1
|
Malik T, Sidisky JM, Jones S, Winters A, Hocking B, Rotay J, Huhulea EN, Moran S, Connors B, Babcock DT. Synaptic defects in adult drosophila motor neurons in a model of amyotrophic lateral sclerosis. Hum Mol Genet 2025:ddaf068. [PMID: 40327885 DOI: 10.1093/hmg/ddaf068] [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: 09/27/2024] [Revised: 03/26/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that primarily affects motor neurons in the brain and spinal cord. Like other neurodegenerative diseases, defects in synaptic integrity are among the earliest hallmarks of ALS. However, the specific impairments to synaptic integrity remain unclear. To better understand synaptic defects in ALS, we expressed either wild-type or mutant Fused in Sarcoma (FUS), an RNA binding protein that is often mis-localized in ALS, in adult motor neurons. Using optogenetic stimulation of the motor neurons innervating the Ventral Abdominal Muscles (VAMs), we found that expression of mutant FUS disrupted the functional integrity of these synapses. This functional deficit was followed by disruption of synaptic gross morphology, localization of pre- and post-synaptic proteins, and cytoskeleton integrity. We found similar synaptic defects using the motor neurons innervating the Dorsal Longitudinal Muscles (DLMs), where expression of mutant FUS resulted in a progressive loss of flight ability along with disruption of active zone distribution. Our findings uncover defects in synaptic function that precede changes in synaptic structure, suggesting that synaptic function is more sensitive to this ALS model. Highlighting the earliest synaptic defects in this disease model should help to identify strategies for preventing later stages of disease progression.
Collapse
Affiliation(s)
- Tulika Malik
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Jessica M Sidisky
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Sam Jones
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Alexander Winters
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Brandon Hocking
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Jocelyn Rotay
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Ellen N Huhulea
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Sara Moran
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Bali Connors
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| | - Daniel T Babcock
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem PA, 18045 United States
| |
Collapse
|
2
|
Thakur RS, O'Connor-Giles KM. PDZD8 promotes autophagy at ER-lysosome membrane contact sites to regulate activity-dependent synaptic growth. Cell Rep 2025; 44:115483. [PMID: 40156832 DOI: 10.1016/j.celrep.2025.115483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/07/2025] [Accepted: 03/07/2025] [Indexed: 04/01/2025] Open
Abstract
Building synaptic connections requires coordinating a host of cellular activities from cell signaling to protein turnover, placing a high demand on intracellular communication. Membrane contact sites (MCSs) formed between organelles have emerged as key signaling hubs for coordinating diverse cellular activities, yet their roles in the developing nervous system remain obscure. We investigate the in vivo function of the endoplasmic reticulum (ER) MCS tethering and lipid-transfer protein PDZD8, which was recently linked to intellectual disability, in the nervous system. We find that PDZD8 is required for activity-dependent synaptic bouton formation in multiple paradigms. PDZD8 is sufficient to drive excess synaptic bouton formation through an autophagy-dependent mechanism and required for synapse development when autophagy is limited. PDZD8 accelerates autophagic flux by promoting lysosome maturation at ER-late endosome/lysosome MCSs. We propose that PDZD8 functions in the nervous system to increase autophagy during periods of high demand, including activity-dependent synaptic growth.
Collapse
Affiliation(s)
- Rajan S Thakur
- Department of Neuroscience, Brown University, Providence, RI, USA.
| | - Kate M O'Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI, USA; Carney Institute for Brain Science, Providence, RI, USA.
| |
Collapse
|
3
|
Astacio H, Bykhovskaia M. High frequency stimulation activates hot spots of spontaneous synaptic transmission. Front Synaptic Neurosci 2025; 17:1539868. [PMID: 40297638 PMCID: PMC12034645 DOI: 10.3389/fnsyn.2025.1539868] [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: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Neuronal transmitters are released at the morphological specializations known as active zones (AZs). Transmitters can be released either in response to a stimulus or spontaneously, and spontaneous transmission is a vital component of neuronal communication. Employing postsynaptically tethered calcium sensor GCaMP, we investigated how nerve stimulation affects spontaneous transmission at individual AZs at the Drosophila neuromuscular synapse. Optical monitoring of spontaneous transmission at individual AZs revealed that prolonged high-frequency stimulation (HFS, 30 Hz for 1 min) selectively activates the hot spots of spontaneous transmission, including the individual AZs with elevated activities as well as AZ clusters. In contrast, a brief tetanus (2 s) activated numerous low-activity AZs. We employed Monte-Carlo simulations of spontaneous transmission based on a three-state model of AZ preparedness, which incorporated longer-lasting (minutes) and shorter-lasting (sub-seconds to seconds) high-activity states of AZs. The simulations produced an accurate quantitative description of the variability and time-course of spontaneous transmission at individual AZs before and after the stimulation and suggested that HFS activates both longer-lasting and shorter-lasting states of AZ preparedness.
Collapse
Affiliation(s)
| | - Maria Bykhovskaia
- Department of Neurology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
4
|
Akbergenova Y, Matthias J, Littleton JT. Active zone maturation state controls synaptic output and release mode and is differentially regulated by neuronal activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636302. [PMID: 39975213 PMCID: PMC11838553 DOI: 10.1101/2025.02.03.636302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Synapse formation requires the gradual accumulation of cytomatrix proteins and voltage-gated Ca2+ channels (VDCCs) at presynaptic active zones (AZs) to support neurotransmitter release. To correlate AZ maturation with synaptic output, quantal imaging was performed at serially imaged time-stamped Drosophila synapses. Evoked release strength correlated strongly with AZ age and accumulation of late AZ scaffolds, while immature sites lacking VDCC accumulation supported spontaneous release. To examine how neuronal activity regulates AZ maturation and protein accumulation, the effects of disruptions to SV fusion or action potential generation were analyzed. Decreasing neuronal activity reduced AZ seeding and caused hyperaccumulation of presynaptic material at existing AZs. Although enlarged AZs are also observed in rab3 mutants, activity reduction acted through an independent mechanism that required postsynaptic glutamate receptor-dependent signaling. Together, these data indicate AZ maturation state sets distinct presynaptic release modes and output strength, with neuronal activity shaping both AZ number and size across development.
Collapse
Affiliation(s)
- Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
5
|
Garduño‐Tamayo NA, Almazán JL, Romo‐Rodríguez R, Valle‐García D, Meza‐Sosa KF, Pérez‐Domínguez M, Pelayo R, Pedraza‐Alva G, Pérez‐Martínez L. Klf10 Regulates the Emergence of Glial Phenotypes During Hypothalamic Development. J Neurosci Res 2025; 103:e70020. [PMID: 39924964 PMCID: PMC11808290 DOI: 10.1002/jnr.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/26/2024] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Abstract
Glial cells play a pivotal role in the Central Nervous System (CNS), constituting most brain cells. Gliogenesis, crucial in CNS development, occurs after neurogenesis. In the hypothalamus, glial progenitors first generate oligodendrocytes and later astrocytes. However, the precise molecular mechanisms governing the emergence of glial lineages in the developing hypothalamus remain incompletely understood. This study reveals the pivotal role of the transcription factor KLF10 in regulating the emergence of both astrocyte and oligodendrocyte lineages during embryonic hypothalamic development. Through transcriptomic and bioinformatic analyses, we identified novel KLF10 putative target genes, which play important roles in the differentiation of neurons, astrocytes, and oligodendrocytes. Notably, in the absence of KLF10, there is an increase in the oligodendrocyte population, while the astrocyte population decreases in the embryonic hypothalamus. Strikingly, this decline in the number of astrocytes persists into adulthood, indicating that the absence of KLF10 leads to an extended period of oligodendrocyte emergence while delaying the appearance of astrocytes. Our findings also unveil a novel signaling pathway for Klf10 gene expression regulation. We demonstrate that Klf10 is a target of CREB and that its expression is upregulated via the BDNF-p38-CREB pathway. Thus, we postulate that KLF10 is an integral part of the hypothalamic developmental program that ensures the correct timing for glial phenotypes' generation. Importantly, we propose that the Klf10-/- mouse model represents a valuable tool for investigating the impact of reduced astrocyte and microglia populations in the homeostasis of the adult hypothalamus.
Collapse
Affiliation(s)
- Norma Angelica Garduño‐Tamayo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Jorge Luis Almazán
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Rubí Romo‐Rodríguez
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de OrienteDelegación PueblaPueblaMexico
| | - David Valle‐García
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Karla F. Meza‐Sosa
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Martha Pérez‐Domínguez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Rosana Pelayo
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de OrienteDelegación PueblaPueblaMexico
| | - Gustavo Pedraza‐Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| | - Leonor Pérez‐Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de BiotecnologíaUniversidad Nacional Autónoma de México (UNAM)CuernavacaMorelosMexico
| |
Collapse
|
6
|
Ermanoska B, Baets J, Rodal AA. Non-muscle myosin II regulates presynaptic actin assemblies and neuronal mechanobiology in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.10.566609. [PMID: 38014140 PMCID: PMC10680633 DOI: 10.1101/2023.11.10.566609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Neuromuscular junctions (NMJs) are evolutionarily ancient, specialized contacts between neurons and muscles. They endure mechanical strain from muscle contractions throughout life, but cellular mechanisms for managing this stress remain unclear. Here we identify a novel actomyosin structure at Drosophila larval NMJs, consisting of a long-lived, low-turnover presynaptic actin core that co-localizes with non-muscle myosin II (NMII). This core is likely to have contractile properties, as manipulating neuronal NMII levels or activity disrupts its organization. Intriguingly, depleting neuronal NMII triggered changes in postsynaptic muscle NMII levels and organization near synapses, suggesting transsynaptic propagation of actomyosin rearrangements. We also found reduced levels of Integrin adhesion receptors both pre- and postsynaptically upon NMII knockdown, indicating disrupted neuron-muscle connections. Mechanical severing of axons caused similar actin core fragmentation and Integrin loss to NMII depletion, suggesting this structure responds to tension. Our findings reveal a presynaptic actomyosin assembly that maintains mechanical continuity between neurons and muscle, possibly facilitating mechanotransduction at the NMJ via Integrin-mediated adhesion.
