Research Center, Korea University, Ansan 15588, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 04763, Republic of Korea
| | - Hae-Chul Park
- Zebrafish Translational Medical Research Center, Korea University, Ansan 15588, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 04763, Republic of Korea
| |
Collapse
|
20
|
Inorganic arsenic alters the development of dopaminergic neurons but not serotonergic neurons and induces motor neuron development via Sonic hedgehog pathway in zebrafish. Neurosci Lett 2023; 795:137042. [PMID: 36587726 DOI: 10.1016/j.neulet.2022.137042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/15/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022]
Abstract
The mechanism of inorganic arsenic-induced neurotoxicity at the cellular level is not known. In zebrafish, teratological effects of inorganic arsenic have been shown at various concentrations. Here, we used similar concentrations of inorganic arsenic to evaluate the effects on specific neuron types. Exposure of zebrafish embryos at 5 h post fertilization (hpf) to sodium arsenite induced developmental toxicity (reduced body length) in 72 hpf larvae, beginning at a concentration of 300 mg/L concentration. Mortality or overt morphological deformity was detected at 500 mg/L sodium arsenite. While 200 mg/L sodium arsenite induced development of tyrosine hydroxylase-positive (dopaminergic) neurons, there was no significant effect on the development of 5-hydroxytryptamine (serotonergic) neurons. Sodium arsenite reduced acetylcholinesterase activity. In the hb9-GFP transgenic larvae, both 200 and 400 mg/L sodium arsenite produced supernumerary motor neurons in the spinal cord. Inhibition of the Sonic hedgehog (Shh) pathway that is essential for motor neuron development, by Gant61, prevented sodium arsenite-induced supernumerary motor neuron development. Inductively coupled plasma mass spectrometry (ICP-MS) revealed that with 200 mg/L and 400 mg/L sodium arsenite treatment, each larva had an average of 387.8 pg and 847.5 pg arsenic, respectively. The data show for the first time that inorganic arsenic alters the development of dopaminergic and motor neurons in the zebrafish larvae and the latter occurs through the Shh pathway. These results may help understand why arsenic-exposed populations suffer from psychiatric disorders and motor neuron disease and Shh may, potentially, serve as a plasma biomarker of arsenic toxicity.
Collapse
|
21
|
Walker LJ, Guevara C, Kawakami K, Granato M. A glia cell dependent mechanism at a peripheral nerve plexus critical for target-selective axon regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522786. [PMID: 36712008 PMCID: PMC9881934 DOI: 10.1101/2023.01.05.522786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A critical step for functional recovery from peripheral nerve injury is for regenerating axons to connect with their pre-injury targets. Reestablishing pre-injury target specificity is particularly challenging for limb-innervating axons as they encounter a plexus, a network where peripheral nerves converge, axons from different nerves intermingle, and then re-sort into target-specific bundles. Here, we examine this process at a plexus located at the base of the zebrafish pectoral fin, equivalent to tetrapod forelimbs. Using live cell imaging and sparse axon labeling, we find that regenerating motor axons from three nerves coalesce into the plexus. There, they intermingle and sort into distinct branches, and then navigate to their original muscle domains with high fidelity that restores functionality. We demonstrate that this regeneration process includes selective retraction of mistargeted axons, suggesting active correction mechanisms. Moreover, we find that Schwann cells are enriched and associate with axons at the plexus, and that Schwann cell ablation during regeneration causes profound axonal mistargeting. Our data provide the first real time account of regenerating vertebrate motor axons navigating a nerve plexus and reveal a previously unappreciated role for Schwann cells to promote axon sorting at a plexus during regeneration.
Collapse
Affiliation(s)
- Lauren J Walker
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Camilo Guevara
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, The Graduate University for Advanced Studies (SOKENDAI), Mishima, Shizuoka 411-8540, Japan
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
22
|
Luderman LN, Michaels MT, Levic DS, Knapik EW. Zebrafish Erc1b mediates motor innervation and organization of craniofacial muscles in control of jaw movement. Dev Dyn 2023; 252:104-123. [PMID: 35708710 DOI: 10.1002/dvdy.511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Movement of the lower jaw, a common behavior observed among vertebrates, is required for eating and processing food. This movement is controlled by signals sent from the trigeminal motor nerve through neuromuscular junctions (NMJs) to the masticatory muscles. Dysfunctional jaw movements contribute to craniomandibular disorders, yet the pathophysiology of these disorders is not well understood, as limited studies have been conducted on the molecular mechanisms of jaw movement. RESULTS Using erc1b/kimm533 genetic loss of function mutant, we evaluated lower jaw muscle organization and innervation by the cranial motor nerves in developing zebrafish. Using time-lapse confocal imaging of the erc1b mutant in a transgenic fluorescent reporter line, we found delayed trigeminal nerve growth and disrupted nerve branching architecture during muscle innervation. By automated 3D image analysis of NMJ distribution, we identified an increased number of small, disorganized NMJ clusters in erc1b mutant larvae compared to WT siblings. Using genetic replacement experiments, we determined the Rab GTPase binding domain of Erc1b is required for cranial motor nerve branching, but not NMJ organization or muscle attachment. CONCLUSIONS We identified Erc1b/ERC1 as a novel component of a genetic pathway contributing to muscle organization, trigeminal nerve outgrowth, and NMJ spatial distribution during development that is required for jaw movement.
Collapse
Affiliation(s)
- Lauryn N Luderman
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mackenzie T Michaels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Daniel S Levic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
- Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Ela W Knapik
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
23
|
Lee HC, Lai WL, Lin CY, Zeng CW, Sheu JC, Chou TB, Tsai HJ. Anp32a Promotes Neuronal Regeneration after Spinal Cord Injury of Zebrafish Embryos. Int J Mol Sci 2022; 23:ijms232415921. [PMID: 36555564 PMCID: PMC9786895 DOI: 10.3390/ijms232415921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
After spinal cord injury (SCI) in mammals, neuronal regeneration is limited; in contrast, such regeneration occurs quickly in zebrafish. Member A of the acidic nuclear phosphoprotein 32 (ANP32a) family is involved in neuronal development, but its function is controversial, and its involvement in zebrafish SCI remains unknown. To determine the role of zebrafish ANP32a in the neuronal regeneration of SCI embryos, we microinjected ANP32a mRNA into embryos from zebrafish transgenic line Tg(mnx1:GFP) prior to SCI. Compared to control SCI embryos, the results showed that the regeneration of spinal cord and resumption of swimming capability were promoted by the overexpression of ANP32a mRNA but reduced by its knockdown. We next combined fluorescence-activated cell sorting with immunochemical staining of anti-GFAP and immunofluorescence staining against anti-PH3 on Tg(gfap:GFP) SCI embryos. The results showed that ANP32a promoted the proliferation and cell number of radial glial cells at the injury epicenter at 24 h post-injury (hpi). Moreover, when we applied BrdU labeling to SCI embryos derived from crossing the Tg(gfap:GFP) and Tg(mnx1:TagRFP) lines, we found that both radial glial cells and motor neurons had proliferated, along with their increased cell numbers in Anp32a-overexpression SCI-embryos. On this basis, we conclude that ANP32a plays a positive role in the regeneration of zebrafish SCI embryos.
