1
|
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
|
2
|
Chen X, Perry S, Fan Z, Wang B, Loxterkamp E, Wang S, Hu J, Dickman D, Han C. Tissue-specific knockout in the Drosophila neuromuscular system reveals ESCRT's role in formation of synapse-derived extracellular vesicles. PLoS Genet 2024; 20:e1011438. [PMID: 39388480 PMCID: PMC11495600 DOI: 10.1371/journal.pgen.1011438] [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: 03/13/2024] [Revised: 10/22/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Tissue-specific gene knockout by CRISPR/Cas9 is a powerful approach for characterizing gene functions during development. However, this approach has not been successfully applied to most Drosophila tissues, including the Drosophila neuromuscular junction (NMJ). To expand tissue-specific CRISPR to this powerful model system, here we present a CRISPR-mediated tissue-restricted mutagenesis (CRISPR-TRiM) toolkit for knocking out genes in motoneurons, muscles, and glial cells. We validated the efficacy of CRISPR-TRiM by knocking out multiple genes in each tissue, demonstrated its orthogonal use with the Gal4/UAS binary expression system, and showed simultaneous knockout of multiple redundant genes. We used CRISPR-TRiM to discover an essential role for SNARE components in NMJ maintenance. Furthermore, we demonstrate that the canonical ESCRT pathway suppresses NMJ bouton growth by downregulating retrograde Gbb signaling. Lastly, we found that axon termini of motoneurons rely on ESCRT-mediated intra-axonal membrane trafficking to release extracellular vesicles at the NMJ. Thus, we have successfully developed an NMJ CRISPR mutagenesis approach which we used to reveal genes important for NMJ structural plasticity.
Collapse
Affiliation(s)
- Xinchen Chen
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Ziwei Fan
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Bei Wang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Elizabeth Loxterkamp
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Shuran Wang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jiayi Hu
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, California, United States of America
| | - Chun Han
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
3
|
Banerjee S, Vernon S, Ruchti E, Limoni G, Jiao W, Asadzadeh J, Van Campenhoudt M, McCabe BD. Trio preserves motor synapses and prolongs motor ability during aging. Cell Rep 2024; 43:114256. [PMID: 38795343 DOI: 10.1016/j.celrep.2024.114256] [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: 05/11/2022] [Revised: 01/24/2024] [Accepted: 05/05/2024] [Indexed: 05/27/2024] Open
Abstract
The decline of motor ability is a hallmark feature of aging and is accompanied by degeneration of motor synaptic terminals. Consistent with this, Drosophila motor synapses undergo characteristic age-dependent structural fragmentation co-incident with diminishing motor ability. Here, we show that motor synapse levels of Trio, an evolutionarily conserved guanine nucleotide exchange factor (GEF), decline with age. We demonstrate that increasing Trio expression in adult Drosophila can abrogate age-dependent synaptic structural fragmentation, postpone the decline of motor ability, and maintain the capacity of motor synapses to sustain high-intensity neurotransmitter release. This preservative activity is conserved in transgenic human Trio, requires Trio Rac GEF function, and can also ameliorate synapse degeneration induced by depletion of miniature neurotransmission. Our results support a paradigm where the structural dissolution of motor synapses precedes and promotes motor behavioral diminishment and where intervening in this process can postpone the decline of motor function during aging.
Collapse
Affiliation(s)
- Soumya Banerjee
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Samuel Vernon
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Marine Van Campenhoudt
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, VD 1015 Lausanne, Switzerland.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Mina E, Wyart E, Sartori R, Angelino E, Zaggia I, Rausch V, Maldotti M, Pagani A, Hsu MY, Friziero A, Sperti C, Menga A, Graziani A, Hirsch E, Oliviero S, Sandri M, Conti L, Kautz L, Silvestri L, Porporato PE. FK506 bypasses the effect of erythroferrone in cancer cachexia skeletal muscle atrophy. Cell Rep Med 2023; 4:101306. [PMID: 38052214 PMCID: PMC10772350 DOI: 10.1016/j.xcrm.2023.101306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/29/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023]
Abstract
Skeletal muscle atrophy is a hallmark of cachexia, a wasting condition typical of chronic pathologies, that still represents an unmet medical need. Bone morphogenetic protein (BMP)-Smad1/5/8 signaling alterations are emerging drivers of muscle catabolism, hence, characterizing these perturbations is pivotal to develop therapeutic approaches. We identified two promoters of "BMP resistance" in cancer cachexia, specifically the BMP scavenger erythroferrone (ERFE) and the intracellular inhibitor FKBP12. ERFE is upregulated in cachectic cancer patients' muscle biopsies and in murine cachexia models, where its expression is driven by STAT3. Moreover, the knock down of Erfe or Fkbp12 reduces muscle wasting in cachectic mice. To bypass the BMP resistance mediated by ERFE and release the brake on the signaling, we targeted FKBP12 with low-dose FK506. FK506 restores BMP-Smad1/5/8 signaling, rescuing myotube atrophy by inducing protein synthesis. In cachectic tumor-bearing mice, FK506 prevents muscle and body weight loss and protects from neuromuscular junction alteration, suggesting therapeutic potential for targeting the ERFE-FKBP12 axis.