Collapse
Affiliation(s)
| | - Jonathan Baets
- Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | | |
Collapse
|
7
|
von Saucken VE, Windner SE, Armetta G, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. J Cell Biol 2025; 224:e202404052. [PMID: 39475469 PMCID: PMC11530350 DOI: 10.1083/jcb.202404052] [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: 04/10/2024] [Revised: 09/13/2024] [Accepted: 10/13/2024] [Indexed: 11/04/2024] Open
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (size scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show that local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
- Biochemistry, Cell and Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giovanna Armetta
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
8
|
Lee JY, Gala DS, Kiourlappou M, Olivares-Abril J, Joha J, Titlow JS, Teodoro RO, Davis I. Murine glial protrusion transcripts predict localized Drosophila glial mRNAs involved in plasticity. J Cell Biol 2024; 223:e202306152. [PMID: 39037431 PMCID: PMC11262410 DOI: 10.1083/jcb.202306152] [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: 06/30/2023] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
The polarization of cells often involves the transport of specific mRNAs and their localized translation in distal projections. Neurons and glia are both known to contain long cytoplasmic processes, while localized transcripts have only been studied extensively in neurons, not glia, especially in intact nervous systems. Here, we predict 1,740 localized Drosophila glial transcripts by extrapolating from our meta-analysis of seven existing studies characterizing the localized transcriptomes and translatomes of synaptically associated mammalian glia. We demonstrate that the localization of mRNAs in mammalian glial projections strongly predicts the localization of their high-confidence Drosophila homologs in larval motor neuron-associated glial projections and are highly statistically enriched for genes associated with neurological diseases. We further show that some of these localized glial transcripts are specifically required in glia for structural plasticity at the nearby neuromuscular junction synapses. We conclude that peripheral glial mRNA localization is a common and conserved phenomenon and propose that it is likely to be functionally important in disease.
Collapse
Affiliation(s)
- Jeffrey Y. Lee
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Dalia S. Gala
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | | - Jana Joha
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Rita O. Teodoro
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ilan Davis
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Dresselhaus EC, Harris KP, Blanchette CR, Koles K, Del Signore SJ, Pescosolido MF, Ermanoska B, Rozencwaig M, Soslowsky RC, Parisi MJ, Stewart BA, Mosca TJ, Rodal AA. ESCRT disruption provides evidence against trans-synaptic signaling via extracellular vesicles. J Cell Biol 2024; 223:e202405025. [PMID: 38842573 PMCID: PMC11157088 DOI: 10.1083/jcb.202405025] [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: 05/05/2024] [Revised: 05/20/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Extracellular vesicles (EVs) are released by many cell types, including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating endosomal sorting complex required for transport (ESCRT) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo evenness interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell-autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.
Collapse
Affiliation(s)
| | - Kathryn P. Harris
- Office of the Vice-Principal, Research and Innovation, University of Toronto Mississauga, Mississauga, Canada
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | | | | | | - Mark Rozencwaig
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | | - Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA, USA
| | - Avital A. Rodal
- Department of Biology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
10
|
Sidisky JM, Winters A, Caratenuto R, Babcock DT. Synaptic defects in a drosophila model of muscular dystrophy. Front Cell Neurosci 2024; 18:1381112. [PMID: 38812789 PMCID: PMC11133739 DOI: 10.3389/fncel.2024.1381112] [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: 02/02/2024] [Accepted: 04/08/2024] [Indexed: 05/31/2024] Open
Abstract
Muscular dystrophies are a devastating class of diseases that result in a progressive loss of muscle integrity. Duchenne Muscular Dystrophy, the most prevalent form of Muscular Dystrophy, is due to the loss of functional Dystrophin. While much is known regarding destruction of muscle tissue in these diseases, much less is known regarding the synaptic defects that also occur in these diseases. Synaptic defects are also among the earliest hallmarks of neurodegenerative diseases, including the neuromuscular disease Amyotrophic Lateral Sclerosis (ALS). Our current study investigates synaptic defects within adult muscle tissues as well as presynaptic motor neurons in Drosophila dystrophin mutants. Here we demonstrate that the progressive, age-dependent loss of flight ability in dystrophin mutants is accompanied by disorganization of Neuromuscular Junctions (NMJs), including impaired localization of both presynaptic and postsynaptic markers. We show that these synaptic defects, including presynaptic defects within motor neurons, are due to the loss of Dystrophin specifically within muscles. These results should help to better understand the early synaptic defects preceding cell loss in neuromuscular disorders.
Collapse
Affiliation(s)
- Jessica M. Sidisky
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
- Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Cambridge, MA, United States
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Alex Winters
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Russell Caratenuto
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Daniel T. Babcock
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| |
Collapse
|
11
|
Dresselhaus EC, Harris KP, Blanchette CR, Koles K, Del Signore SJ, Pescosolido MF, Ermanoska B, Rozencwaig M, Soslowsky RC, Parisi MJ, Stewart BA, Mosca TJ, Rodal AA. ESCRT disruption provides evidence against transsynaptic signaling functions for extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.22.537920. [PMID: 38746182 PMCID: PMC11092503 DOI: 10.1101/2023.04.22.537920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Extracellular vesicles (EVs) are released by many cell types including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating ESCRT (endosomal sorting complex required for transport) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo Evenness Interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.
Collapse
Affiliation(s)
| | - Kathryn P. Harris
- Office of the Vice-Principal, Research and Innovation, University of Toronto, Mississauga, Mississauga, Canada
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA
| | | | | | | | | | | | - Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA
| | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada; Department of Cell and Systems Biology University of Toronto, Toronto, Canada
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA
| | | |
Collapse
|
12
|
Mousavi H, Rimaz M, Zeynizadeh B. Practical Three-Component Regioselective Synthesis of Drug-Like 3-Aryl(or heteroaryl)-5,6-dihydrobenzo[ h]cinnolines as Potential Non-Covalent Multi-Targeting Inhibitors To Combat Neurodegenerative Diseases. ACS Chem Neurosci 2024; 15:1828-1881. [PMID: 38647433 DOI: 10.1021/acschemneuro.4c00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Neurodegenerative diseases (NDs) are one of the prominent health challenges facing contemporary society, and many efforts have been made to overcome and (or) control it. In this research paper, we described a practical one-pot two-step three-component reaction between 3,4-dihydronaphthalen-1(2H)-one (1), aryl(or heteroaryl)glyoxal monohydrates (2a-h), and hydrazine monohydrate (NH2NH2•H2O) for the regioselective preparation of some 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnoline derivatives (3a-h). After synthesis and characterization of the mentioned cinnolines (3a-h), the in silico multi-targeting inhibitory properties of these heterocyclic scaffolds have been investigated upon various Homo sapiens-type enzymes, including hMAO-A, hMAO-B, hAChE, hBChE, hBACE-1, hBACE-2, hNQO-1, hNQO-2, hnNOS, hiNOS, hPARP-1, hPARP-2, hLRRK-2(G2019S), hGSK-3β, hp38α MAPK, hJNK-3, hOGA, hNMDA receptor, hnSMase-2, hIDO-1, hCOMT, hLIMK-1, hLIMK-2, hRIPK-1, hUCH-L1, hPARK-7, and hDHODH, which have confirmed their functions and roles in the neurodegenerative diseases (NDs), based on molecular docking studies, and the obtained results were compared with a wide range of approved drugs and well-known (with IC50, EC50, etc.) compounds. In addition, in silico ADMET prediction analysis was performed to examine the prospective drug properties of the synthesized heterocyclic compounds (3a-h). The obtained results from the molecular docking studies and ADMET-related data demonstrated that these series of 3-aryl(or heteroaryl)-5,6-dihydrobenzo[h]cinnolines (3a-h), especially hit ones, can really be turned into the potent core of new drugs for the treatment of neurodegenerative diseases (NDs), and/or due to the having some reactionable locations, they are able to have further organic reactions (such as cross-coupling reactions), and expansion of these compounds (for example, with using other types of aryl(or heteroaryl)glyoxal monohydrates) makes a new avenue for designing novel and efficient drugs for this purpose.
Collapse
Affiliation(s)
- Hossein Mousavi
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| | - Mehdi Rimaz
- Department of Chemistry, Payame Noor University, P.O. Box 19395-3697, Tehran 19395-3697, Iran
| | - Behzad Zeynizadeh
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia 5756151818, Iran
| |
Collapse
|
13
|
Chang YC, Gao Y, Lee JY, Peng YJ, Langen J, Chang KT. Identification of secretory autophagy as a mechanism modulating activity-induced synaptic remodeling. Proc Natl Acad Sci U S A 2024; 121:e2315958121. [PMID: 38588427 PMCID: PMC11032469 DOI: 10.1073/pnas.2315958121] [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: 09/14/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood, as neurodevelopment and structural plasticity are tightly linked. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity and synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We report that while both synapse development and activity-induced synaptic remodeling at the fly NMJ require macroautophagy (hereafter referred to as autophagy), bifurcation in the autophagy pathway differentially impacts development and synaptic plasticity. We demonstrate that neuronal activity enhances autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a transsynaptic signaling mechanism modulating synaptic plasticity.
Collapse
Affiliation(s)
- Yen-Ching Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yuan Gao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Joo Yeun Lee
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yi-Jheng Peng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Jennifer Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Karen T. Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
14
|
von Saucken VE, Windner SE, Baylies MK. Postsynaptic BMP signaling regulates myonuclear properties in Drosophila larval muscles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588944. [PMID: 38645063 PMCID: PMC11030338 DOI: 10.1101/2024.04.10.588944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The syncytial mammalian muscle fiber contains a heterogeneous population of (myo)nuclei. At the neuromuscular junction (NMJ), myonuclei have specialized positioning and gene expression. However, it remains unclear how myonuclei are recruited and what regulates myonuclear output at the NMJ. Here, we identify specific properties of myonuclei located near the Drosophila larval NMJ. These synaptic myonuclei have increased size in relation to their surrounding cytoplasmic domain (scaling), increased DNA content (ploidy), and increased levels of transcription factor pMad, a readout for BMP signaling activity. Our genetic manipulations show local BMP signaling affects muscle size, nuclear size, ploidy, and NMJ size and function. In support, RNA sequencing analysis reveals that pMad regulates genes involved in muscle growth, ploidy (i.e., E2f1), and neurotransmission. Our data suggest that muscle BMP signaling instructs synaptic myonuclear output that then positively shapes the NMJ synapse. This study deepens our understanding of how myonuclear heterogeneity supports local signaling demands to fine tune cellular function and NMJ activity.