Collapse
Affiliation(s)
- Hung-Chieh Lee
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
| | - Wei-Lin Lai
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Cheng-Yung Lin
- Institute of Biomedical Science, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Chih-Wei Zeng
- Liver Disease Prevention and Treatment Research Foundation, Taipei 100008, Taiwan
| | - Jin-Chuan Sheu
- Liver Disease Prevention and Treatment Research Foundation, Taipei 100008, Taiwan
| | - Tze-Bin Chou
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Huai-Jen Tsai
- Department of Life Science, Fu Jen Catholic University, New Taipei City 242062, Taiwan
- Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City 242062, Taiwan
- Correspondence:
| |
Collapse
|
24
|
Nikolaou N, Gordon PM, Hamid F, Taylor R, Lloyd-Jones J, Makeyev EV, Houart C. Cytoplasmic pool of U1 spliceosome protein SNRNP70 shapes the axonal transcriptome and regulates motor connectivity. Curr Biol 2022; 32:5099-5115.e8. [PMID: 36384140 DOI: 10.1016/j.cub.2022.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 09/09/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022]
Abstract
Regulation of pre-mRNA splicing and polyadenylation plays a profound role in neurons by diversifying the proteome and modulating gene expression in response to physiological cues. Although most of the pre-mRNA processing is thought to occur in the nucleus, numerous splicing regulators are also found in neurites. Here, we show that U1-70K/SNRNP70, a component of the major spliceosome, localizes in RNA-associated granules in zebrafish axons. We identify the extra-nuclear SNRNP70 as an important regulator of motor axonal growth, nerve-dependent acetylcholine receptor (AChR) clustering, and neuromuscular synaptogenesis. This cytoplasmic pool has a protective role for a limited number of transcripts regulating their abundance and trafficking inside axons. Moreover, non-nuclear SNRNP70 regulates splice variants of transcripts such as agrin, thereby controlling synapse formation. Our results point to an unexpected, yet essential, function of non-nuclear SNRNP70 in axonal development, indicating a role of spliceosome proteins in cytoplasmic RNA metabolism during neuronal connectivity.
Collapse
Affiliation(s)
- Nikolas Nikolaou
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK; Department of Life Sciences, University of Bath, Bath BA2 7AY, UK.
| | - Patricia M Gordon
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Richard Taylor
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | | | - Eugene V Makeyev
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Corinne Houart
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
25
|
Barker CM, Miles KD, Doll CA. Fmrp regulates neuronal balance in embryonic motor circuit formation. Front Neurosci 2022; 16:962901. [PMID: 36408418 PMCID: PMC9669763 DOI: 10.3389/fnins.2022.962901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
Motor behavior requires the balanced production and integration of a variety of neural cell types. Motor neurons are positioned in discrete locations in the spinal cord, targeting specific muscles to drive locomotive contractions. Specialized spinal interneurons modulate and synchronize motor neuron activity to achieve coordinated motor output. Changes in the ratios and connectivity of spinal interneurons could drastically alter motor output by tipping the balance of inhibition and excitation onto target motor neurons. Importantly, individuals with Fragile X syndrome (FXS) and associated autism spectrum disorders often have significant motor challenges, including repetitive behaviors and epilepsy. FXS stems from the transcriptional silencing of the gene Fragile X Messenger Ribonucleoprotein 1 (FMR1), which encodes an RNA binding protein that is implicated in a multitude of crucial neurodevelopmental processes, including cell specification. Our work shows that Fmrp regulates the formation of specific interneurons and motor neurons that comprise early embryonic motor circuits. We find that zebrafish fmr1 mutants generate surplus ventral lateral descending (VeLD) interneurons, an early-born cell derived from the motor neuron progenitor domain (pMN). As VeLD interneurons are hypothesized to act as central pattern generators driving the earliest spontaneous movements, this imbalance could influence the formation and long-term function of motor circuits driving locomotion. fmr1 embryos also show reduced expression of proteins associated with inhibitory synapses, including the presynaptic transporter vGAT and the postsynaptic scaffold Gephyrin. Taken together, we show changes in embryonic motor circuit formation in fmr1 mutants that could underlie persistent hyperexcitability.
Collapse
Affiliation(s)
- Chase M. Barker
- Section of Developmental Biology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kaleb D. Miles
- Section of Developmental Biology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
- Biomedical Sciences and Biotechnology Program, Graduate School, University of Colorado, Aurora, CO, United States
| | - Caleb A. Doll
- Section of Developmental Biology, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
26
|
Wong HTC, Drerup CM. Using fluorescent indicators for in vivo quantification of spontaneous or evoked motor neuron presynaptic activity in transgenic zebrafish. STAR Protoc 2022; 3:101766. [PMID: 36240058 PMCID: PMC9568885 DOI: 10.1016/j.xpro.2022.101766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
In this protocol, we describe steps that utilize the optical clarity of the zebrafish larvae and the stereotyped motor neuron axon structure in the trunk to measure spontaneous or evoked motor neuron axon activity. This activity is detected with transgenic fluorescent indicators introduced into the larvae by zygotic injection. Fluorescent indicator intensity changes in the small neuromuscular junctions are quantified to measure the presynaptic calcium activity and consequent synaptic vesicle release. For complete details on the use and execution of this protocol, please refer to Mandal et al. (2020).
Collapse
Affiliation(s)
- Hiu-tung Candy Wong
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA,Corresponding author
| | - Catherine M. Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA,Corresponding author
| |
Collapse
|
27
|
Hua J, Wang X, Zhu J, Wang Q, Zhang W, Lei L, Zhu B, Han J, Yang L, Zhou B. Decabromodiphenyl ethane induced hyperactivity in developing zebrafish at environmentally relevant concentrations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114044. [PMID: 36055044 DOI: 10.1016/j.ecoenv.2022.114044] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/11/2022] [Accepted: 08/29/2022] [Indexed: 06/15/2023]
Abstract
Decabromodiphenyl ethane (DBDPE), a widely used novel brominated flame retardant, is gaining concerns due to rapidly increased contents in various environmental and biota samples. In the present study, zebrafish (Danio rerio) embryos were exposed to 2.91, 9.71, 29.14 and 97.12 μg/L of DBDPE until 120 h post-fertilization (hpf) to investigate the potential developmental neurotoxicity and underlying mechanisms. Chemical analysis revealed concentration-dependently increased body burdens of DBDPE in zebrafish larvae, with bioaccumulation factors (BCFs) ranging from 414 to 726. Embryonic exposure to DBDPE caused hyperactivity without affecting the development of secondary motoneuron axons and muscle fibers. However, further results implicated that DBDPE may affect the locomotor regulatory network via different mechanisms at lower and higher concentrations. On the one hand, embryonic exposure to 2.91 μg/L DBDPE transiently promoted spontaneous coiling contractions, but showed no effects on touch-response and swimming activity in zebrafish larvae. The whole-body contents of neurotransmitters were significantly decreased. Significant decreased protein abundances of α1-TUBULIN and SYN2a and molecular docking results pointed out possible interactions of DBDPE with these two proteins. However, these changes may be unconcerned with the transient hyperactivity, and the exact molecular mechanisms need further investigation. On the other hand, 29.14 and 97.12 μg/L DBDPE exposure caused longer-lasting effects in promoting spontaneous coiling contractions, and also touch-response and swimming activity. At the same time, increased ACh contents (without changes of other neurotransmitters) and ChAT activity and inhibited transcription of nAChRs were observed at higher concentrations. Molecular docking indicated direct interaction of DBDPE with ChAT. The results suggested that DBDPE induced hyperactivity at higher concentrations was probably involved with disrupted cholinergic system, with ChAT as a potential target. Given that the body burden of DBDPE in lower concentration group was comparable with those detected in wild fish, the current results may provide useful information for ecological risk assessment.