Collapse
Affiliation(s)
- Erica Mina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Roberta Sartori
- Department of Biomedical Sciences, University of Padova, Padova, Italy; VIMM: Veneto Institute of Molecular Medicine, Padova, Italy
| | - Elia Angelino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Ivan Zaggia
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Valentina Rausch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Mara Maldotti
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Italian Institute for Genomic Medicine (IIGM), Sp142 Km 3.95, 10060 Candiolo, Torino, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Myriam Y Hsu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Division of Cell Fate Dynamics and Therapeutics, Department of Biosystems Science, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto, Japan
| | - Alberto Friziero
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; General Surgery 1, Padova University Hospital, Padova, Italy
| | - Cosimo Sperti
- General Surgery 2, Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, Padova University Hospital, Padova, Italy
| | - Alessio Menga
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Andrea Graziani
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Italian Institute for Genomic Medicine (IIGM), Sp142 Km 3.95, 10060 Candiolo, Torino, Italy
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy; VIMM: Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Léon Kautz
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, University Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Laura Silvestri
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita Salute San Raffaele University, Milan, Italy
| | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy.
| |
Collapse
|
7
|
Bertin F, Jara-Wilde J, Auer B, Köhler-Solís A, González-Silva C, Thomas U, Sierralta J. Drosophila Atlastin regulates synaptic vesicle mobilization independent of bone morphogenetic protein signaling. Biol Res 2023; 56:49. [PMID: 37710314 PMCID: PMC10503011 DOI: 10.1186/s40659-023-00462-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) contacts endosomes in all parts of a motor neuron, including the axon and presynaptic terminal, to move structural proteins, proteins that send signals, and lipids over long distances. Atlastin (Atl), a large GTPase, is required for membrane fusion and the structural dynamics of the ER tubules. Atl mutations are the second most common cause of Hereditary Spastic Paraplegia (HSP), which causes spasticity in both sexes' lower extremities. Through an unknown mechanism, Atl mutations stimulate the BMP (bone morphogenetic protein) pathway in vertebrates and Drosophila. Synaptic defects are caused by atl mutations, which affect the abundance and distribution of synaptic vesicles (SV) in the bouton. We hypothesize that BMP signaling, does not cause Atl-dependent SV abnormalities in Drosophila. RESULTS We show that atl knockdown in motor neurons (Atl-KD) increases synaptic and satellite boutons in the same way that constitutively activating the BMP-receptor Tkv (thick veins) (Tkv-CA) increases the bouton number. The SV proteins Cysteine string protein (CSP) and glutamate vesicular transporter are reduced in Atl-KD and Tkv-CA larvae. Reducing the activity of the BMP receptor Wishful thinking (wit) can rescue both phenotypes. Unlike Tkv-CA larvae, Atl-KD larvae display altered activity-dependent distributions of CSP staining. Furthermore, Atl-KD larvae display an increased FM 1-43 unload than Control and Tkv-CA larvae. As decreasing wit function does not reduce the phenotype, our hypothesis that BMP signaling is not involved is supported. We also found that Rab11/CSP colocalization increased in Atl-KD larvae, which supports the concept that late recycling endosomes regulate SV movements. CONCLUSIONS Our findings reveal that Atl modulates neurotransmitter release in motor neurons via SV distribution independently of BMP signaling, which could explain the observed SV accumulation and synaptic dysfunction. Our data suggest that Atl is involved in membrane traffic as well as formation and/or recycling of the late endosome.
Collapse
Affiliation(s)
- Francisca Bertin
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jorge Jara-Wilde
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Computational Sciences, Faculty of Physical and Mathematical Sciences, Universidad de Chile, Santiago, Chile
| | - Benedikt Auer
- Laboratory of Neuronal and Synaptic Signals, Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Andrés Köhler-Solís
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Carolina González-Silva
- Biomedical Neuroscience Institute (BNI), Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ulrich Thomas
- Functional Genetics of the Synapse, Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Jimena Sierralta
- Biomedical Neuroscience Institute (BNI), Santiago, Chile.