Collapse
Affiliation(s)
- Victoria E. von Saucken
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
- Weill Cornell-Rockefeller-Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10065 USA
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065 USA
| | - Stefanie E. Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Mary K. Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| |
Collapse
|
15
|
Dominicci-Cotto C, Vazquez M, Marie B. The Wingless planar cell polarity pathway is essential for optimal activity-dependent synaptic plasticity. Front Synaptic Neurosci 2024; 16:1322771. [PMID: 38633293 PMCID: PMC11021733 DOI: 10.3389/fnsyn.2024.1322771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
From fly to man, the Wingless (Wg)/Wnt signaling molecule is essential for both the stability and plasticity of the nervous system. The Drosophila neuromuscular junction (NMJ) has proven to be a useful system for deciphering the role of Wg in directing activity-dependent synaptic plasticity (ADSP), which, in the motoneuron, has been shown to be dependent on both the canonical and the noncanonical calcium Wg pathways. Here we show that the noncanonical planar cell polarity (PCP) pathway is an essential component of the Wg signaling system controlling plasticity at the motoneuron synapse. We present evidence that disturbing the PCP pathway leads to a perturbation in ADSP. We first show that a PCP-specific allele of disheveled (dsh) affects the de novo synaptic structures produced during ADSP. We then show that the Rho GTPases downstream of Dsh in the PCP pathway are also involved in regulating the morphological changes that take place after repeated stimulation. Finally, we show that Jun kinase is essential for this phenomenon, whereas we found no indication of the involvement of the transcription factor complex AP1 (Jun/Fos). This work shows the involvement of the neuronal PCP signaling pathway in supporting ADSP. Because we find that AP1 mutants can perform ADSP adequately, we hypothesize that, upon Wg activation, the Rho GTPases and Jun kinase are involved locally at the synapse, in instructing cytoskeletal dynamics responsible for the appearance of the morphological changes occurring during ADSP.
Collapse
Affiliation(s)
- Carihann Dominicci-Cotto
- Department of Anatomy and Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, United States
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, United States
| | - Mariam Vazquez
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, United States
- Molecular Sciences Research Center, University of Puerto Rico, San Juan, PR, United States
| | - Bruno Marie
- Department of Anatomy and Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, United States
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, United States
- Molecular Sciences Research Center, University of Puerto Rico, San Juan, PR, United States
| |
Collapse
|
16
|
Akiyama T, Raftery LA, Wharton KA. Bone morphogenetic protein signaling: the pathway and its regulation. Genetics 2024; 226:iyad200. [PMID: 38124338 PMCID: PMC10847725 DOI: 10.1093/genetics/iyad200] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/27/2023] [Indexed: 12/23/2023] Open
Abstract
In the mid-1960s, bone morphogenetic proteins (BMPs) were first identified in the extracts of bone to have the remarkable ability to induce heterotopic bone. When the Drosophila gene decapentaplegic (dpp) was first identified to share sequence similarity with mammalian BMP2/BMP4 in the late-1980s, it became clear that secreted BMP ligands can mediate processes other than bone formation. Following this discovery, collaborative efforts between Drosophila geneticists and mammalian biochemists made use of the strengths of their respective model systems to identify BMP signaling components and delineate the pathway. The ability to conduct genetic modifier screens in Drosophila with relative ease was critical in identifying the intracellular signal transducers for BMP signaling and the related transforming growth factor-beta/activin signaling pathway. Such screens also revealed a host of genes that encode other core signaling components and regulators of the pathway. In this review, we provide a historical account of this exciting time of gene discovery and discuss how the field has advanced over the past 30 years. We have learned that while the core BMP pathway is quite simple, composed of 3 components (ligand, receptor, and signal transducer), behind the versatility of this pathway lies multiple layers of regulation that ensures precise tissue-specific signaling output. We provide a sampling of these discoveries and highlight many questions that remain to be answered to fully understand the complexity of BMP signaling.
Collapse
Affiliation(s)
- Takuya Akiyama
- Department of Biology, Rich and Robin Porter Cancer Research Center, The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Laurel A Raftery
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology, and Biochemistry, Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
17
|
DePew AT, Bruckner JJ, O’Connor-Giles KM, Mosca TJ. Neuronal LRP4 directs the development, maturation, and cytoskeletal organization of peripheral synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.564481. [PMID: 37961323 PMCID: PMC10635100 DOI: 10.1101/2023.11.03.564481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synapse development requires multiple signaling pathways to accomplish the myriad of steps needed to ensure a successful connection. Transmembrane receptors on the cell surface are optimally positioned to facilitate communication between the synapse and the rest of the neuron and often function as synaptic organizers to synchronize downstream signaling events. One such organizer, the LDL receptor-related protein LRP4, is a cell surface receptor most well-studied postsynaptically at mammalian neuromuscular junctions. Recent work, however, has identified emerging roles for LRP4 as a presynaptic molecule, but how LRP4 acts as a presynaptic organizer, what roles LRP4 plays in organizing presynaptic biology, and the downstream mechanisms of LRP4 are not well understood. Here we show that LRP4 functions presynaptically at Drosophila neuromuscular synapses, acting in motor neurons to instruct multiple aspects of pre- and postsynaptic development. Loss of presynaptic LRP4 results in a range of developmental defects, impairing active zone organization, synapse growth, physiological function, microtubule organization, synaptic ultrastructure, and synapse maturation. We further demonstrate that LRP4 promotes most aspects of presynaptic development via a downstream SR-protein kinase, SRPK79D. SRPK79D overexpression suppresses synaptic defects associated with loss of lrp4. These data demonstrate a function for LRP4 as a peripheral synaptic organizer acting presynaptically, highlight a downstream mechanism conserved with its CNS function, and indicate previously unappreciated roles for LRP4 in cytoskeletal organization, synapse maturation, and active zone organization, underscoring its developmental importance.
Collapse
Affiliation(s)
- Alison T. DePew
- Dept. of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Joseph J. Bruckner
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Kate M. O’Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI 02912 USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Timothy J. Mosca
- Dept. of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107 USA
- Lead Contact
| |
Collapse
|
18
|
Chang YC, Gao Y, Lee JY, Langen J, Chang KT. Identification of secretory autophagy as a novel mechanism modulating activity-induced synaptic remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.560931. [PMID: 38328055 PMCID: PMC10849665 DOI: 10.1101/2023.10.06.560931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity from synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We further demonstrate that neuronal activity stimulates autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a novel trans-synaptic signaling mechanism modulating structural plasticity.
Collapse
|
19
|
Furusawa K, Ishii K, Tsuji M, Tokumitsu N, Hasegawa E, Emoto K. Presynaptic Ube3a E3 ligase promotes synapse elimination through down-regulation of BMP signaling. Science 2023; 381:1197-1205. [PMID: 37708280 DOI: 10.1126/science.ade8978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Inactivation of the ubiquitin ligase Ube3a causes the developmental disorder Angelman syndrome, whereas increased Ube3a dosage is associated with autism spectrum disorders. Despite the enriched localization of Ube3a in the axon terminals including presynapses, little is known about the presynaptic function of Ube3a and mechanisms underlying its presynaptic localization. We show that developmental synapse elimination requires presynaptic Ube3a activity in Drosophila neurons. We further identified the domain of Ube3a that is required for its interaction with the kinesin motor. Angelman syndrome-associated missense mutations in the interaction domain attenuate presynaptic targeting of Ube3a and prevent synapse elimination. Conversely, increased Ube3a activity in presynapses leads to precocious synapse elimination and impairs synaptic transmission. Our findings reveal the physiological role of Ube3a and suggest potential pathogenic mechanisms associated with Ube3a dysregulation.
Collapse
Affiliation(s)
- Kotaro Furusawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kenichi Ishii
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masato Tsuji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nagomi Tokumitsu
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Eri Hasegawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
20
|
Fernandes AR, Martins JP, Gomes ER, Mendes CS, Teodoro RO. Drosophila motor neuron boutons remodel through membrane blebbing coupled with muscle contraction. Nat Commun 2023; 14:3352. [PMID: 37291089 PMCID: PMC10250368 DOI: 10.1038/s41467-023-38421-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Wired neurons form new presynaptic boutons in response to increased synaptic activity, however the mechanism(s) by which this occurs remains uncertain. Drosophila motor neurons (MNs) have clearly discernible boutons that display robust structural plasticity, being therefore an ideal system in which to study activity-dependent bouton genesis. Here, we show that in response to depolarization and in resting conditions, MNs form new boutons by membrane blebbing, a pressure-driven mechanism that occurs in 3-D cell migration, but to our knowledge not previously described to occur in neurons. Accordingly, F-actin is decreased in boutons during outgrowth, and non-muscle myosin-II is dynamically recruited to newly formed boutons. Furthermore, muscle contraction plays a mechanical role, which we hypothesize promotes bouton addition by increasing MN confinement. Overall, we identified a mechanism by which established circuits form new boutons allowing their structural expansion and plasticity, using trans-synaptic physical forces as the main driving force.
Collapse
Affiliation(s)
- Andreia R Fernandes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - João P Martins
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - César S Mendes
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Rita O Teodoro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
21
|
Cheng X, Yan Z, Su Z, Liu J. The transforming growth factor beta ligand TIG-2 modulates the function of neuromuscular junction and muscle energy metabolism in Caenorhabditis elegans. Front Mol Neurosci 2022; 15:962974. [PMID: 36385772 PMCID: PMC9650414 DOI: 10.3389/fnmol.2022.962974] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Deciphering the physiological function of TGF-β (the transforming growth factor beta) family ligands is import for understanding the role of TGF-β in animals' development and aging. Here, we investigate the function of TIG-2, one of the ligands in Caenorhabditis elegans TGF-β family, in animals' behavioral modulation. Our results show that a loss-of-function mutation in tig-2 gene result in slower locomotion speed in the early adulthood and an increased density of cholinergic synapses, but a decreased neurotransmitter release at neuromuscular junctions (NMJs). Further tissue-specific rescue results reveal that neuronal and intestinal TIG-2 are essential for the formation of cholinergic synapses at NMJs. Interestingly, tig-2(ok3416) mutant is characterized with reduced muscle mitochondria content and adenosine triphosphate (ATP) production, although the function of muscle acetylcholine receptors and the morphology muscle fibers in the mutant are comparable to that in wild-type animals. Our result suggests that TIG-2 from different neuron and intestine regulates worm locomotion by modulating synaptogenesis and neurotransmission at NMJs, as well as energy metabolism in postsynaptic muscle cells.