Collapse
Affiliation(s)
- Jianghuan Hua
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiulin Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaping Zhu
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Qiangwei Wang
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou 310058, China
| | - Wei Zhang
- State Environmental Protection Key Laboratory of Environmental Risk Assessment and Control on Chemical Process, School of Resource and Environmental Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lei Lei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Biran Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jian Han
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
28
|
Xing L, Chai R, Wang J, Lin J, Li H, Wang Y, Lai B, Sun J, Chen G. Expression of myelin transcription factor 1 and lamin B receptor mediate neural progenitor fate transition in the zebrafish spinal cord pMN domain. J Biol Chem 2022; 298:102452. [PMID: 36063998 PMCID: PMC9530849 DOI: 10.1016/j.jbc.2022.102452] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 02/05/2023] Open
Abstract
The pMN domain is a restricted domain in the ventral spinal cord, defined by the expression of the olig2 gene. Though it is known that the pMN progenitor cells can sequentially generate motor neurons and oligodendrocytes, the lineages of these progenitors are controversial and how their progeny are generated is not well understood. Using single-cell RNA sequencing, here, we identified a previously unknown heterogeneity among pMN progenitors with distinct fates and molecular signatures in zebrafish. Notably, we characterized two distinct motor neuron lineages using bioinformatic analysis. We then went on to investigate specific molecular programs that regulate neural progenitor fate transition. We validated experimentally that expression of the transcription factor myt1 (myelin transcription factor 1) and inner nuclear membrane integral proteins lbr (lamin B receptor) were critical for the development of motor neurons and neural progenitor maintenance, respectively. We anticipate that the transcriptome features and molecular programs identified in zebrafish pMN progenitors will not only provide an in-depth understanding of previous findings regarding the lineage analysis of oligodendrocyte progenitor cells and motor neurons but will also help in further understanding of the molecular programming involved in neural progenitor fate transition.
Collapse
Affiliation(s)
- Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China,For correspondence: Lingyan Xing; Gang Chen
| | - Rui Chai
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jiaqi Wang
- Department of Physiology, School of Medicine, Nantong University, Nantong, China
| | - Jiaqi Lin
- Department of Physiology, School of Medicine, Nantong University, Nantong, China
| | - Hanyang Li
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yueqi Wang
- School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Biqin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China,Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China,For correspondence: Lingyan Xing; Gang Chen
| |
Collapse
|
29
|
Chaytow H, Carroll E, Gordon D, Huang YT, van der Hoorn D, Smith HL, Becker T, Becker CG, Faller KME, Talbot K, Gillingwater TH. Targeting phosphoglycerate kinase 1 with terazosin improves motor neuron phenotypes in multiple models of amyotrophic lateral sclerosis. EBioMedicine 2022; 83:104202. [PMID: 35963713 PMCID: PMC9482929 DOI: 10.1016/j.ebiom.2022.104202] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with heterogeneous aetiology and a complex genetic background. Effective therapies are therefore likely to act on convergent pathways such as dysregulated energy metabolism, linked to multiple neurodegenerative diseases including ALS. METHODS Activity of the glycolysis enzyme phosphoglycerate kinase 1 (PGK1) was increased genetically or pharmacologically using terazosin in zebrafish, mouse and ESC-derived motor neuron models of ALS. Multiple disease phenotypes were assessed to determine the therapeutic potential of this approach, including axon growth and motor behaviour, survival and cell death following oxidative stress. FINDINGS We have found that targeting a single bioenergetic protein, PGK1, modulates motor neuron vulnerability in vivo. In zebrafish models of ALS, overexpression of PGK1 rescued motor axon phenotypes and improved motor behaviour. Treatment with terazosin, an FDA-approved compound with a known non-canonical action of increasing PGK1 activity, also improved these phenotypes. Terazosin treatment extended survival, improved motor phenotypes and increased motor neuron number in Thy1-hTDP-43 mice. In ESC-derived motor neurons expressing TDP-43M337V, terazosin protected against oxidative stress-induced cell death and increased basal glycolysis rates, while rescuing stress granule assembly. INTERPRETATION Our data demonstrate that terazosin protects motor neurons via multiple pathways, including upregulating glycolysis and rescuing stress granule formation. Repurposing terazosin therefore has the potential to increase the limited therapeutic options across all forms of ALS, irrespective of disease cause. FUNDING This work was supported by project grant funding from MND Scotland, the My Name'5 Doddie Foundation, Medical Research Council Doctoral Student Training Fellowship [Ref: BST0010Z] and Academy of Medical Sciences grant [SGL023\1100].
Collapse
Affiliation(s)
- Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK
| | - Emily Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford, UK
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford, UK
| | - Yu-Ting Huang
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK
| | - Dinja van der Hoorn
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK
| | - Hannah Louise Smith
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK
| | - Thomas Becker
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK; Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, Dresden, Germany
| | - Catherina Gwynne Becker
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK; Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, Dresden, Germany
| | | | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford; Oxford, UK
| | - Thomas Henry Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh; Edinburgh, UK; Euan MacDonald Centre for Motor Neuron Disease Research; Edinburgh, UK.
| |
Collapse
|
30
|
Petel Légaré V, Rampal CJ, Gurberg TJN, Harji ZA, Allard-Chamard X, Rodríguez EC, Armstrong GAB. Development of an endogenously myc-tagged TARDBP (TDP-43) zebrafish model using the CRISPR/Cas9 system and homology directed repair. Comp Biochem Physiol B Biochem Mol Biol 2022; 261:110756. [PMID: 35580804 DOI: 10.1016/j.cbpb.2022.110756] [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: 03/12/2022] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 11/25/2022]
Abstract
Many of the modern advances in cellular biology have been made by the expression of engineered constructs with epitope tags for subsequent biochemical investigations. While the utility of epitope tags has permitted insights in cellular and animal models, these are often expressed using traditional transgenic approaches. Using the CRISPR/Cas9 system and homology directed repair we recombine a single myc epitope sequence following the start codon of the zebrafish ortholog of TARDBP (TDP-43). TDP-43 is an RNA binding protein that is involved in the neurodegenerative disease amyotrophic lateral sclerosis and frontotemporal dementia. We report that zebrafish expressing the myc-tardbp engendered allele produced a stable protein that was detected by both western blot and immunofluorescence. Furthermore, both heterozygous and homozygous carriers of the myc-tardbp allele developed to sexual maturity. We propose that the methodology used here will be useful for zebrafish researchers and other comparative animal biologists interested in developing animal models expressing endogenously tagged proteins.