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Terauchi A, Yee P, Johnson-Venkatesh EM, Seiglie MP, Kim L, Pitino JC, Kritzer E, Zhang Q, Zhou J, Li Y, Ginty DD, Lee WCA, Umemori H. The projection-specific signals that establish functionally segregated dopaminergic synapses. Cell 2023; 186:3845-3861.e24. [PMID: 37591240 PMCID: PMC10540635 DOI: 10.1016/j.cell.2023.07.023] [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: 08/10/2022] [Revised: 05/28/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023]
Abstract
Dopaminergic projections regulate various brain functions and are implicated in many neuropsychiatric disorders. There are two anatomically and functionally distinct dopaminergic projections connecting the midbrain to striatum: nigrostriatal, which controls movement, and mesolimbic, which regulates motivation. However, how these discrete dopaminergic synaptic connections are established is unknown. Through an unbiased search, we identify that two groups of antagonistic TGF-β family members, bone morphogenetic protein (BMP)6/BMP2 and transforming growth factor (TGF)-β2, regulate dopaminergic synapse development of nigrostriatal and mesolimbic neurons, respectively. Projection-preferential expression of their receptors contributes to specific synapse development. Downstream, Smad1 and Smad2 are specifically activated and required for dopaminergic synapse development and function in nigrostriatal vs. mesolimbic projections. Remarkably, Smad1 mutant mice show motor defects, whereas Smad2 mutant mice show lack of motivation. These results uncover the molecular logic underlying the proper establishment of functionally segregated dopaminergic synapses and may provide strategies to treat relevant, projection-specific disease symptoms by targeting specific BMPs/TGF-β and/or Smads.
Collapse
Affiliation(s)
- Akiko Terauchi
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patricia Yee
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erin M Johnson-Venkatesh
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mariel P Seiglie
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa Kim
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Julia C Pitino
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eli Kritzer
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qiyu Zhang
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jie Zhou
- Department of Computer Science, Northern Illinois University, DeKalb, IL 60115, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - David D Ginty
- Department of Neurobiology and Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Wei-Chung A Lee
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
9
|
Zhou X, Gan G, Sun Y, Ou M, Geng J, Wang J, Yang X, Huang S, Jia D, Xie W, He H. GTPase-activating protein TBC1D5 coordinates with retromer to constrain synaptic growth by inhibiting BMP signaling. J Genet Genomics 2023; 50:163-177. [PMID: 36473687 DOI: 10.1016/j.jgg.2022.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Formation and plasticity of neural circuits rely on precise regulation of synaptic growth. At Drosophila neuromuscular junction (NMJ), Bone Morphogenetic Protein (BMP) signaling is critical for many aspects of synapse formation and function. The evolutionarily conserved retromer complex and its associated GTPase-activating protein TBC1D5 are critical regulators of membrane trafficking and cellular signaling. However, their functions in regulating the formation of NMJ are less understood. Here, we report that TBC1D5 is required for inhibition of synaptic growth, and loss of TBC1D5 leads to abnormal presynaptic terminal development, including excessive satellite boutons and branch formation. Ultrastructure analysis reveals that the size of synaptic vesicles and the density of subsynaptic reticulum are increased in TBC1D5 mutant boutons. Disruption of interactions of TBC1D5 with Rab7 and retromer phenocopies the loss of TBC1D5. Unexpectedly, we find that TBC1D5 is functionally linked to Rab6, in addition to Rab7, to regulate synaptic growth. Mechanistically, we show that loss of TBC1D5 leads to upregulated BMP signaling by increasing the protein level of BMP type II receptor Wishful Thinking (Wit) at NMJ. Overall, our data establish that TBC1D5 in coordination with retromer constrains synaptic growth by regulating Rab7 activity, which negatively regulates BMP signaling through inhibiting Wit level.
Collapse
Affiliation(s)
- Xiu Zhou
- State Key Laboratory of Biotherapy, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Guangming Gan
- The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210096, China; The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yichen Sun
- The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210096, China
| | - Mengzhu Ou
- The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210096, China
| | - Junhua Geng
- The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210096, China
| | - Jing Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xi Yang
- State Key Laboratory of Biotherapy, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shu Huang
- State Key Laboratory of Biotherapy, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease (MOE), School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Haihuai He
- State Key Laboratory of Biotherapy, Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
10
|
Vuilleumier R, Miao M, Medina-Giro S, Ell CM, Flibotte S, Lian T, Kauwe G, Collins A, Ly S, Pyrowolakis G, Haghighi A, Allan D. Dichotomous cis-regulatory motifs mediate the maturation of the neuromuscular junction by retrograde BMP signaling. Nucleic Acids Res 2022; 50:9748-9764. [PMID: 36029115 PMCID: PMC9508838 DOI: 10.1093/nar/gkac730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/20/2022] [Accepted: 08/19/2022] [Indexed: 11/26/2022] Open
Abstract
Retrograde bone morphogenetic protein (BMP) signaling at the Drosophila neuromuscular junction (NMJ) has served as a paradigm to study TGF-β-dependent synaptic function and maturation. Yet, how retrograde BMP signaling transcriptionally regulates these functions remains unresolved. Here, we uncover a gene network, enriched for neurotransmission-related genes, that is controlled by retrograde BMP signaling in motor neurons through two Smad-binding cis-regulatory motifs, the BMP-activating (BMP-AE) and silencer (BMP-SE) elements. Unpredictably, both motifs mediate direct gene activation, with no involvement of the BMP derepression pathway regulators Schnurri and Brinker. Genome editing of candidate BMP-SE and BMP-AE within the locus of the active zone gene bruchpilot, and a novel Ly6 gene witty, demonstrated the role of these motifs in upregulating genes required for the maturation of pre- and post-synaptic NMJ compartments. Our findings uncover how Smad-dependent transcriptional mechanisms specific to motor neurons directly orchestrate a gene network required for synaptic maturation by retrograde BMP signaling.