Collapse
Affiliation(s)
- Xinran Cheng
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Zhenzhen Yan
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Zexiong Su
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Jie Liu
- Neuroscience Program, Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
22
|
AP2 Regulates Thickveins Trafficking to Attenuate NMJ Growth Signaling in Drosophila. eNeuro 2022; 9:ENEURO.0044-22.2022. [PMID: 36180220 PMCID: PMC9581581 DOI: 10.1523/eneuro.0044-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 09/09/2022] [Accepted: 09/23/2022] [Indexed: 12/15/2022] Open
Abstract
Compromised endocytosis in neurons leads to synapse overgrowth and altered organization of synaptic proteins. However, the molecular players and the signaling pathways which regulate the process remain poorly understood. Here, we show that σ2-adaptin, one of the subunits of the AP2-complex, genetically interacts with Mad, Medea and Dad (components of BMP signaling) to control neuromuscular junction (NMJ) growth in Drosophila Ultrastructural analysis of σ2-adaptin mutants show an accumulation of large vesicles and membranous structures akin to endosomes at the synapse. We found that mutations in σ2-adaptin lead to an accumulation of Tkv receptors at the presynaptic membrane. Interestingly, the level of small GTPase Rab11 was significantly reduced in the σ2-adaptin mutant synapses. However, expression of Rab11 does not restore the synaptic defects of σ2-adaptin mutations. We propose a model in which AP2 regulates Tkv internalization and endosomal recycling to control synaptic growth.
Collapse
|
23
|
Kim YJ. Activity-induced synaptic structural modifications by Akt. Biochem Biophys Res Commun 2022; 621:94-100. [PMID: 35820284 DOI: 10.1016/j.bbrc.2022.06.093] [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: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
The activity-dependent regulation of synaptic structures plays a key role in synaptic development and plasticity; however, the signaling mechanisms involved remain largely unknown. The serine/threonine protein kinase Akt, a downstream effector of phosphoinositide 3-kinase (PI3K), plays a pivotal role in a wide range of physiological functions. We focused on the importance of Akt in rapid synaptic structural changes after stimulation at the Drosophila neuromuscular junction, a well-studied model synapse. Compared with wild-type larvae, akt mutants showed significantly reduced muscle size and an increased number of boutons per area, suggesting that Akt is required for proper pre- and postsynaptic growth. In addition, the level of cysteine string protein (CSP) was significantly increased, and its distribution was different in akt mutants. After high K+ single stimulation, the CSP level of akt mutant NMJs increased dramatically compared with that of wild-type NMJs. Interestingly, ghost boutons without postsynaptic specialization were found in akt mutant NMJs, and the number of these boutons was significantly increased by patterned stimulation. In contrast, the postsynaptic change in the subsynaptic reticulum (SSR) in the akt mutant occurred independent of stimulation. These results suggest that Akt functions in both pre- and postsynaptic growth and differentiation, and in particular, presynaptic action occurs in an activity-dependent manner.
Collapse
Affiliation(s)
- Yoon-Jung Kim
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, South Korea.
| |
Collapse
|
24
|
Restrepo LJ, DePew AT, Moese ER, Tymanskyj SR, Parisi MJ, Aimino MA, Duhart JC, Fei H, Mosca TJ. γ-secretase promotes Drosophila postsynaptic development through the cleavage of a Wnt receptor. Dev Cell 2022; 57:1643-1660.e7. [PMID: 35654038 DOI: 10.1016/j.devcel.2022.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 12/27/2022]
Abstract
Developing synapses mature through the recruitment of specific proteins that stabilize presynaptic and postsynaptic structure and function. Wnt ligands signaling via Frizzled (Fz) receptors play many crucial roles in neuronal and synaptic development, but whether and how Wnt and Fz influence synaptic maturation is incompletely understood. Here, we show that Fz2 receptor cleavage via the γ-secretase complex is required for postsynaptic development and maturation. In the absence of γ-secretase, Drosophila neuromuscular synapses fail to recruit postsynaptic scaffolding and cytoskeletal proteins, leading to behavioral deficits. Introducing presenilin mutations linked to familial early-onset Alzheimer's disease into flies leads to synaptic maturation phenotypes that are identical to those seen in null alleles. This conserved role for γ-secretase in synaptic maturation and postsynaptic development highlights the importance of Fz2 cleavage and suggests that receptor processing by proteins linked to neurodegeneration may be a shared mechanism with aspects of synaptic development.
Collapse
Affiliation(s)
- Lucas J Restrepo
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Alison T DePew
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Elizabeth R Moese
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Stephen R Tymanskyj
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael J Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael A Aimino
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Juan Carlos Duhart
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Hong Fei
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Timothy J Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA.
| |
Collapse
|
25
|
Vicidomini R, Serpe M. Local BMP signaling: A sensor for synaptic activity that balances synapse growth and function. Curr Top Dev Biol 2022; 150:211-254. [PMID: 35817503 PMCID: PMC11102767 DOI: 10.1016/bs.ctdb.2022.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Synapse development is coordinated by intercellular communication between the pre- and postsynaptic compartments, and by neuronal activity itself. In flies as in vertebrates, neuronal activity induces input-specific changes in the synaptic strength so that the entire circuit maintains stable function in the face of many challenges, including changes in synapse number and strength. But how do neurons sense synapse activity? In several studies carried out using the Drosophila neuromuscular junction (NMJ), we demonstrated that local BMP signaling provides an exquisite sensor for synapse activity. Here we review the main features of this exquisite sensor and discuss its functioning beyond monitoring the synapse activity but rather as a key controller that operates in coordination with other BMP signaling pathways to balance synapse growth, maturation and function.
Collapse
Affiliation(s)
- Rosario Vicidomini
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mihaela Serpe
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shiver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
26
|
Blanchette CR, Scalera AL, Harris KP, Zhao Z, Dresselhaus EC, Koles K, Yeh A, Apiki JK, Stewart BA, Rodal AA. Local regulation of extracellular vesicle traffic by the synaptic endocytic machinery. J Cell Biol 2022; 221:e202112094. [PMID: 35320349 PMCID: PMC8952828 DOI: 10.1083/jcb.202112094] [Citation(s) in RCA: 9] [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: 12/17/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Neuronal extracellular vesicles (EVs) are locally released from presynaptic terminals, carrying cargoes critical for intercellular signaling and disease. EVs are derived from endosomes, but it is unknown how these cargoes are directed to the EV pathway rather than for conventional endolysosomal degradation. Here, we find that endocytic machinery plays an unexpected role in maintaining a release-competent pool of EV cargoes at synapses. Endocytic mutants, including nervous wreck (nwk), shibire/dynamin, and AP-2, unexpectedly exhibit local presynaptic depletion specifically of EV cargoes. Accordingly, nwk mutants phenocopy synaptic plasticity defects associated with loss of the EV cargo synaptotagmin-4 (Syt4) and suppress lethality upon overexpression of the EV cargo amyloid precursor protein (APP). These EV defects are genetically separable from canonical endocytic functions in synaptic vesicle recycling and synaptic growth. Endocytic machinery opposes the endosomal retromer complex to regulate EV cargo levels and acts upstream of synaptic cargo removal by retrograde axonal transport. Our data suggest a novel molecular mechanism that locally promotes cargo loading into synaptic EVs.
Collapse
Affiliation(s)
| | | | - Kathryn P. Harris
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Zechuan Zhao
- Department of Biology, Brandeis University, Waltham, MA
| | | | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA
| | - Anna Yeh
- Department of Biology, Brandeis University, Waltham, MA
| | | | - Bryan A. Stewart
- Department of Biology, University of Toronto Mississauga, Mississauga, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | | |
Collapse
|
27
|
Ribba AS, Fraboulet S, Sadoul K, Lafanechère L. The Role of LIM Kinases during Development: A Lens to Get a Glimpse of Their Implication in Pathologies. Cells 2022; 11:cells11030403. [PMID: 35159213 PMCID: PMC8834001 DOI: 10.3390/cells11030403] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/24/2022] Open
Abstract
The organization of cell populations within animal tissues is essential for the morphogenesis of organs during development. Cells recognize three-dimensional positions with respect to the whole organism and regulate their cell shape, motility, migration, polarization, growth, differentiation, gene expression and cell death according to extracellular signals. Remodeling of the actin filaments is essential to achieve these cell morphological changes. Cofilin is an important binding protein for these filaments; it increases their elasticity in terms of flexion and torsion and also severs them. The activity of cofilin is spatiotemporally inhibited via phosphorylation by the LIM domain kinases 1 and 2 (LIMK1 and LIMK2). Phylogenetic analysis indicates that the phospho-regulation of cofilin has evolved as a mechanism controlling the reorganization of the actin cytoskeleton during complex multicellular processes, such as those that occur during embryogenesis. In this context, the main objective of this review is to provide an update of the respective role of each of the LIM kinases during embryonic development.
Collapse
|
28
|
Ariawan D, Au C, Paric E, Fath T, Ke YD, Kassiou M, van Eersel J, Ittner LM. The Nature of Diamino Linker and Halogen Bonding Define Selectivity of Pyrrolopyrimidine-Based LIMK1 Inhibitors. Front Chem 2021; 9:781213. [PMID: 34966720 PMCID: PMC8711653 DOI: 10.3389/fchem.2021.781213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/16/2021] [Indexed: 12/23/2022] Open
Abstract
The LIM-domain kinase (LIMK) family consists of two isoforms, LIMK1 and LIMK2, which are highly homologous, making selective inhibitor development challenging. LIMK regulates dynamics of the actin cytoskeleton, thereby impacting many cellular functions including cell morphology and motility. Here, we designed and synthesised analogues of a known pyrrolopyrimidine LIMK inhibitor with moderate selectivity for LIMK1 over LIMK2 to gain insights into which features contribute to both activity and selectivity. We incorporated a different stereochemistry around a cyclohexyl central moiety to achieve better selectivity for different LIMK isoforms. Inhibitory activity was assessed by kinase assays, and biological effects in cells were determined using an in vitro wound closure assay. Interestingly, a slight change in stereochemistry alters LIMK isoform selectivity. Finally, a docking study was performed to predict how the new compounds interact with the target.
Collapse
Affiliation(s)
- Daryl Ariawan
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Carol Au
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Esmeralda Paric
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Thomas Fath
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yazi D Ke
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Darlington, NSW, Australia
| | - Janet van Eersel
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Lars M Ittner
- Dementia Research Centre, Department of Biomedical Science, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
29
|
Maldonado-Díaz C, Vazquez M, Marie B. A comparison of three different methods of eliciting rapid activity-dependent synaptic plasticity at the Drosophila NMJ. PLoS One 2021; 16:e0260553. [PMID: 34847197 PMCID: PMC8631638 DOI: 10.1371/journal.pone.0260553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/11/2021] [Indexed: 11/29/2022] Open
Abstract
The Drosophila NMJ is a system of choice for investigating the mechanisms underlying the structural and functional modifications evoked during activity-dependent synaptic plasticity. Because fly genetics allows considerable versatility, many strategies can be employed to elicit this activity. Here, we compare three different stimulation methods for eliciting activity-dependent changes in structure and function at the Drosophila NMJ. We find that the method using patterned stimulations driven by a K+-rich solution creates robust structural modifications but reduces muscle viability, as assessed by resting potential and membrane resistance. We argue that, using this method, electrophysiological studies that consider the frequency of events, rather than their amplitude, are the only reliable studies. We contrast these results with the expression of CsChrimson channels and red-light stimulation at the NMJ, as well as with the expression of TRPA channels and temperature stimulation. With both these methods we observed reliable modifications of synaptic structures and consistent changes in electrophysiological properties. Indeed, we observed a rapid appearance of immature boutons that lack postsynaptic differentiation, and a potentiation of spontaneous neurotransmission frequency. Surprisingly, a patterned application of temperature changes alone is sufficient to provoke both structural and functional plasticity. In this context, temperature-dependent TRPA channel activation induces additional structural plasticity but no further increase in the frequency of spontaneous neurotransmission, suggesting an uncoupling of these mechanisms.