Collapse
Affiliation(s)
- Virginie Petel Légaré
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University. https://twitter.com/virginiepet
| | - Christian J Rampal
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University. https://twitter.com/ChristianRampal
| | - Tyler J N Gurberg
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University
| | - Ziyaan A Harji
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University. https://twitter.com/ziyaanharji
| | - Xavier Allard-Chamard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University
| | - Esteban C Rodríguez
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University
| | - Gary A B Armstrong
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University.
| |
Collapse
|
31
|
Bermedo-García F, Zelada D, Martínez E, Tabares L, Henríquez JP. Functional regeneration of the murine neuromuscular synapse relies on long-lasting morphological adaptations. BMC Biol 2022; 20:158. [PMID: 35804361 PMCID: PMC9270767 DOI: 10.1186/s12915-022-01358-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 05/05/2022] [Indexed: 12/04/2022] Open
Abstract
Background In a broad variety of species, muscle contraction is controlled at the neuromuscular junction (NMJ), the peripheral synapse composed of a motor nerve terminal, a muscle specialization, and non-myelinating terminal Schwann cells. While peripheral nerve damage leads to successful NMJ reinnervation in animal models, muscle fiber reinnervation in human patients is largely inefficient. Interestingly, some hallmarks of NMJ denervation and early reinnervation in murine species, such as fragmentation and poly-innervation, are also phenotypes of aged NMJs or even of unaltered conditions in other species, including humans. We have reasoned that rather than features of NMJ decline, such cellular responses could represent synaptic adaptations to accomplish proper functional recovery. Here, we have experimentally tackled this idea through a detailed comparative study of the short- and long-term consequences of irreversible (chronic) and reversible (partial) NMJ denervation in the convenient cranial levator auris longus muscle. Results Our findings reveal that irreversible muscle denervation results in highly fragmented postsynaptic domains and marked ectopic acetylcholine receptor clustering along with significant terminal Schwann cells sprouting and progressive detachment from the NMJ. Remarkably, even though reversible nerve damage led to complete reinnervation after 11 days, we found that more than 30% of NMJs are poly-innervated and around 65% of postsynaptic domains are fragmented even 3 months after injury, whereas synaptic transmission is fully recovered two months after nerve injury. While postsynaptic stability was irreversibly decreased after chronic denervation, this parameter was only transiently affected by partial NMJ denervation. In addition, we found that a combination of morphometric analyses and postsynaptic stability determinations allows discriminating two distinct forms of NMJ fragmentation, stable-smooth and unstable-blurred, which correlate with their regeneration potential. Conclusions Together, our data unveil that reversible nerve damage imprints a long-lasting reminiscence in the NMJ that results in the rearrangement of its cellular components. Instead of being predictive of NMJ decline, these traits may represent an efficient adaptive response for proper functional recovery. As such, these features are relevant targets to be considered in strategies aimed to restore motor function in detrimental conditions for peripheral innervation. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01358-4.
Collapse
Affiliation(s)
- Francisca Bermedo-García
- Laboratory of Neuromuscular Studies (NeSt Lab), Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Diego Zelada
- Laboratory of Neuromuscular Studies (NeSt Lab), Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Esperanza Martínez
- Laboratory of Neuromuscular Studies (NeSt Lab), Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Lucía Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, Universidad de Sevilla, Sevilla, Spain
| | - Juan Pablo Henríquez
- Laboratory of Neuromuscular Studies (NeSt Lab), Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
32
|
Zheng YQ, Suo GH, Liu D, Li HY, Wu YJ, Ni H. Nexmifa Regulates Axon Morphogenesis in Motor Neurons in Zebrafish. Front Mol Neurosci 2022; 15:848257. [PMID: 35431796 PMCID: PMC9009263 DOI: 10.3389/fnmol.2022.848257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Nexmif is mainly expressed in the central nervous system (CNS) and plays important roles in cell migration, cell to cell and cell-matrix adhesion, and maintains normal synaptic formation and function. Nevertheless, it is unclear how nexmif is linked to motor neuron morphogenesis. Here, we provided in situ hybridization evidence that nexmifa (zebrafish paralog) was localized to the brain and spinal cord and acted as a vital regulator of motor neuron morphogenesis. Nexmifa deficiency in zebrafish larvae generated abnormal primary motor neuron (PMN) development, including truncated Cap axons and decreased branches in Cap axons. Importantly, RNA-sequencing showed that nexmifa-depleted zebrafish embryos caused considerable CNS related gene expression alterations. Differentially expressed genes (DEGs) were mainly involved in axon guidance and several synaptic pathways, including glutamatergic, GABAergic, dopaminergic, cholinergic, and serotonergic synapse pathways, according to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation. In particular, when compared with other pathways, DEGs were highest (84) in the axon guidance pathway, according to Organismal Systems. Efna5b, bmpr2b, and sema6ba were decreased markedly in nexmifa-depleted zebrafish embryos. Moreover, both overexpression of efna5b mRNA and sema6ba mRNA could partially rescued motor neurons morphogenesis. These observations supported nexmifa as regulating axon morphogenesis of motor neurons in zebrafish. Taken together, nexmifa elicited crucial roles during motor neuron development by regulating the morphology of neuronal axons.
Collapse
Affiliation(s)
- Yu-qin Zheng
- Division of Brain Science, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Gui-hai Suo
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Dong Liu
- School of Life Sciences, Nantong University, Nantong, China
| | - Hai-ying Li
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - You-jia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
- You-jia Wu,
| | - Hong Ni
- Division of Brain Science, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
- *Correspondence: Hong Ni,
| |
Collapse
|
33
|
Li DD, Deng J, Jin B, Han S, Gu XY, Zhou XF, Yin XF. Effects of delayed repair of peripheral nerve injury on the spatial distribution of motor endplates in target muscle. Neural Regen Res 2022; 17:459-464. [PMID: 34269223 PMCID: PMC8464005 DOI: 10.4103/1673-5374.317990] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Motor endplates (MEPs) are important sites of information exchange between motor neurons and skeletal muscle, and are distributed in an organized pattern of lamellae in the muscle. Delayed repair of peripheral nerve injury typically results in unsatisfactory functional recovery because of MEP degeneration. In this study, the mouse tibial nerve was transected and repaired with a biodegradable chitin conduit, immediately following or 1 or 3 months after the injury. Fluorescent α-bungarotoxin was injected to label MEPs. Tissue optical clearing combined with light-sheet microscopy revealed that MEPs were distributed in an organized pattern of lamellae in skeletal muscle after delayed repair for 1 and 3 months. However, the total number of MEPs, the number of MEPs per lamellar cluster, and the maturation of single MEPs in gastrocnemius muscle gradually decreased with increasing denervation time. These findings suggest that delayed repair can restore the spatial distribution of MEPs, but it has an adverse effect on the homogeneity of MEPs in the lamellar clusters and the total number of MEPs in the target muscle. The study procedures were approved by the Animal Ethics Committee of the Peking University People's Hospital (approval No. 2019PHC015) on April 8, 2019.