Collapse
Affiliation(s)
- Robin Vuilleumier
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Mo Miao
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Sonia Medina-Giro
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Clara-Maria Ell
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, 79104, Germany
- CIBSS - Centre for Integrative Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Hilde Mangold Haus, Habsburgerstrasse 49, University of Freiburg, Freiburg, 79104, Germany
| | - Stephane Flibotte
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Annie Collins
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Sophia Ly
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - George Pyrowolakis
- CIBSS - Centre for Integrative Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Hilde Mangold Haus, Habsburgerstrasse 49, University of Freiburg, Freiburg, 79104, Germany
| | | | - Douglas W Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| |
Collapse
|
11
|
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
|
12
|
Ho CH, Paolantoni C, Bawankar P, Tang Z, Brown S, Roignant J, Treisman JE. An exon junction complex-independent function of Barentsz in neuromuscular synapse growth. EMBO Rep 2022; 23:e53231. [PMID: 34726300 PMCID: PMC8728599 DOI: 10.15252/embr.202153231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
The exon junction complex controls the translation, degradation, and localization of spliced mRNAs, and three of its core subunits also play a role in splicing. Here, we show that a fourth subunit, Barentsz, has distinct functions within and separate from the exon junction complex in Drosophila neuromuscular development. The distribution of mitochondria in larval muscles requires Barentsz as well as other exon junction complex subunits and is not rescued by a Barentsz transgene in which residues required for binding to the core subunit eIF4AIII are mutated. In contrast, interactions with the exon junction complex are not required for Barentsz to promote the growth of neuromuscular synapses. We find that the Activin ligand Dawdle shows reduced expression in barentsz mutants and acts downstream of Barentsz to control synapse growth. Both barentsz and dawdle are required in motor neurons, muscles, and glia for normal synapse growth, and exogenous Dawdle can rescue synapse growth in the absence of barentsz. These results identify a biological function for Barentsz that is independent of the exon junction complex.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| | - Chiara Paolantoni
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
| | - Praveen Bawankar
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Zuojian Tang
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
Computational Biology at Ridgefield US, Global Computational Biology and Digital ScienceBoehringer IngelheimRidgefieldCTUSA
| | - Stuart Brown
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
ExxonMobil Corporate Strategic ResearchAnnandaleNJUSA
| | - Jean‐Yves Roignant
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Jessica E Treisman
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| |
Collapse
|
13
|
Cornejo F, Cortés BI, Findlay GM, Cancino GI. LAR Receptor Tyrosine Phosphatase Family in Healthy and Diseased Brain. Front Cell Dev Biol 2021; 9:659951. [PMID: 34966732 PMCID: PMC8711739 DOI: 10.3389/fcell.2021.659951] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
Protein phosphatases are major regulators of signal transduction and they are involved in key cellular mechanisms such as proliferation, differentiation, and cell survival. Here we focus on one class of protein phosphatases, the type IIA Receptor-type Protein Tyrosine Phosphatases (RPTPs), or LAR-RPTP subfamily. In the last decade, LAR-RPTPs have been demonstrated to have great importance in neurobiology, from neurodevelopment to brain disorders. In vertebrates, the LAR-RPTP subfamily is composed of three members: PTPRF (LAR), PTPRD (PTPδ) and PTPRS (PTPσ), and all participate in several brain functions. In this review we describe the structure and proteolytic processing of the LAR-RPTP subfamily, their alternative splicing and enzymatic regulation. Also, we review the role of the LAR-RPTP subfamily in neural function such as dendrite and axon growth and guidance, synapse formation and differentiation, their participation in synaptic activity, and in brain development, discussing controversial findings and commenting on the most recent studies in the field. Finally, we discuss the clinical outcomes of LAR-RPTP mutations, which are associated with several brain disorders.
Collapse
Affiliation(s)
- Francisca Cornejo
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Bastián I Cortés
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Greg M Findlay
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Gonzalo I Cancino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| |
Collapse
|
14
|
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
|
15
|
Sartori R, Hagg A, Zampieri S, Armani A, Winbanks CE, Viana LR, Haidar M, Watt KI, Qian H, Pezzini C, Zanganeh P, Turner BJ, Larsson A, Zanchettin G, Pierobon ES, Moletta L, Valmasoni M, Ponzoni A, Attar S, Da Dalt G, Sperti C, Kustermann M, Thomson RE, Larsson L, Loveland KL, Costelli P, Megighian A, Merigliano S, Penna F, Gregorevic P, Sandri M. Perturbed BMP signaling and denervation promote muscle wasting in cancer cachexia. Sci Transl Med 2021; 13:eaay9592. [PMID: 34349036 DOI: 10.1126/scitranslmed.aay9592] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/18/2021] [Indexed: 02/05/2023]
Abstract
Most patients with advanced solid cancers exhibit features of cachexia, a debilitating syndrome characterized by progressive loss of skeletal muscle mass and strength. Because the underlying mechanisms of this multifactorial syndrome are incompletely defined, effective therapeutics have yet to be developed. Here, we show that diminished bone morphogenetic protein (BMP) signaling is observed early in the onset of skeletal muscle wasting associated with cancer cachexia in mouse models and in patients with cancer. Cancer-mediated factors including Activin A and IL-6 trigger the expression of the BMP inhibitor Noggin in muscle, which blocks the actions of BMPs on muscle fibers and motor nerves, subsequently causing disruption of the neuromuscular junction (NMJ), denervation, and muscle wasting. Increasing BMP signaling in the muscles of tumor-bearing mice by gene delivery or pharmacological means can prevent muscle wasting and preserve measures of NMJ function. The data identify perturbed BMP signaling and denervation of muscle fibers as important pathogenic mechanisms of muscle wasting associated with tumor growth. Collectively, these findings present interventions that promote BMP-mediated signaling as an attractive strategy to counteract the loss of functional musculature in patients with cancer.