Collapse
Affiliation(s)
- Carolina Maldonado-Díaz
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Mariam Vazquez
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Bruno Marie
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
- Department of Anatomy & Neurobiology, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
30
|
Decapentaplegic Acutely Defines the Connectivity of Central Pacemaker Neurons in Drosophila. J Neurosci 2021; 41:8338-8350. [PMID: 34429376 DOI: 10.1523/jneurosci.0397-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/12/2021] [Accepted: 08/11/2021] [Indexed: 11/21/2022] Open
Abstract
Rhythmic rest-activity cycles are controlled by an endogenous clock. In Drosophila, this clock resides in ∼150 neurons organized in clusters whose hierarchy changes in response to environmental conditions. The concerted activity of the circadian network is necessary for the adaptive responses to synchronizing environmental stimuli. Thus far, work was devoted to unravel the logic of the coordination of different clusters focusing on neurotransmitters and neuropeptides. We further explored communication in the adult male brain through ligands belonging to the bone morphogenetic protein (BMP) pathway. Herein we show that the lateral ventral neurons (LNvs) express the small morphogen decapentaplegic (DPP). DPP expression in the large LNvs triggered a period lengthening phenotype, the downregulation of which caused reduced rhythmicity and affected anticipation at dawn and dusk, underscoring DPP per se conveys time-of-day relevant information. Surprisingly, DPP expression in the large LNvs impaired circadian remodeling of the small LNv axonal terminals, likely through local modulation of the guanine nucleotide exchange factor Trio. These findings open the provocative possibility that the BMP pathway is recruited to strengthen/reduce the connectivity among specific clusters along the day and thus modulate the contribution of the clusters to the circadian network.SIGNIFICANCE STATEMENT The circadian clock relies on the communication between groups of so-called clock neurons to coordinate physiology and behavior to the optimal times across the day, predicting and adapting to a changing environment. The circadian network relies on neurotransmitters and neuropeptides to fine-tune connectivity among clock neurons and thus give rise to a coherent output. Herein we show that decapentaplegic, a ligand belonging to the BMP retrograde signaling pathway required for coordinated growth during development, is recruited by a group of circadian neurons in the adult brain to trigger structural remodeling of terminals on a daily basis.
Collapse
|
31
|
Del Signore SJ, Kelley CF, Messelaar EM, Lemos T, Marchan MF, Ermanoska B, Mund M, Fai TG, Kaksonen M, Rodal AA. An autoinhibitory clamp of actin assembly constrains and directs synaptic endocytosis. eLife 2021; 10:69597. [PMID: 34324418 PMCID: PMC8321554 DOI: 10.7554/elife.69597] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 01/05/2023] Open
Abstract
Synaptic membrane-remodeling events such as endocytosis require force-generating actin assembly. The endocytic machinery that regulates these actin and membrane dynamics localizes at high concentrations to large areas of the presynaptic membrane, but actin assembly and productive endocytosis are far more restricted in space and time. Here we describe a mechanism whereby autoinhibition clamps the presynaptic endocytic machinery to limit actin assembly to discrete functional events. We found that collective interactions between the Drosophila endocytic proteins Nwk/FCHSD2, Dap160/intersectin, and WASp relieve Nwk autoinhibition and promote robust membrane-coupled actin assembly in vitro. Using automated particle tracking to quantify synaptic actin dynamics in vivo, we discovered that Nwk-Dap160 interactions constrain spurious assembly of WASp-dependent actin structures. These interactions also promote synaptic endocytosis, suggesting that autoinhibition both clamps and primes the synaptic endocytic machinery, thereby constraining actin assembly to drive productive membrane remodeling in response to physiological cues. Neurons constantly talk to each other by sending chemical signals across the tiny gap, or ‘synapse’, that separates two cells. While inside the emitting cell, these molecules are safely packaged into small, membrane-bound vessels. Upon the right signal, the vesicles fuse with the external membrane of the neuron and spill their contents outside, for the receiving cell to take up and decode. The emitting cell must then replenish its vesicle supply at the synapse through a recycling mechanism known as endocytosis. To do so, it uses dynamically assembling rod-like ‘actin’ filaments, which work in concert with many other proteins to pull in patches of membrane as new vesicles. The proteins that control endocytosis and actin assembly abound at neuronal synapses, and, when mutated, are linked to many neurological diseases. Unlike other cell types, neurons appear to ‘pre-deploy’ these actin-assembly proteins to synaptic membranes, but to keep them inactive under normal conditions. How neurons control the way this machinery is recruited and activated remains unknown. To investigate this question, Del Signore et al. conducted two sets of studies. First, they exposed actin to several different purified proteins in initial ‘test tube’ experiments. This revealed that, depending on the conditions, a group of endocytosis proteins could prevent or promote actin assembly: assembly occurred only if the proteins were associated with membranes. Next, Del Signore et al. mutated these proteins in fruit fly larvae, and performed live cell microscopy to determine their impact on actin assembly and endocytosis. Consistent with the test tube findings, endocytosis mutants had more actin assembly overall, implying that the proteins were required to prevent random actin assembly. However, the same mutants had reduced levels of endocytosis, suggesting that the proteins were also necessary for productive actin assembly. Together, these experiments suggest that, much like a mousetrap holds itself poised ready to spring, some endocytic proteins play a dual role to restrain actin assembly when and where it is not needed, and to promote it at sites of endocytosis. These results shed new light on how neurons might build and maintain effective, working synapses. Del Signore et al. hope that this knowledge may help to better understand and combat neurological diseases, such as Alzheimer’s, which are linked to impaired membrane traffic and cell signalling.
Collapse
Affiliation(s)
| | | | | | - Tania Lemos
- Department of Biology, Brandeis University, Walltham, United States
| | | | | | - Markus Mund
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Thomas G Fai
- Department of Mathematics, Brandeis University, Waltham, United States
| | - Marko Kaksonen
- Department of Biochemistry and NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
32
|
Function of Drosophila Synaptotagmins in membrane trafficking at synapses. Cell Mol Life Sci 2021; 78:4335-4364. [PMID: 33619613 PMCID: PMC8164606 DOI: 10.1007/s00018-021-03788-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
The Synaptotagmin (SYT) family of proteins play key roles in regulating membrane trafficking at neuronal synapses. Using both Ca2+-dependent and Ca2+-independent interactions, several SYT isoforms participate in synchronous and asynchronous fusion of synaptic vesicles (SVs) while preventing spontaneous release that occurs in the absence of stimulation. Changes in the function or abundance of the SYT1 and SYT7 isoforms alter the number and route by which SVs fuse at nerve terminals. Several SYT family members also regulate trafficking of other subcellular organelles at synapses, including dense core vesicles (DCV), exosomes, and postsynaptic vesicles. Although SYTs are linked to trafficking of multiple classes of synaptic membrane compartments, how and when they interact with lipids, the SNARE machinery and other release effectors are still being elucidated. Given mutations in the SYT family cause disorders in both the central and peripheral nervous system in humans, ongoing efforts are defining how these proteins regulate vesicle trafficking within distinct neuronal compartments. Here, we review the Drosophila SYT family and examine their role in synaptic communication. Studies in this invertebrate model have revealed key similarities and several differences with the predicted activity of their mammalian counterparts. In addition, we highlight the remaining areas of uncertainty in the field and describe outstanding questions on how the SYT family regulates membrane trafficking at nerve terminals.
Collapse
|
33
|
Aponte-Santiago NA, Littleton JT. Synaptic Properties and Plasticity Mechanisms of Invertebrate Tonic and Phasic Neurons. Front Physiol 2020; 11:611982. [PMID: 33391026 PMCID: PMC7772194 DOI: 10.3389/fphys.2020.611982] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Defining neuronal cell types and their associated biophysical and synaptic diversity has become an important goal in neuroscience as a mechanism to create comprehensive brain cell atlases in the post-genomic age. Beyond broad classification such as neurotransmitter expression, interneuron vs. pyramidal, sensory or motor, the field is still in the early stages of understanding closely related cell types. In both vertebrate and invertebrate nervous systems, one well-described distinction related to firing characteristics and synaptic release properties are tonic and phasic neuronal subtypes. In vertebrates, these classes were defined based on sustained firing responses during stimulation (tonic) vs. transient responses that rapidly adapt (phasic). In crustaceans, the distinction expanded to include synaptic release properties, with tonic motoneurons displaying sustained firing and weaker synapses that undergo short-term facilitation to maintain muscle contraction and posture. In contrast, phasic motoneurons with stronger synapses showed rapid depression and were recruited for short bursts during fast locomotion. Tonic and phasic motoneurons with similarities to those in crustaceans have been characterized in Drosophila, allowing the genetic toolkit associated with this model to be used for dissecting the unique properties and plasticity mechanisms for these neuronal subtypes. This review outlines general properties of invertebrate tonic and phasic motoneurons and highlights recent advances that characterize distinct synaptic and plasticity pathways associated with two closely related glutamatergic neuronal cell types that drive invertebrate locomotion.