Collapse
Affiliation(s)
- Dong-Dong Li
- Department of Trauma and Orthopedics, Peking University People's Hospital; Department of Orthopedics, PLA Strategic Support Force Medical Center, Beijing, China
| | - Jin Deng
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| | - Bo Jin
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| | - Shuai Han
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| | - Xin-Yi Gu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| | - Xue-Feng Zhou
- Department of Orthopedics, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiao-Feng Yin
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| |
Collapse
|
34
|
Quan FB, Gaillard AL, Alejevski F, Pézeron G, Tostivint H. Urotensin II-related peptide (Urp) is expressed in motoneurons in zebrafish, but is dispensable for locomotion in larva. Peptides 2021; 146:170675. [PMID: 34655691 DOI: 10.1016/j.peptides.2021.170675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
The urotensin 2 (uts2) gene family consists of four paralogs called uts2, uts2-related peptide (urp), urp1 and urp2. uts2 is known to exert a large array of biological effects, including osmoregulation, control of cardiovascular functions and regulation of endocrine activities. Lately, urp1 and urp2 have been shown to regulate axial straightening during embryogenesis. In contrast, much less is known about the roles of urp. The aim of the present study was to investigate the expression and the functions of urp by using the zebrafish as a model. For this purpose, we determined the expression pattern of the urp gene. We found that urp is expressed in motoneurons of the brainstem and the spinal cord, as in tetrapods. This was confirmed with a new Tg(urp:gfp) fluorescent reporter line. We also generated a urp knockout mutant by using CRISPR/Cas9-mediated genome editing and analysed its locomotor activity in larvae. urp mutant did not exhibit any apparent defect of spontaneous swimming when compared to wild-type. We also tested the idea that urp may represent an intermediary of urp1 and urp2 in their role on axial straightening. We found that the upward bending of the tail induced by the overexpression of urp2 in 24-hpf embryos was not altered in urp mutants. Our results indicate that urp does probably not act as a relay downstream of urp2. In conclusion, the present study showed that zebrafish urp gene is primarily expressed in motoneurons but is apparently dispensable for locomotor activity in the early larval stages.
Collapse
Affiliation(s)
- Feng B Quan
- Molecular Physiology and Adaptation (PhyMA - UMR7221), Muséum National d'Histoire naturelle, CNRS, Paris, France
| | - Anne-Laure Gaillard
- Molecular Physiology and Adaptation (PhyMA - UMR7221), Muséum National d'Histoire naturelle, CNRS, Paris, France
| | - Faredin Alejevski
- Molecular Physiology and Adaptation (PhyMA - UMR7221), Muséum National d'Histoire naturelle, CNRS, Paris, France
| | - Guillaume Pézeron
- Molecular Physiology and Adaptation (PhyMA - UMR7221), Muséum National d'Histoire naturelle, CNRS, Paris, France.
| | - Hervé Tostivint
- Molecular Physiology and Adaptation (PhyMA - UMR7221), Muséum National d'Histoire naturelle, CNRS, Paris, France.
| |
Collapse
|
35
|
Walker LJ, Roque RA, Navarro MF, Granato M. Agrin/Lrp4 signal constrains MuSK-dependent neuromuscular synapse development in appendicular muscle. Development 2021; 148:272655. [PMID: 34714331 PMCID: PMC8602948 DOI: 10.1242/dev.199790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/24/2021] [Indexed: 12/17/2022]
Abstract
The receptor tyrosine kinase MuSK, its co-receptor Lrp4 and the Agrin ligand constitute a signaling pathway that is crucial in axial muscle for neuromuscular synapse development, yet whether this pathway functions similarly in appendicular muscle is unclear. Here, using the larval zebrafish pectoral fin, equivalent to tetrapod forelimbs, we show that, similar to axial muscle, developing appendicular muscles form aneural acetylcholine receptor (AChR) clusters prior to innervation. As motor axons arrive, neural AChR clusters form, eventually leading to functional synapses in a MuSK-dependent manner. We find that loss of Agrin or Lrp4 function, which abolishes synaptic AChR clusters in axial muscle, results in enlarged presynaptic nerve regions and progressively expanding appendicular AChR clusters, mimicking the consequences of motoneuron ablation. Moreover, musk depletion in lrp4 mutants partially restores synaptic AChR patterning. Combined, our results provide compelling evidence that, in addition to the canonical pathway in which Agrin/Lrp4 stimulates MuSK activity, Agrin/Lrp4 signaling in appendicular muscle constrains MuSK-dependent neuromuscular synapse organization. Thus, we reveal a previously unappreciated role for Agrin/Lrp4 signaling, thereby highlighting distinct differences between axial and appendicular synapse development.
Collapse
|
36
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
37
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
38
|
Liu KC, Villasenor A, Bertuzzi M, Schmitner N, Radros N, Rautio L, Mattonet K, Matsuoka RL, Reischauer S, Stainier DY, Andersson O. Insulin-producing β-cells regenerate ectopically from a mesodermal origin under the perturbation of hemato-endothelial specification. eLife 2021; 10:65758. [PMID: 34403334 PMCID: PMC8370765 DOI: 10.7554/elife.65758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/06/2021] [Indexed: 12/19/2022] Open
Abstract
To investigate the role of the vasculature in pancreatic β-cell regeneration, we crossed a zebrafish β-cell ablation model into the avascular npas4l mutant (i.e. cloche). Surprisingly, β-cell regeneration increased markedly in npas4l mutants owing to the ectopic differentiation of β-cells in the mesenchyme, a phenotype not previously reported in any models. The ectopic β-cells expressed endocrine markers of pancreatic β-cells, and also responded to glucose with increased calcium influx. Through lineage tracing, we determined that the vast majority of these ectopic β-cells has a mesodermal origin. Notably, ectopic β-cells were found in npas4l mutants as well as following knockdown of the endothelial/myeloid determinant Etsrp. Together, these data indicate that under the perturbation of endothelial/myeloid specification, mesodermal cells possess a remarkable plasticity enabling them to form β-cells, which are normally endodermal in origin. Understanding the restriction of this differentiation plasticity will help exploit an alternative source for β-cell regeneration.