Collapse
Affiliation(s)
- Roberta Sartori
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Adam Hagg
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Sandra Zampieri
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
- Myology Center, University of Padova, 35122 Padua, Italy
| | - Andrea Armani
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | | | - Laís R Viana
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, São Paulo 13083-97, Brazil
| | - Mouna Haidar
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Kevin I Watt
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Hongwei Qian
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Camilla Pezzini
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Pardis Zanganeh
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Anna Larsson
- Theme Cancer, Karolinska University Hospital, Solna 171 76, Sweden
| | - Gianpietro Zanchettin
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Elisa S Pierobon
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Lucia Moletta
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Michele Valmasoni
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Alberto Ponzoni
- Department of Radiology, Padova General Hospital, 35121 Padova, Italy
| | - Shady Attar
- Department of Medicine, University Hospital of Padova, 35121 Padova, Italy
| | - Gianfranco Da Dalt
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Monika Kustermann
- Center for Anatomy and Cell Biology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rachel E Thomson
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kate L Loveland
- Centre for Reproductive Health. Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, and Anatomy and Developmental Biology, Monash University, VIC 3800, Australia
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Stefano Merigliano
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia.
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Marco Sandri
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy.
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Myology Center, University of Padova, 35122 Padua, Italy
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
16
|
Spierer AN, Mossman JA, Smith SP, Crawford L, Ramachandran S, Rand DM. Natural variation in the regulation of neurodevelopmental genes modifies flight performance in Drosophila. PLoS Genet 2021; 17:e1008887. [PMID: 33735180 PMCID: PMC7971549 DOI: 10.1371/journal.pgen.1008887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/26/2021] [Indexed: 12/28/2022] Open
Abstract
The winged insects of the order Diptera are colloquially named for their most recognizable phenotype: flight. These insects rely on flight for a number of important life history traits, such as dispersal, foraging, and courtship. Despite the importance of flight, relatively little is known about the genetic architecture of flight performance. Accordingly, we sought to uncover the genetic modifiers of flight using a measure of flies’ reaction and response to an abrupt drop in a vertical flight column. We conducted a genome wide association study (GWAS) using 197 of the Drosophila Genetic Reference Panel (DGRP) lines, and identified a combination of additive and marginal variants, epistatic interactions, whole genes, and enrichment across interaction networks. Egfr, a highly pleiotropic developmental gene, was among the most significant additive variants identified. We functionally validated 13 of the additive candidate genes’ (Adgf-A/Adgf-A2/CG32181, bru1, CadN, flapper (CG11073), CG15236, flippy (CG9766), CREG, Dscam4, form3, fry, Lasp/CG9692, Pde6, Snoo), and introduce a novel approach to whole gene significance screens: PEGASUS_flies. Additionally, we identified ppk23, an Acid Sensing Ion Channel (ASIC) homolog, as an important hub for epistatic interactions. We propose a model that suggests genetic modifiers of wing and muscle morphology, nervous system development and function, BMP signaling, sexually dimorphic neural wiring, and gene regulation are all important for the observed differences flight performance in a natural population. Additionally, these results represent a snapshot of the genetic modifiers affecting drop-response flight performance in Drosophila, with implications for other insects. Insect flight is a widely recognizable phenotype of many winged insects, hence the name: flies. While fruit flies, or Drosophila melanogaster, are a genetically tractable model, flight performance is a highly integrative phenotype, and therefore challenging to identify comprehensively which genetic modifiers contribute to its genetic architecture. Accordingly, we screened 197 Drosophila Genetic Reference Panel lines for their ability to react and respond to an abrupt drop. Using several computational approaches, we identified additive, marginal, and epistatic variants, as well as whole genes and altered sub-networks of gene-gene and protein-protein interaction networks that contribute to variation in flight performance. More generally, we demonstrate the benefits of employing multiple methodologies to elucidate the genetic architecture of complex traits. Many variants and genes mapped to regions of the genome that affect neurodevelopment, wing and muscle development, and regulation of gene expression. We also introduce PEGASUS_flies, a Drosophila-adapted version of the PEGASUS platform first used in human studies, to infer gene-level significance of association based on the gene’s distribution of individual variant P-values. Our results contribute to the debate over the relative importance of individual, additive factors and epistatic, or higher order, interactions, in the mapping of genotype to phenotype.