Collapse
Affiliation(s)
- Nicole A. Aponte-Santiago
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - J. Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
34
|
Chou VT, Johnson SA, Van Vactor D. Synapse development and maturation at the drosophila neuromuscular junction. Neural Dev 2020; 15:11. [PMID: 32741370 PMCID: PMC7397595 DOI: 10.1186/s13064-020-00147-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Synapses are the sites of neuron-to-neuron communication and form the basis of the neural circuits that underlie all animal cognition and behavior. Chemical synapses are specialized asymmetric junctions between a presynaptic neuron and a postsynaptic target that form through a series of diverse cellular and subcellular events under the control of complex signaling networks. Once established, the synapse facilitates neurotransmission by mediating the organization and fusion of synaptic vesicles and must also retain the ability to undergo plastic changes. In recent years, synaptic genes have been implicated in a wide array of neurodevelopmental disorders; the individual and societal burdens imposed by these disorders, as well as the lack of effective therapies, motivates continued work on fundamental synapse biology. The properties and functions of the nervous system are remarkably conserved across animal phyla, and many insights into the synapses of the vertebrate central nervous system have been derived from studies of invertebrate models. A prominent model synapse is the Drosophila melanogaster larval neuromuscular junction, which bears striking similarities to the glutamatergic synapses of the vertebrate brain and spine; further advantages include the simplicity and experimental versatility of the fly, as well as its century-long history as a model organism. Here, we survey findings on the major events in synaptogenesis, including target specification, morphogenesis, and the assembly and maturation of synaptic specializations, with a emphasis on work conducted at the Drosophila neuromuscular junction.
Collapse
Affiliation(s)
- Vivian T Chou
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Seth A Johnson
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Guangming G, Junhua G, Chenchen Z, Yang M, Wei X. Neurexin and Neuroligins Maintain the Balance of Ghost and Satellite Boutons at the Drosophila Neuromuscular Junction. Front Neuroanat 2020; 14:19. [PMID: 32581727 PMCID: PMC7296126 DOI: 10.3389/fnana.2020.00019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/26/2020] [Indexed: 11/22/2022] Open
Abstract
Neurexins and neuroligins are common synaptic adhesion molecules that are associated with autism and interact with each other in the synaptic cleft. The Drosophila neuromuscular junction (NMJ) bouton is a well-known model system in neuroscience, and ghost and satellite boutons, respectively, indicate the poor development and overgrowth of the NMJ boutons. However, the Drosophila neurexin (DNrx) and Drosophila neuroligins (DNlgs) are mainly observed in type Ib boutons, indicating the ultrastructural and developmental phenotypes of the Drosophila NMJ. Here, we identified the ultrastructural and developmental features of ghost and satellite boutons by utilizing dneurexin (dnrx) and dneuroligins (dnlgs) fly mutants and other associated fly strains. Ghost boutons contain synaptic vesicles with multiple diameters but very rarely contain T-bar structures and swollen or thin subsynaptic reticulum (SSR) membranes. The muscle cell membrane is invaginated at different sites, stretches to the ghost bouton from different directions, forms several layers that enwrap the ghost bouton, and then branches into the complex SSR. Satellite boutons share a common SSR membrane and present either a typical profile in which a main bouton is encircled by small boutons or two atypical profiles in which the small boutons are grouped together or distributed in beads without a main bouton. Electron and confocal microscopy data showed that dnrx, dnlg1, dnlg2, dnlg3, and dnlg4 mutations led to ghost boutons; the overexpression of dnrx, dnlg1, dnlg2, dnlg3, and dnlg4 led to satellite boutons; and the dnlg2;dnlg3 double mutation also led to satellite boutons. These results suggested that DNrx and DNlgs jointly maintain the development and function of NMJ boutons by regulating the balance of ghost and satellite boutons in Drosophila.
Collapse
Affiliation(s)
- Gan Guangming
- School of Medicine, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Geng Junhua
- The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhang Chenchen
- School of Medicine, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Mou Yang
- School of Medicine, Southeast University, Nanjing, China
| | - Xie Wei
- The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, China.,Institute of Life Sciences, The Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| |
Collapse
|
36
|
Lnenicka GA. Crayfish and Drosophila NMJs. Neurosci Lett 2020; 732:135110. [PMID: 32497734 DOI: 10.1016/j.neulet.2020.135110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 01/06/2023]
Abstract
Many synaptic studies have utilized the experimental advantages of the Arthropod NMJ and the most prominent preparations have been the crayfish and Drosophila larval NMJs. Early cellular studies in the crayfish established the framework for later molecular studies in Drosophila. The two neuromuscular systems are compared including the advantages presented by each preparation for cellular analysis. Beginning with the early work in the crayfish, research developments are followed in the areas of structure/function relationships, activity-dependent synaptic plasticity/development and synaptic homeostasis. A reoccurring theme in these studies is the regulation of active zone structure and function. Early studies in the crayfish focused on the role of active zone number/size and possible functional heterogeneity in regulating transmitter release. Recent studies in Drosophila have begun to characterize this heterogeneity using new approaches that combine imaging of transmitter release, Ca2+ influx and molecular composition for individual active zones.
Collapse
Affiliation(s)
- Gregory A Lnenicka
- Department of Biological Sciences, University at Albany, SUNY, Albany, NY 12222, United States.
| |
Collapse
|
37
|
Guan Z, Quiñones-Frías MC, Akbergenova Y, Littleton JT. Drosophila Synaptotagmin 7 negatively regulates synaptic vesicle release and replenishment in a dosage-dependent manner. eLife 2020; 9:e55443. [PMID: 32343229 PMCID: PMC7224696 DOI: 10.7554/elife.55443] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023] Open
Abstract
Synchronous neurotransmitter release is triggered by Ca2+ binding to the synaptic vesicle protein Synaptotagmin 1, while asynchronous fusion and short-term facilitation is hypothesized to be mediated by plasma membrane-localized Synaptotagmin 7 (SYT7). We generated mutations in Drosophila Syt7 to determine if it plays a conserved role as the Ca2+ sensor for these processes. Electrophysiology and quantal imaging revealed evoked release was elevated 2-fold. Syt7 mutants also had a larger pool of readily-releasable vesicles, faster recovery following stimulation, and intact facilitation. Syt1/Syt7 double mutants displayed more release than Syt1 mutants alone, indicating SYT7 does not mediate the residual asynchronous release remaining in the absence of SYT1. SYT7 localizes to an internal membrane tubular network within the peri-active zone, but does not enrich at active zones. These findings indicate the two Ca2+ sensor model of SYT1 and SYT7 mediating all phases of neurotransmitter release and facilitation is not applicable at Drosophila synapses.
Collapse
Affiliation(s)
- Zhuo Guan
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Monica C Quiñones-Frías
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
38
|
Naik AS, Lin JM, Taroc EZM, Katreddi RR, Frias JA, Lemus AA, Sammons MA, Forni PE. Smad4-dependent morphogenic signals control the maturation and axonal targeting of basal vomeronasal sensory neurons to the accessory olfactory bulb. Development 2020; 147:147/8/dev184036. [PMID: 32341026 PMCID: PMC7197725 DOI: 10.1242/dev.184036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
The vomeronasal organ (VNO) contains two main types of vomeronasal sensory neurons (VSNs) that express distinct vomeronasal receptor (VR) genes and localize to specific regions of the neuroepithelium. Morphogenic signals are crucial in defining neuronal identity and network formation; however, if and what signals control maturation and homeostasis of VSNs is largely unexplored. Here, we found transforming growth factor β (TGFβ) and bone morphogenetic protein (BMP) signal transduction in postnatal mice, with BMP signaling being restricted to basal VSNs and at the marginal zones of the VNO: the site of neurogenesis. Using different Smad4 conditional knockout mouse models, we disrupted canonical TGFβ/BMP signaling in either maturing basal VSNs (bVSNs) or all mature VSNs. Smad4 loss of function in immature bVSNs compromises dendritic knob formation, pheromone induced activation, correct glomeruli formation in the accessory olfactory bulb (AOB) and survival. However, Smad4 loss of function in all mature VSNs only compromises correct glomeruli formation in the posterior AOB. Our results indicate that Smad4-mediated signaling drives the functional maturation and connectivity of basal VSNs. Summary: Genetic disruption of TGFβ/BMP signaling in maturing basal vomeronasal sensory neurons (VSNs) or in all mature VSNs indicates that Smad4 signaling drives maturation and connectivity of basal VSNs.
Collapse
Affiliation(s)
- Ankana S Naik
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jennifer M Lin
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Ed Zandro M Taroc
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Raghu R Katreddi
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jesus A Frias
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Alex A Lemus
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Paolo E Forni
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
39
|
Berke B, Le L, Keshishian H. Target-dependent retrograde signaling mediates synaptic plasticity at the Drosophila neuromuscular junction. Dev Neurobiol 2020; 79:895-912. [PMID: 31950660 DOI: 10.1002/dneu.22731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/26/2022]
Abstract
Neurons that innervate multiple targets often establish synapses with target-specific strengths, and local forms of synaptic plasticity. We have examined the molecular-genetic mechanisms that allow a single Drosophila motoneuron, the ventral Common Exciter (vCE), to establish connections with target-specific properties at its various synaptic partners. By driving transgenes in a subset of vCE's targets, we found that individual target cells are able to independently control the properties of vCE's innervating branch and synapses. This is achieved by means of a trans-synaptic growth factor secreted by the target cell. At the larval neuromuscular junction, postsynaptic glutamate receptor activity stimulates the release of the BMP4/5/6 homolog Glass bottom boat (Gbb). As larvae mature and motoneuron terminals grow, Gbb activates the R-Smad transcriptional regulator phosphorylated Mad (pMad) to facilitate presynaptic development. We found that manipulations affecting glutamate receptors or Gbb within subsets of target muscles led to local effects either specific to the manipulated muscle or by a limited gradient within the presynaptic branches. While presynaptic development depends on pMad transcriptional activity within the motoneuron nucleus, we find that the Gbb growth factor may also act locally within presynaptic terminals. Local Gbb signaling and presynaptic pMad accumulation within boutons may therefore participate in a "synaptic tagging" mechanism, to influence synaptic growth and plasticity in Drosophila.
Collapse
Affiliation(s)
- Brett Berke
- Molecular, Cellular, and Developmental Biology Department, Yale University, New Haven, CT, USA
| | - Linh Le
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Haig Keshishian
- Molecular, Cellular, and Developmental Biology Department, Yale University, New Haven, CT, USA
| |
Collapse
|
40
|
Chou VT, Johnson S, Long J, Vounatsos M, Van Vactor D. dTACC restricts bouton addition and regulates microtubule organization at the Drosophila neuromuscular junction. Cytoskeleton (Hoboken) 2020; 77:4-15. [PMID: 31702858 PMCID: PMC7027520 DOI: 10.1002/cm.21578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022]
Abstract
Regulation of the synaptic cytoskeleton is essential to proper neuronal development and wiring. Perturbations in neuronal microtubules (MTs) are associated with numerous pathologies, yet it remains unclear how changes in MTs may be coupled to synapse morphogenesis. Studies have identified many MT regulators that promote synapse growth. However, less is known about the factors that restrict growth, despite the potential links of synaptic overgrowth to severe neurological conditions. Here, we report that dTACC, which is implicated in MT assembly and stability, prevents synapse overgrowth at the Drosophila neuromuscular junction by restricting addition of new boutons throughout larval development. dTACC localizes to the axonal MT lattice and is required to maintain tubulin levels and the integrity of higher-order MT structures in motor axon terminals. While previous reports have demonstrated the roles of MT-stabilizing proteins in promoting synapse growth, our findings suggest that in certain contexts, MT stabilization may correlate with restricted growth.