Collapse
Affiliation(s)
- Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Alethia Villasenor
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Maria Bertuzzi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Nicole Schmitner
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Niki Radros
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Linn Rautio
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ryota L Matsuoka
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Cardio-Pulmonary Institute, Frankfurt, Germany; Medical Clinic I, (Cardiology/Angiology) and Campus Kerckhoff, Justus-Liebig-University Giessen, Giessen, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Terni B, Llobet A. Axon terminals control endolysosome diffusion to support synaptic remodelling. Life Sci Alliance 2021; 4:4/8/e202101105. [PMID: 34226200 PMCID: PMC8321675 DOI: 10.26508/lsa.202101105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/27/2022] Open
Abstract
Endolysosomes present in the presynaptic terminal move by diffusion constrained by F-actin and increase their mobility during the remodelling of synaptic connectivity to support a local degradative activity. Endolysosomes are acidic organelles formed by the fusion of endosomes with lysosomes. In the presynaptic compartment they contribute to protein homeostasis, the maintenance of vesicle pools and synaptic stability. Here, we evaluated the mobility of endolysosomes found in axon terminals of olfactory sensory neurons of Xenopus tropicalis tadpoles. F-actin restricts the motion of these presynaptic acidic organelles which is characterized by a diffusion coefficient of 6.7 × 10−3 μm2·s−1. Local injection of secreted protein acidic and rich in cysteine (SPARC) in the glomerular layer of the olfactory bulb disrupts the structure of synaptic F-actin patches and increases the presence and mobility of endolysosomal organelles found in axon terminals. The increased motion of endolysosomes is localized to the presynaptic compartment and does not promote their access to axonal regions for retrograde transportation to the cell body. Local activation of synaptic degradation mechanisms mediated by SPARC coincides with a loss of the ability of tadpoles to detect waterborne odorants. Together, these observations show that the diffusion of presynaptic endolysosomes increases during conditions of synaptic remodelling to support their local degradative activity.
Collapse
Affiliation(s)
- Beatrice Terni
- Department of Pathology and Experimental Therapy, School of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain .,Laboratory of Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Artur Llobet
- Department of Pathology and Experimental Therapy, School of Medicine, Institute of Neurosciences, University of Barcelona, Barcelona, Spain .,Laboratory of Neurobiology, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
40
|
Meserve JH, Nelson JC, Marsden KC, Hsu J, Echeverry FA, Jain RA, Wolman MA, Pereda AE, Granato M. A forward genetic screen identifies Dolk as a regulator of startle magnitude through the potassium channel subunit Kv1.1. PLoS Genet 2021; 17:e1008943. [PMID: 34061829 PMCID: PMC8195410 DOI: 10.1371/journal.pgen.1008943] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 06/11/2021] [Accepted: 05/04/2021] [Indexed: 11/19/2022] Open
Abstract
The acoustic startle response is an evolutionarily conserved avoidance behavior. Disruptions in startle behavior, particularly startle magnitude, are a hallmark of several human neurological disorders. While the neural circuitry underlying startle behavior has been studied extensively, the repertoire of genes and genetic pathways that regulate this locomotor behavior has not been explored using an unbiased genetic approach. To identify such genes, we took advantage of the stereotypic startle behavior in zebrafish larvae and performed a forward genetic screen coupled with whole genome analysis. We uncovered mutations in eight genes critical for startle behavior, including two genes encoding proteins associated with human neurological disorders, Dolichol kinase (Dolk), a broadly expressed regulator of the glycoprotein biosynthesis pathway, and the potassium Shaker-like channel subunit Kv1.1. We demonstrate that Kv1.1 and Dolk play critical roles in the spinal cord to regulate movement magnitude during the startle response and spontaneous swim movements. Moreover, we show that Kv1.1 protein is mislocalized in dolk mutants, suggesting they act in a common genetic pathway. Combined, our results identify a diverse set of eight genes, all associated with human disorders, that regulate zebrafish startle behavior and reveal a previously unappreciated role for Dolk and Kv1.1 in regulating movement magnitude via a common genetic pathway.
Collapse
Affiliation(s)
- Joy H. Meserve
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jessica C. Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kurt C. Marsden
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jerry Hsu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fabio A. Echeverry
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Roshan A. Jain
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Marc A. Wolman
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alberto E. Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
41
|
Issa FA, Hall MK, Hatchett CJ, Weidner DA, Fiorenza AC, Schwalbe RA. Compromised N-Glycosylation Processing of Kv3.1b Correlates with Perturbed Motor Neuron Structure and Locomotor Activity. BIOLOGY 2021; 10:486. [PMID: 34070741 PMCID: PMC8229559 DOI: 10.3390/biology10060486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022]
Abstract
Neurological difficulties commonly accompany individuals suffering from congenital disorders of glycosylation, resulting from defects in the N-glycosylation pathway. Vacant N-glycosylation sites (N220 and N229) of Kv3, voltage-gated K+ channels of high-firing neurons, deeply perturb channel activity in neuroblastoma (NB) cells. Here we examined neuron development, localization, and activity of Kv3 channels in wildtype AB zebrafish and CRISPR/Cas9 engineered NB cells, due to perturbations in N-glycosylation processing of Kv3.1b. We showed that caudal primary (CaP) motor neurons of zebrafish spinal cord transiently expressing fully glycosylated (WT) Kv3.1b have stereotypical morphology, while CaP neurons expressing partially glycosylated (N220Q) Kv3.1b showed severe maldevelopment with incomplete axonal branching and extension around the ventral musculature. Consequently, larvae expressing N220Q in CaP neurons had impaired swimming locomotor activity. We showed that replacement of complex N-glycans with oligomannose attached to Kv3.1b and at cell surface lessened Kv3.1b dispersal to outgrowths by altering the number, size, and density of Kv3.1b-containing particles in membranes of rat neuroblastoma cells. Opening and closing rates were slowed in Kv3 channels containing Kv3.1b with oligomannose, instead of complex N-glycans, which suggested a reduction in the intrinsic dynamics of the Kv3.1b α-subunit. Thus, N-glycosylation processing of Kv3.1b regulates neuronal development and excitability, thereby controlling motor activity.
Collapse
Affiliation(s)
- Fadi A. Issa
- Department of Biology, East Carolina University, Greenville, NC 27858, USA;
| | - M. Kristen Hall
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (M.K.H.); (C.J.H.); (A.C.F.)
| | - Cody J. Hatchett
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (M.K.H.); (C.J.H.); (A.C.F.)
| | - Douglas A. Weidner
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| | - Alexandria C. Fiorenza
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (M.K.H.); (C.J.H.); (A.C.F.)
| | - Ruth A. Schwalbe
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; (M.K.H.); (C.J.H.); (A.C.F.)
| |
Collapse
|
42
|
Xing G, Jing H, Yu Z, Chen P, Wang H, Xiong WC, Mei L. Membraneless condensates by Rapsn phase separation as a platform for neuromuscular junction formation. Neuron 2021; 109:1963-1978.e5. [PMID: 34033754 DOI: 10.1016/j.neuron.2021.04.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/27/2021] [Accepted: 04/22/2021] [Indexed: 12/29/2022]
Abstract
Our daily life depends on muscle contraction, a process that is controlled by the neuromuscular junction (NMJ). However, the mechanisms of NMJ assembly remain unclear. Here we show that Rapsn, a protein critical for NMJ formation, undergoes liquid-liquid phase separation (LLPS) and condensates into liquid-like assemblies. Such assemblies can recruit acetylcholine receptors (AChRs), cytoskeletal proteins, and signaling proteins for postsynaptic differentiation. Rapsn LLPS requires multivalent binding of tetratricopeptide repeats (TPRs) and is increased by Musk signaling. The capacity of Rapsn to condensate and co-condensate with interaction proteins is compromised by mutations of congenital myasthenic syndromes (CMSs). NMJ formation is impaired in mutant mice carrying a CMS-associated, LLPS-deficient mutation. These results reveal a critical role of Rapsn LLPS in forming a synaptic semi-membraneless compartment for NMJ formation.