Collapse
Affiliation(s)
- Adam N Spierer
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
| | - Jim A Mossman
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Samuel Pattillo Smith
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Lorin Crawford
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Microsoft Research New England, Cambridge, Massachusetts, United States of America
| | - Sohini Ramachandran
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - David M Rand
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
17
|
Sidisky JM, Weaver D, Hussain S, Okumus M, Caratenuto R, Babcock D. Mayday sustains trans-synaptic BMP signaling required for synaptic maintenance with age. eLife 2021; 10:e54932. [PMID: 33667157 PMCID: PMC7935490 DOI: 10.7554/elife.54932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/23/2021] [Indexed: 01/12/2023] Open
Abstract
Maintaining synaptic structure and function over time is vital for overall nervous system function and survival. The processes that underly synaptic development are well understood. However, the mechanisms responsible for sustaining synapses throughout the lifespan of an organism are poorly understood. Here, we demonstrate that a previously uncharacterized gene, CG31475, regulates synaptic maintenance in adult Drosophila NMJs. We named CG31475 mayday due to the progressive loss of flight ability and synapse architecture with age. Mayday is functionally homologous to the human protein Cab45, which sorts secretory cargo from the Trans Golgi Network (TGN). We find that Mayday is required to maintain trans-synaptic BMP signaling at adult NMJs in order to sustain proper synaptic structure and function. Finally, we show that mutations in mayday result in the loss of both presynaptic motor neurons as well as postsynaptic muscles, highlighting the importance of maintaining synaptic integrity for cell viability.
Collapse
Affiliation(s)
- Jessica M Sidisky
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Daniel Weaver
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Sarrah Hussain
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Meryem Okumus
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Russell Caratenuto
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Daniel Babcock
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| |
Collapse
|
18
|
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
|
19
|
Berndt AJ, Othonos KM, Lian T, Flibotte S, Miao M, Bhuiyan SA, Cho RY, Fong JS, Hur SA, Pavlidis P, Allan DW. A low affinity cis-regulatory BMP response element restricts target gene activation to subsets of Drosophila neurons. eLife 2020; 9:59650. [PMID: 33124981 PMCID: PMC7669266 DOI: 10.7554/elife.59650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/29/2020] [Indexed: 11/19/2022] Open
Abstract
Retrograde BMP signaling and canonical pMad/Medea-mediated transcription regulate diverse target genes across subsets of Drosophila efferent neurons, to differentiate neuropeptidergic neurons and promote motor neuron terminal maturation. How a common BMP signal regulates diverse target genes across many neuronal subsets remains largely unresolved, although available evidence implicates subset-specific transcription factor codes rather than differences in BMP signaling. Here we examine the cis-regulatory mechanisms restricting BMP-induced FMRFa neuropeptide expression to Tv4-neurons. We find that pMad/Medea bind at an atypical, low affinity motif in the FMRFa enhancer. Converting this motif to high affinity caused ectopic enhancer activity and eliminated Tv4-neuron expression. In silico searches identified additional motif instances functional in other efferent neurons, implicating broader functions for this motif in BMP-dependent enhancer activity. Thus, differential interpretation of a common BMP signal, conferred by low affinity pMad/Medea binding motifs, can contribute to the specification of BMP target genes in efferent neuron subsets.
Collapse
Affiliation(s)
- Anthony Je Berndt
- Department of Food & Fuel for the 21st Century, University of California San Diego, San Diego, United States
| | - Katerina M Othonos
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Stephane Flibotte
- UBC/LSI Bioinformatics Facility, University of British Columbia, Vancouver, Canada
| | - Mo Miao
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | | | - Raymond Y Cho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Justin S Fong
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Seo Am Hur
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Paul Pavlidis
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | - Douglas W Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
20
|
Spinner MA, Pinter K, Drerup CM, Herman TG. A Conserved Role for Vezatin Proteins in Cargo-Specific Regulation of Retrograde Axonal Transport. Genetics 2020; 216:431-445. [PMID: 32788307 PMCID: PMC7536845 DOI: 10.1534/genetics.120.303499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Active transport of organelles within axons is critical for neuronal health. Retrograde axonal transport, in particular, relays neurotrophic signals received by axon terminals to the nucleus and circulates new material among enpassant synapses. A single motor protein complex, cytoplasmic dynein, is responsible for nearly all retrograde transport within axons: its linkage to and transport of diverse cargos is achieved by cargo-specific regulators. Here, we identify Vezatin as a conserved regulator of retrograde axonal transport. Vertebrate Vezatin (Vezt) is required for the maturation and maintenance of cell-cell junctions and has not previously been implicated in axonal transport. However, a related fungal protein, VezA, has been shown to regulate retrograde transport of endosomes in hyphae. In a forward genetic screen, we identified a loss-of-function mutation in the Drosophila vezatin-like (vezl) gene. We here show that vezl loss prevents a subset of endosomes, including signaling endosomes containing activated BMP receptors, from initiating transport out of motor neuron terminal boutons. vezl loss also decreases the transport of endosomes and dense core vesicles, but not mitochondria, within axon shafts. We disrupted vezt in zebrafish and found that vezt loss specifically impairs the retrograde axonal transport of late endosomes, causing their accumulation in axon terminals. Our work establishes a conserved, cargo-specific role for Vezatin proteins in retrograde axonal transport.