Collapse
Affiliation(s)
- Vivian T. Chou
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Seth Johnson
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Jennifer Long
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Maxime Vounatsos
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - David Van Vactor
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| |
Collapse
|
41
|
Hoover KM, Gratz SJ, Qi N, Herrmann KA, Liu Y, Perry-Richardson JJ, Vanderzalm PJ, O'Connor-Giles KM, Broihier HT. The calcium channel subunit α 2δ-3 organizes synapses via an activity-dependent and autocrine BMP signaling pathway. Nat Commun 2019; 10:5575. [PMID: 31811118 PMCID: PMC6898181 DOI: 10.1038/s41467-019-13165-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022] Open
Abstract
Synapses are highly specialized for neurotransmitter signaling, yet activity-dependent growth factor release also plays critical roles at synapses. While efficient neurotransmitter signaling relies on precise apposition of release sites and neurotransmitter receptors, molecular mechanisms enabling high-fidelity growth factor signaling within the synaptic microenvironment remain obscure. Here we show that the auxiliary calcium channel subunit α2δ-3 promotes the function of an activity-dependent autocrine Bone Morphogenetic Protein (BMP) signaling pathway at the Drosophila neuromuscular junction (NMJ). α2δ proteins have conserved synaptogenic activity, although how they execute this function has remained elusive. We find that α2δ-3 provides an extracellular scaffold for an autocrine BMP signal, suggesting a mechanistic framework for understanding α2δ's conserved role in synapse organization. We further establish a transcriptional requirement for activity-dependent, autocrine BMP signaling in determining synapse density, structure, and function. We propose that activity-dependent, autocrine signals provide neurons with continuous feedback on their activity state for modulating both synapse structure and function.
Collapse
Affiliation(s)
- Kendall M Hoover
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, RI, 02912, USA
| | - Nova Qi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Kelsey A Herrmann
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yizhou Liu
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jahci J Perry-Richardson
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Pamela J Vanderzalm
- Department of Biology, John Carroll University, University Heights, OH, 44118, USA
| | | | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
42
|
Wang W, Halasz E, Townes-Anderson E. Actin Dynamics, Regulated by RhoA-LIMK-Cofilin Signaling, Mediates Rod Photoreceptor Axonal Retraction After Retinal Injury. Invest Ophthalmol Vis Sci 2019; 60:2274-2285. [PMID: 31112612 PMCID: PMC6530517 DOI: 10.1167/iovs.18-26077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Purpose Retraction of the axon terminals of rod photoreceptors after retinal detachment breaks the first synapse in the visual pathway, resulting in visual impairment. Previous work showed that the mechanism of axonal retraction involves RhoA signaling and its downstream effector LIM Kinase (LIMK) activation. We examined the response of the downstream component cofilin, a direct binding protein of actin filaments, as well as the regulation by RhoA-LIMK-Cofilin signaling of actin assembly/disassembly, in the presynaptic ribbon terminal of injured rod cells. Methods Injury was produced by retinal detachment or rod cell isolation. Detached porcine retina was probed for levels and localization of phosphorylated cofilin with Western blots and confocal microscopy, whereas rod cell cultures of dissociated salamander retina were examined for filamentous actin assembly/disassembly with a barbed end assay and phalloidin staining. Results A detachment increased phosphorylation of cofilin in retinal explants; phosphorylation occurred in rod terminals in sections of detached retina. Isolation of rod cells resulted in axon retraction accompanied by an increase in actin barbed ends and a decrease in net filament labeling. All changes were significantly reduced by either Rho kinase (ROCK) or LIMK inhibition, using Y27632 or BMS-5, respectively. Cytochalasin D also reduced retraction and stabilized filaments in isolated rod cells. Conclusions These results indicate that actin depolymerization via activation of RhoA downstream kinases and cofilin contributes to axon retraction. Preventing depolymerization, in addition to actomyosin contraction, may stabilize ribbon synapses after trauma.
Collapse
Affiliation(s)
- Weiwei Wang
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Eva Halasz
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| | - Ellen Townes-Anderson
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Graduate School of Biomedical Sciences, Rutgers Biomedical and Health Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey, United States
| |
Collapse
|
43
|
Ojelade SA, Lee TV, Giagtzoglou N, Yu L, Ugur B, Li Y, Duraine L, Zuo Z, Petyuk V, De Jager PL, Bennett DA, Arenkiel BR, Bellen HJ, Shulman JM. cindr, the Drosophila Homolog of the CD2AP Alzheimer's Disease Risk Gene, Is Required for Synaptic Transmission and Proteostasis. Cell Rep 2019; 28:1799-1813.e5. [PMID: 31412248 PMCID: PMC6703184 DOI: 10.1016/j.celrep.2019.07.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/30/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
The Alzheimer's disease (AD) susceptibility gene, CD2-associated protein (CD2AP), encodes an actin binding adaptor protein, but its function in the nervous system is largely unknown. Loss of the Drosophila ortholog cindr enhances neurotoxicity of human Tau, which forms neurofibrillary tangle pathology in AD. We show that Cindr is expressed in neurons and present at synaptic terminals. cindr mutants show impairments in synapse maturation and both synaptic vesicle recycling and release. Cindr associates and genetically interacts with 14-3-3ζ, regulates the ubiquitin-proteasome system, and affects turnover of Synapsin and the plasma membrane calcium ATPase (PMCA). Loss of cindr elevates PMCA levels and reduces cytosolic calcium. Studies of Cd2ap null mice support a conserved role in synaptic proteostasis, and CD2AP protein levels are inversely related to Synapsin abundance in human postmortem brains. Our results reveal CD2AP neuronal requirements with relevance to AD susceptibility, including for proteostasis, calcium handling, and synaptic structure and function.
Collapse
Affiliation(s)
- Shamsideen A Ojelade
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Tom V Lee
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nikolaos Giagtzoglou
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Berrak Ugur
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yarong Li
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lita Duraine
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vlad Petyuk
- Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute, Columbia University Medical Center, New York, NY 10032, USA; Cell Circuits Program, Broad Institute, Cambridge, MA 02142, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Benjamin R Arenkiel
- Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
44
|
Two Pathways for the Activity-Dependent Growth and Differentiation of Synaptic Boutons in Drosophila. eNeuro 2019; 6:ENEURO.0060-19.2019. [PMID: 31387877 PMCID: PMC6709223 DOI: 10.1523/eneuro.0060-19.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/15/2019] [Accepted: 07/26/2019] [Indexed: 11/21/2022] Open
Abstract
Synapse formation can be promoted by intense activity. At the Drosophila larval neuromuscular junction (NMJ), new synaptic boutons can grow acutely in response to patterned stimulation. We combined confocal imaging with electron microscopy and tomography to investigate the initial stages of growth and differentiation of new presynaptic boutons at the Drosophila NMJ. We found that the new boutons can form rapidly in intact larva in response to intense crawling activity, and we observed two different patterns of bouton formation and maturation. The first pathway involves the growth of filopodia followed by a formation of boutons that are initially devoid of synaptic vesicles (SVs) but filled with filamentous matrix. The second pathway involves rapid budding of synaptic boutons packed with SVs, and these more mature boutons are sometimes capable of exocytosis/endocytosis. We demonstrated that intense activity predominantly promotes the second pathway, i.e., budding of more mature boutons filled with SVs. We also showed that this pathway depends on synapsin (Syn), a neuronal protein which reversibly associates with SVs and mediates their clustering via a protein kinase A (PKA)-dependent mechanism. Finally, we took advantage of the temperature-sensitive mutant sei to demonstrate that seizure activity can promote very rapid budding of new boutons filled with SVs, and this process occurs at scale of minutes. Altogether, these results demonstrate that intense activity acutely and selectively promotes rapid budding of new relatively mature presynaptic boutons filled with SVs, and that this process is regulated via a PKA/Syn-dependent pathway.
Collapse
|
45
|
Anbalagan S, Blechman J, Gliksberg M, Gordon L, Rotkopf R, Dadosh T, Shimoni E, Levkowitz G. Robo2 regulates synaptic oxytocin content by affecting actin dynamics. eLife 2019; 8:45650. [PMID: 31180321 PMCID: PMC6590984 DOI: 10.7554/elife.45650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/08/2019] [Indexed: 12/28/2022] Open
Abstract
The regulation of neuropeptide level at the site of release is essential for proper neurophysiological functions. We focused on a prominent neuropeptide, oxytocin (OXT) in the zebrafish as an in vivo model to visualize and quantify OXT content at the resolution of a single synapse. We found that OXT-loaded synapses were enriched with polymerized actin. Perturbation of actin filaments by either cytochalasin-D or conditional Cofilin expression resulted in decreased synaptic OXT levels. Genetic loss of robo2 or slit3 displayed decreased synaptic OXT content and robo2 mutants displayed reduced mobility of the actin probe Lifeact-EGFP in OXT synapses. Using a novel transgenic reporter allowing real-time monitoring of OXT-loaded vesicles, we show that robo2 mutants display slower rate of vesicles accumulation. OXT-specific expression of dominant-negative Cdc42, which is a key regulator of actin dynamics and a downstream effector of Robo2, led to a dose-dependent increase in OXT content in WT, and a dampened effect in robo2 mutants. Our results link Slit3-Robo2-Cdc42, which controls local actin dynamics, with the maintenance of synaptic neuropeptide levels.