Collapse
Affiliation(s)
- Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Peng Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
43
|
Cavone L, McCann T, Drake LK, Aguzzi EA, Oprişoreanu AM, Pedersen E, Sandi S, Selvarajah J, Tsarouchas TM, Wehner D, Keatinge M, Mysiak KS, Henderson BEP, Dobie R, Henderson NC, Becker T, Becker CG. A unique macrophage subpopulation signals directly to progenitor cells to promote regenerative neurogenesis in the zebrafish spinal cord. Dev Cell 2021; 56:1617-1630.e6. [PMID: 34033756 DOI: 10.1016/j.devcel.2021.04.031] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022]
Abstract
Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.
Collapse
Affiliation(s)
- Leonardo Cavone
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Tess McCann
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Louisa K Drake
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Erika A Aguzzi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Elisa Pedersen
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Soe Sandi
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jathurshan Selvarajah
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Themistoklis M Tsarouchas
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Daniel Wehner
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Max Planck Institute for the Science of Light, Staudtstraße 2, Erlangen 91058, Germany; Max-Planck-Zentrum für Physik und Medizin, Staudtstraße 2, Erlangen 91058, Germany
| | - Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Karolina S Mysiak
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Beth E P Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, 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
|
44
|
Oprişoreanu AM, Smith HL, Krix S, Chaytow H, Carragher NO, Gillingwater TH, Becker CG, Becker T. Automated in vivo drug screen in zebrafish identifies synapse-stabilising drugs with relevance to spinal muscular atrophy. Dis Model Mech 2021; 14:259422. [PMID: 33973627 PMCID: PMC8106959 DOI: 10.1242/dmm.047761] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Synapses are particularly vulnerable in many neurodegenerative diseases and often the first to degenerate, for example in the motor neuron disease spinal muscular atrophy (SMA). Compounds that can counteract synaptic destabilisation are rare. Here, we describe an automated screening paradigm in zebrafish for small-molecule compounds that stabilize the neuromuscular synapse in vivo. We make use of a mutant for the axonal C-type lectin chondrolectin (chodl), one of the main genes dysregulated in SMA. In chodl-/- mutants, neuromuscular synapses that are formed at the first synaptic site by growing axons are not fully mature, causing axons to stall, thereby impeding further axon growth beyond that synaptic site. This makes axon length a convenient read-out for synapse stability. We screened 982 small-molecule compounds in chodl chodl-/- mutants and found four that strongly rescued motor axon length. Aberrant presynaptic neuromuscular synapse morphology was also corrected. The most-effective compound, the adenosine uptake inhibitor drug dipyridamole, also rescued axon growth defects in the UBA1-dependent zebrafish model of SMA. Hence, we describe an automated screening pipeline that can detect compounds with relevance to SMA. This versatile platform can be used for drug and genetic screens, with wider relevance to synapse formation and stabilisation.
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
| | - Hannah L Smith
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB
| | - Sophia Krix
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB
| | - Helena Chaytow
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, EH16 4SB Edinburgh, UK
| | - Neil O Carragher
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR Edinburgh, UK
| | - Thomas H Gillingwater
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, EH16 4SB Edinburgh, UK
| | - Catherina G Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, EH16 4SB Edinburgh, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB
| |
Collapse
|
45
|
Exploiting pyocyanin to treat mitochondrial disease due to respiratory complex III dysfunction. Nat Commun 2021; 12:2103. [PMID: 33833234 PMCID: PMC8032734 DOI: 10.1038/s41467-021-22062-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/25/2021] [Indexed: 01/13/2023] Open
Abstract
Mitochondrial diseases impair oxidative phosphorylation and ATP production, while effective treatment is still lacking. Defective complex III is associated with a highly variable clinical spectrum. We show that pyocyanin, a bacterial redox cycler, can replace the redox functions of complex III, acting as an electron shunt. Sub-μM pyocyanin was harmless, restored respiration and increased ATP production in fibroblasts from five patients harboring pathogenic mutations in TTC19, BCS1L or LYRM7, involved in assembly/stabilization of complex III. Pyocyanin normalized the mitochondrial membrane potential, and mildly increased ROS production and biogenesis. These in vitro effects were confirmed in both DrosophilaTTC19KO and in Danio rerioTTC19KD, as administration of low concentrations of pyocyanin significantly ameliorated movement proficiency. Importantly, daily administration of pyocyanin for two months was not toxic in control mice. Our results point to utilization of redox cyclers for therapy of complex III disorders.
Collapse
|
46
|
Asakawa K, Handa H, Kawakami K. Illuminating ALS Motor Neurons With Optogenetics in Zebrafish. Front Cell Dev Biol 2021; 9:640414. [PMID: 33816488 PMCID: PMC8012537 DOI: 10.3389/fcell.2021.640414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by progressive degeneration of motor neurons in the brain and spinal cord. Spinal motor neurons align along the spinal cord length within the vertebral column, and extend long axons to connect with skeletal muscles covering the body surface. Due to this anatomy, spinal motor neurons are among the most difficult cells to observe in vivo. Larval zebrafish have transparent bodies that allow non-invasive visualization of whole cells of single spinal motor neurons, from somas to the neuromuscular synapses. This unique feature, combined with its amenability to genome editing, pharmacology, and optogenetics, enables functional analyses of ALS-associated proteins in the spinal motor neurons in vivo with subcellular resolution. Here, we review the zebrafish skeletal neuromuscular system and the optical methods used to study it. We then introduce a recently developed optogenetic zebrafish ALS model that uses light illumination to control oligomerization, phase transition and aggregation of the ALS-associated DNA/RNA-binding protein called TDP-43. Finally, we will discuss how this disease-in-a-fish ALS model can help solve key questions about ALS pathogenesis and lead to new ALS therapeutics.