Collapse
Affiliation(s)
- Michael A Spinner
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403
| | - Katherine Pinter
- Unit on Neuronal Cell Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Catherine M Drerup
- Unit on Neuronal Cell Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Tory G Herman
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403
| |
Collapse
|
21
|
Aponte-Santiago NA, Ormerod KG, Akbergenova Y, Littleton JT. Synaptic Plasticity Induced by Differential Manipulation of Tonic and Phasic Motoneurons in Drosophila. J Neurosci 2020; 40:6270-6288. [PMID: 32631939 PMCID: PMC7424871 DOI: 10.1523/jneurosci.0925-20.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 12/12/2022] Open
Abstract
Structural and functional plasticity induced by neuronal competition is a common feature of developing nervous systems. However, the rules governing how postsynaptic cells differentiate between presynaptic inputs are unclear. In this study, we characterized synaptic interactions following manipulations of tonic Ib or phasic Is glutamatergic motoneurons that coinnervate postsynaptic muscles of male or female Drosophila melanogaster larvae. After identifying drivers for each neuronal subtype, we performed ablation or genetic manipulations to alter neuronal activity and examined the effects on synaptic innervation and function at neuromuscular junctions. Ablation of either Ib or Is resulted in decreased muscle response, with some functional compensation occurring in the Ib input when Is was missing. In contrast, the Is terminal failed to show functional or structural changes following loss of the coinnervating Ib input. Decreasing the activity of the Ib or Is neuron with tetanus toxin light chain resulted in structural changes in muscle innervation. Decreased Ib activity resulted in reduced active zone (AZ) number and decreased postsynaptic subsynaptic reticulum volume, with the emergence of filopodial-like protrusions from synaptic boutons of the Ib input. Decreased Is activity did not induce structural changes at its own synapses, but the coinnervating Ib motoneuron increased the number of synaptic boutons and AZs it formed. These findings indicate that tonic Ib and phasic Is motoneurons respond independently to changes in activity, with either functional or structural alterations in the Ib neuron occurring following ablation or reduced activity of the coinnervating Is input, respectively.SIGNIFICANCE STATEMENT Both invertebrate and vertebrate nervous systems display synaptic plasticity in response to behavioral experiences, indicating that underlying mechanisms emerged early in evolution. How specific neuronal classes innervating the same postsynaptic target display distinct types of plasticity is unclear. Here, we examined whether Drosophila tonic Ib and phasic Is motoneurons display competitive or cooperative interactions during innervation of the same muscle, or compensatory changes when the output of one motoneuron is altered. We established a system to differentially manipulate the motoneurons and examined the effects of cell type-specific changes to one of the inputs. Our findings indicate Ib and Is motoneurons respond differently to activity mismatch or loss of the coinnervating input, with the Ib subclass responding robustly compared with Is motoneurons.
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, Massachusetts 02139
| | - Kiel G Ormerod
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - 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, Massachusetts 02139
| |
Collapse
|
22
|
Rushton E, Kopke DL, Broadie K. Extracellular heparan sulfate proteoglycans and glycan-binding lectins orchestrate trans-synaptic signaling. J Cell Sci 2020; 133:133/15/jcs244186. [PMID: 32788209 DOI: 10.1242/jcs.244186] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The exceedingly narrow synaptic cleft (<20 nm) and adjacent perisynaptic extracellular space contain an astonishing array of secreted and membrane-anchored glycoproteins. A number of these extracellular molecules regulate intercellular trans-synaptic signaling by binding to ligands, acting as co-receptors or modulating ligand-receptor interactions. Recent work has greatly expanded our understanding of extracellular proteoglycan and glycan-binding lectin families as key regulators of intercellular signaling at the synapse. These secreted proteins act to regulate the compartmentalization of glycoprotein ligands and receptors, crosslink dynamic extracellular and cell surface lattices, modulate both exocytosis and endocytosis vesicle cycling, and control postsynaptic receptor trafficking. Here, we focus closely on the Drosophila glutamatergic neuromuscular junction (NMJ) as a model synapse for understanding extracellular roles of the many heparan sulfate proteoglycan (HSPG) and lectin proteins that help determine synaptic architecture and neurotransmission strength. We particularly concentrate on the roles of extracellular HSPGs and lectins in controlling trans-synaptic signaling, especially that mediated by the Wnt and BMP pathways. These signaling mechanisms are causally linked to a wide spectrum of neurological disease states that impair coordinated movement and cognitive functions.