Collapse
Affiliation(s)
- Savani Anbalagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, LSCF, Weizmann Institute of Science, Rehovot, Israel.,Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Shimoni
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
46
|
Semaphorin4D Induces Inhibitory Synapse Formation by Rapid Stabilization of Presynaptic Boutons via MET Coactivation. J Neurosci 2019; 39:4221-4237. [PMID: 30914448 DOI: 10.1523/jneurosci.0215-19.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 11/21/2022] Open
Abstract
Changes in inhibitory connections are essential for experience-dependent circuit adaptations. Defects in inhibitory synapses are linked to neurodevelopmental disorders, but the molecular processes underlying inhibitory synapse formation are not well understood. Here we use high-resolution two-photon microscopy in organotypic hippocampal slices from GAD65-GFP mice of both sexes to examine the signaling pathways induced by the postsynaptic signaling molecule Semaphorin4D (Sema4D) during inhibitory synapse formation. By monitoring changes in individual GFP-labeled presynaptic boutons, we found that the primary action of Sema4D is to induce stabilization of presynaptic boutons within tens of minutes. Stabilized boutons rapidly recruited synaptic vesicles, followed by accumulation of postsynaptic gephyrin and were functional after 24 h, as determined by electrophysiology and immunohistochemistry. Inhibitory boutons are only sensitive to Sema4D at a specific stage during synapse formation and sensitivity to Sema4D is regulated by network activity. We further examined the intracellular signaling cascade triggered by Sema4D and found that bouton stabilization occurs through rapid remodeling of the actin cytoskeleton. This could be mimicked by the actin-depolymerizing drug latrunculin B or by reducing ROCK activity. We discovered that the intracellular signaling cascade requires activation of the receptor tyrosine kinase MET, which is a well known autism risk factor. By using a viral approach to reduce MET levels specifically in inhibitory neurons, we found that their axons are no longer sensitive to Sema4D signaling. Together, our data yield important insights into the molecular pathway underlying activity-dependent Sema4D-induced synapse formation and reveal a novel role for presynaptic MET at inhibitory synapses.SIGNIFICANCE STATEMENT GABAergic synapses provide the main inhibitory control of neuronal activity in the brain. We wanted to unravel the sequence of molecular events that take place when formation of inhibitory synapses is triggered by a specific signaling molecule, Sema4D. We find that this signaling pathway depends on network activity and involves specific remodeling of the intracellular actin cytoskeleton. We also reveal a previously unknown role for MET at inhibitory synapses. Our study provides novel insights into the dynamic process of inhibitory synapse formation. As defects in GABAergic synapses have been implied in many brain disorders, and mutations in MET are strong risk factors for autism, our findings urge for a further investigation of the role of MET at inhibitory synapses.
Collapse
|
47
|
Held A, Major P, Sahin A, Reenan RA, Lipscombe D, Wharton KA. Circuit Dysfunction in SOD1-ALS Model First Detected in Sensory Feedback Prior to Motor Neuron Degeneration Is Alleviated by BMP Signaling. J Neurosci 2019; 39:2347-2364. [PMID: 30659087 PMCID: PMC6433758 DOI: 10.1523/jneurosci.1771-18.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/24/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease for which the origin and underlying cellular defects are not fully understood. Although motor neuron degeneration is the signature feature of ALS, it is not clear whether motor neurons or other cells of the motor circuit are the site of disease initiation. To better understand the contribution of multiple cell types in ALS, we made use of a Drosophila Sod1G85R knock-in model, in which all cells harbor the disease allele. End-stage dSod1G85R animals of both sexes exhibit severe motor deficits with clear degeneration of motor neurons. Interestingly, earlier in dSod1G85R larvae, motor function is also compromised, but their motor neurons exhibit only subtle morphological and electrophysiological changes that are unlikely to cause the observed decrease in locomotion. We analyzed the intact motor circuit and identified a defect in sensory feedback that likely accounts for the altered motor activity of dSod1G85R We found cell-autonomous activation of bone morphogenetic protein signaling in proprioceptor sensory neurons which are critical for the relay of the contractile status of muscles back to the central nerve cord, completely rescues early-stage motor defects and partially rescue late-stage motor function to extend lifespan. Identification of a defect in sensory feedback as a potential initiating event in ALS motor dysfunction, coupled with the ability of modified proprioceptors to alleviate such motor deficits, underscores the critical role that nonmotor neurons play in disease progression and highlights their potential as a site to identify early-stage ALS biomarkers and for therapeutic intervention.SIGNIFICANCE STATEMENT At diagnosis, many cellular processes are already disrupted in the amyotrophic lateral sclerosis (ALS) patient. Identifying the initiating cellular events is critical for achieving an earlier diagnosis to slow or prevent disease progression. Our findings indicate that neurons relaying sensory information underlie early stage motor deficits in a Drosophila knock-in model of ALS that best replicates gene dosage in familial ALS (fALS). Importantly, studies on intact motor circuits revealed defects in sensory feedback before evidence of motor neuron degeneration. These findings strengthen our understanding of how neural circuit dysfunctions lead to neurodegeneration and, coupled with our demonstration that the activation of bone morphogenetic protein signaling in proprioceptors alleviates both early and late motor dysfunction, underscores the importance of considering nonmotor neurons as therapeutic targets.
Collapse
Affiliation(s)
- Aaron Held
- Department of Molecular Biology, Cell Biology and Biochemistry
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Paxton Major
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Asli Sahin
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Robert A Reenan
- Department of Molecular Biology, Cell Biology and Biochemistry
| | - Diane Lipscombe
- Department of Neuroscience, and
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Kristi A Wharton
- Department of Molecular Biology, Cell Biology and Biochemistry,
- The Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
48
|
DePew AT, Aimino MA, Mosca TJ. The Tenets of Teneurin: Conserved Mechanisms Regulate Diverse Developmental Processes in the Drosophila Nervous System. Front Neurosci 2019; 13:27. [PMID: 30760977 PMCID: PMC6363694 DOI: 10.3389/fnins.2019.00027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/11/2019] [Indexed: 01/26/2023] Open
Abstract
To successfully integrate a neuron into a circuit, a myriad of developmental events must occur correctly and in the correct order. Neurons must be born and grow out toward a destination, responding to guidance cues to direct their path. Once arrived, each neuron must segregate to the correct sub-region before sorting through a milieu of incorrect partners to identify the correct partner with which they can connect. Finally, the neuron must make a synaptic connection with their correct partner; a connection that needs to be broadly maintained throughout the life of the animal while remaining responsive to modes of plasticity and pruning. Though many intricate molecular mechanisms have been discovered to regulate each step, recent work showed that a single family of proteins, the Teneurins, regulates a host of these developmental steps in Drosophila – an example of biological adaptive reuse. Teneurins first influence axon guidance during early development. Once neurons arrive in their target regions, Teneurins enable partner matching and synapse formation in both the central and peripheral nervous systems. Despite these diverse processes and systems, the Teneurins use conserved mechanisms to achieve these goals, as defined by three tenets: (1) transsynaptic interactions with each other, (2) membrane stabilization via an interaction with and regulation of the cytoskeleton, and (3) a role for presynaptic Ten-a in regulating synaptic function. These processes are further distinguished by (1) the nature of the transsynaptic interaction – homophilic interactions (between the same Teneurins) to engage partner matching and heterophilic interactions (between different Teneurins) to enable synaptic connectivity and the proper apposition of pre- and postsynaptic sites and (2) the location of cytoskeletal regulation (presynaptic cytoskeletal regulation in the CNS and postsynaptic regulation of the cytoskeleton at the NMJ). Thus, both the roles and the mechanisms governing them are conserved across processes and synapses. Here, we will highlight the contributions of Drosophila synaptic biology to our understanding of the Teneurins, discuss the mechanistic conservation that allows the Teneurins to achieve common neurodevelopmental goals, and present new data in support of these points. Finally, we will posit the next steps for understanding how this remarkably versatile family of proteins functions to control multiple distinct events in the creation of a nervous system.
Collapse
Affiliation(s)
- Alison T DePew
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Michael A Aimino
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Timothy J Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
49
|
Dason JS, Allen AM, Vasquez OE, Sokolowski MB. Distinct functions of a cGMP-dependent protein kinase in nerve terminal growth and synaptic vesicle cycling. J Cell Sci 2019; 132:jcs.227165. [DOI: 10.1242/jcs.227165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/26/2019] [Indexed: 01/20/2023] Open
Abstract
Sustained neurotransmission requires the tight coupling of synaptic vesicle (SV) exocytosis and endocytosis. The mechanisms underlying this coupling are poorly understood. We tested the hypothesis that a cGMP-dependent protein kinase (PKG), encoded by the foraging (for) gene in Drosophila melanogaster, is critical for this process using a for null mutant, genomic rescues, and tissue specific rescues. We uncoupled FOR's exocytic and endocytic functions in neurotransmission using a temperature-sensitive shibire mutant in conjunction with fluorescein-assisted light inactivation of FOR. We discovered a dual role for presynaptic FOR, where FOR inhibits SV exocytosis during low frequency stimulation by negatively regulating presynaptic Ca2+ levels and maintains neurotransmission during high frequency stimulation by facilitating SV endocytosis. Additionally, glial FOR negatively regulated nerve terminal growth through TGF-β signaling and this developmental effect was independent from FOR's effects on neurotransmission. Overall, FOR plays a critical role in coupling SV exocytosis and endocytosis, thereby balancing these two components to maintain sustained neurotransmission.
Collapse
Affiliation(s)
- Jeffrey S. Dason
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, N9B 3P4, Canada
| | - Aaron M. Allen
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Present Address: Centre for Neural Circuits and Behaviour, University of Oxford, OX1 3SR Oxford, UK
| | - Oscar E. Vasquez
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
| | - Marla B. Sokolowski
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, M5S 3B2, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
50
|
Oswald MC, Brooks PS, Zwart MF, Mukherjee A, West RJ, Giachello CN, Morarach K, Baines RA, Sweeney ST, Landgraf M. Reactive oxygen species regulate activity-dependent neuronal plasticity in Drosophila. eLife 2018; 7:39393. [PMID: 30540251 PMCID: PMC6307858 DOI: 10.7554/elife.39393] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/12/2018] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) have been extensively studied as damaging agents associated with ageing and neurodegenerative conditions. Their role in the nervous system under non-pathological conditions has remained poorly understood. Working with the Drosophila larval locomotor network, we show that in neurons ROS act as obligate signals required for neuronal activity-dependent structural plasticity, of both pre- and postsynaptic terminals. ROS signaling is also necessary for maintaining evoked synaptic transmission at the neuromuscular junction, and for activity-regulated homeostatic adjustment of motor network output, as measured by larval crawling behavior. We identified the highly conserved Parkinson’s disease-linked protein DJ-1β as a redox sensor in neurons where it regulates structural plasticity, in part via modulation of the PTEN-PI3Kinase pathway. This study provides a new conceptual framework of neuronal ROS as second messengers required for neuronal plasticity and for network tuning, whose dysregulation in the ageing brain and under neurodegenerative conditions may contribute to synaptic dysfunction.
Collapse
Affiliation(s)
- Matthew Cw Oswald
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Paul S Brooks
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | | | - Amrita Mukherjee
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Ryan Jh West
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Department of Biology, University of York, York, United Kingdom
| | - Carlo Ng Giachello
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Khomgrit Morarach
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Richard A Baines
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Sean T Sweeney
- Department of Biology, University of York, York, United Kingdom
| | - Matthias Landgraf
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|