Collapse
Affiliation(s)
- Kazuhide Asakawa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan.,Department of Genetics, Graduate University for Advanced Studies (SOKENDAI), Mishima, Japan
| |
Collapse
|
47
|
Dong Z, Chen X, Li Y, Zhuo R, Lai X, Liu M. Microtubule Severing Protein Fignl2 Contributes to Endothelial and Neuronal Branching in Zebrafish Development. Front Cell Dev Biol 2021; 8:593234. [PMID: 33585441 PMCID: PMC7873885 DOI: 10.3389/fcell.2020.593234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
Previously, fidgetin (fign) and its family members fidgetin-like 1 (fignl1) and fidgetin-like 2 (fignl2) were found to be highly expressed during zebrafish brain development, suggesting their functions in the nervous system. In this study, we report the effects of loss-of-function of these genes on development. We designed and identified single-guide RNAs targeted to generate fign, fignl1, and fignl2 mutants and then observed the overall morphological and behavioral changes. Our findings showed that while fign and fignl1 null mutants displayed no significant defects, fignl2 null zebrafish mutants displayed pericardial edema, reduced heart rate, and smaller eyes; fignl2 null mutants responded to the light-darkness shift with a lower swimming velocity. fignl2 mRNAs were identified in vascular endothelial cells by in situ hybridization and re-analysis of an online dataset of single-cell RNAseq results. Finally, we used morpholino oligonucleotides to confirm that fignl2 knockdown resulted in severe heart edema, which was caused by abnormal vascular branching. The zebrafish fignl2 morphants also showed longer axonal length and more branches of caudal primary neurons. Taken together, we summarize that Fignl2 functions on cellular branches in endothelial cells and neurons. This study reported for the first time that the microtubule-severing protein Fignl2 contributes to cell branching during development.
Collapse
Affiliation(s)
- Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xu Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuanyuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Run Zhuo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaona Lai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
48
|
Retrograde Mitochondrial Transport Is Essential for Organelle Distribution and Health in Zebrafish Neurons. J Neurosci 2020; 41:1371-1392. [PMID: 33376159 PMCID: PMC7896009 DOI: 10.1523/jneurosci.1316-20.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
In neurons, mitochondria are transported by molecular motors throughout the cell to form and maintain functional neural connections. These organelles have many critical functions in neurons and are of high interest as their dysfunction is associated with disease. While the mechanics and impact of anterograde mitochondrial movement toward axon terminals are beginning to be understood, the frequency and function of retrograde (cell body directed) mitochondrial transport in neurons are still largely unexplored. While existing evidence indicates that some mitochondria are retrogradely transported for degradation in the cell body, the precise impact of disrupting retrograde transport on the organelles and the axon was unknown. Using long-term, in vivo imaging, we examined mitochondrial motility in zebrafish sensory and motor axons. We show that retrograde transport of mitochondria from axon terminals allows replacement of the axon terminal population within a day. By tracking these organelles, we show that not all mitochondria that leave the axon terminal are degraded; rather, they persist over several days. Disrupting retrograde mitochondrial flux in neurons leads to accumulation of aged organelles in axon terminals and loss of cell body mitochondria. Assays of neural circuit activity demonstrated that disrupting mitochondrial transport and function has no effect on sensory axon terminal activity but does negatively impact motor neuron axons. Taken together, our work supports a previously unappreciated role for retrograde mitochondrial transport in the maintenance of a homeostatic distribution of mitochondria in neurons and illustrates the downstream effects of disrupting this process on sensory and motor circuits. SIGNIFICANCE STATEMENT Disrupted mitochondrial transport has been linked to neurodegenerative disease. Retrograde transport of this organelle has been implicated in turnover of aged organelles through lysosomal degradation in the cell body. Consistent with this, we provide evidence that retrograde mitochondrial transport is important for removing aged organelles from axons; however, we show that these organelles are not solely degraded, rather they persist in neurons for days. Disrupting retrograde mitochondrial transport impacts the homeostatic distribution of mitochondria throughout the neuron and the function of motor, but not sensory, axon synapses. Together, our work shows the conserved reliance on retrograde mitochondrial transport for maintaining a healthy mitochondrial pool in neurons and illustrates the disparate effects of disrupting this process on sensory versus motor circuits.
Collapse
|
49
|
Zelada D, Bermedo-García F, Collao N, Henríquez JP. Motor function recovery: deciphering a regenerative niche at the neuromuscular synapse. Biol Rev Camb Philos Soc 2020; 96:752-766. [PMID: 33336525 PMCID: PMC7986695 DOI: 10.1111/brv.12675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022]
Abstract
The coordinated movement of many organisms relies on efficient nerve–muscle communication at the neuromuscular junction (NMJ), a peripheral synapse composed of a presynaptic motor axon terminal, a postsynaptic muscle specialization, and non‐myelinating terminal Schwann cells. NMJ dysfunctions are caused by traumatic spinal cord or peripheral nerve injuries as well as by severe motor pathologies. Compared to the central nervous system, the peripheral nervous system displays remarkable regenerating abilities; however, this capacity is limited by the denervation time frame and depends on the establishment of permissive regenerative niches. At the injury site, detailed information is available regarding the cells, molecules, and mechanisms involved in nerve regeneration and repair. However, a regenerative niche at the final functional step of peripheral motor innervation, i.e. at the mature neuromuscular synapse, has not been deciphered. In this review, we integrate classic and recent evidence describing the cells and molecules that could orchestrate a dynamic ecosystem to accomplish successful NMJ regeneration. We propose that such a regenerative niche must ensure at least two fundamental steps for successful NMJ regeneration: the proper arrival of incoming regenerating axons to denervated postsynaptic muscle domains, and the resilience of those postsynaptic domains, in morphological and functional terms. We here describe and combine the main cellular and molecular responses involved in each of these steps as potential targets to help successful NMJ regeneration.
Collapse
Affiliation(s)
- Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Francisca Bermedo-García
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Nicolás Collao
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Juan P Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| |
Collapse
|
50
|
Hypothalamic Pomc Neurons Innervate the Spinal Cord and Modulate the Excitability of Premotor Circuits. Curr Biol 2020; 30:4579-4593.e7. [PMID: 32976803 DOI: 10.1016/j.cub.2020.08.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 07/30/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022]
Abstract
Locomotion requires energy, yet animals need to increase locomotion in order to find and consume food in energy-deprived states. While such energy homeostatic coordination suggests brain origin, whether the central melanocortin 4 receptor (Mc4r) system directly modulates locomotion through motor circuits is unknown. Here, we report that hypothalamic Pomc neurons in zebrafish and mice have long-range projections into spinal cord regions harboring Mc4r-expressing V2a interneurons, crucial components of the premotor networks. Furthermore, in zebrafish, Mc4r activation decreases the excitability of spinal V2a neurons as well as swimming and foraging, while systemic or V2a neuron-specific blockage of Mc4r promotes locomotion. In contrast, in mice, electrophysiological recordings revealed that two-thirds of V2a neurons in lamina X are excited by the Mc4r agonist α-MSH, and acute inhibition of Mc4r signaling reduces locomotor activity. In addition, we found other Mc4r neurons in spinal lamina X that are inhibited by α-MSH, which is in line with previous studies in rodents where Mc4r agonists reduced locomotor activity. Collectively, our studies identify spinal V2a interneurons as evolutionary conserved second-order neurons of the central Mc4r system, providing a direct anatomical and functional link between energy homeostasis and locomotor control systems. The net effects of this modulatory system on locomotor activity can vary between different vertebrate species and, possibly, even within one species. We discuss the biological sense of this phenomenon in light of the ambiguity of locomotion on energy balance and the different living conditions of the different species.
Collapse
|