Collapse
Affiliation(s)
- Emma Rushton
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Danielle L Kopke
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
23
|
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: 34] [Impact Index Per Article: 6.8] [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
|
24
|
Vickers E, Osypenko D, Clark C, Okur Z, Scheiffele P, Schneggenburger R. LTP of inhibition at PV interneuron output synapses requires developmental BMP signaling. Sci Rep 2020; 10:10047. [PMID: 32572071 PMCID: PMC7308402 DOI: 10.1038/s41598-020-66862-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/27/2020] [Indexed: 11/09/2022] Open
Abstract
Parvalbumin (PV)-expressing interneurons (PV-INs) mediate well-timed inhibition of cortical principal neurons, and plasticity of these interneurons is involved in map remodeling of primary sensory cortices during critical periods of development. To assess whether bone morphogenetic protein (BMP) signaling contributes to the developmental acquisition of the synapse- and plasticity properties of PV-INs, we investigated conditional/conventional double KO mice of BMP-receptor 1a (BMPR1a; targeted to PV-INs) and 1b (BMPR1a/1b (c)DKO mice). We report that spike-timing dependent LTP at the synapse between PV-INs and principal neurons of layer 4 in the auditory cortex was absent, concomitant with a decreased paired-pulse ratio (PPR). On the other hand, baseline synaptic transmission at this connection, and action potential (AP) firing rates of PV-INs were unchanged. To explore possible gene expression targets of BMP signaling, we measured the mRNA levels of the BDNF receptor TrkB and of P/Q-type Ca2+ channel α-subunits, but did not detect expression changes of the corresponding genes in PV-INs of BMPR1a/1b (c)DKO mice. Our study suggests that BMP-signaling in PV-INs during and shortly after the critical period is necessary for the expression of LTP at PV-IN output synapses, involving gene expression programs that need to be addressed in future work.
Collapse
Affiliation(s)
- Evan Vickers
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
- Institute of Neuroscience, University of Oregon, Eugene, OR, 97403, USA
| | - Denys Osypenko
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Christopher Clark
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
- Institute for Regenerative Medicine, University of Zürich, 8952, Schlieren, Switzerland
| | - Zeynep Okur
- Biozentrum, University of Basel, 4056, Basel, Switzerland
| | | | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland.
| |
Collapse
|
25
|
Katrancha SM, Shaw JE, Zhao AY, Myers SA, Cocco AR, Jeng AT, Zhu M, Pittenger C, Greer CA, Carr SA, Xiao X, Koleske AJ. Trio Haploinsufficiency Causes Neurodevelopmental Disease-Associated Deficits. Cell Rep 2020; 26:2805-2817.e9. [PMID: 30840899 PMCID: PMC6436967 DOI: 10.1016/j.celrep.2019.02.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/22/2018] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Heterozygous coding mutations in TRIO are associated with neurodevelopmental disorders, including autism, schizophrenia, bipolar disorder, and epilepsy, and impair TRIO's biochemical activities. To model mutant alleles, we ablated one or both Trio alleles from excitatory neurons in the cortex and hippocampus of mice. Trio haploinsufficiency increases anxiety and impairs social preference and motor coordination. Trio loss reduces forebrain size and dendritic arborization but increases dendritic spine densities. Cortical synapses in Trio haploinsufficient mice are small, exhibit pre- and postsynaptic deficits, and cannot undergo long-term potentiation. Similar phenotypes are observed in Trio knockout mice. Overall, Trio haploinsufficiency causes severe disease-relevant deficits in behavior and neuronal structure and function. Interestingly, phosphodiesterase 4A5 (PDE4A5) levels are reduced and protein kinase A (PKA) signaling is increased when TRIO levels are reduced. Elevation of PDE4A5 and drug-based attenuation of PKA signaling rescue Trio haploinsufficiency-related dendritic spine defects, suggesting an avenue for therapeutic intervention for TRIO-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sara Marie Katrancha
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Juliana E Shaw
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Amy Y Zhao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Samuel A Myers
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Amanda T Jeng
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA
| | - Minsheng Zhu
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Christopher Pittenger
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Psychiatry, Yale University, New Haven, CT 06510, USA; Child Study Center, Yale University, New Haven, CT 06510, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA; Department of Neurosurgery, Yale University, New Haven, CT 06510, USA
| | - Steven A Carr
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Xiao
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai 200433, China.
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
26
|
Paskus JD, Herring BE, Roche KW. Kalirin and Trio: RhoGEFs in Synaptic Transmission, Plasticity, and Complex Brain Disorders. Trends Neurosci 2020; 43:505-518. [PMID: 32513570 DOI: 10.1016/j.tins.2020.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/15/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Changes in the actin cytoskeleton are a primary mechanism mediating the morphological and functional plasticity that underlies learning and memory. The synaptic Ras homologous (Rho) guanine nucleotide exchange factors (GEFs) Kalirin and Trio have emerged as central regulators of actin dynamics at the synapse. The increased attention surrounding Kalirin and Trio stems from the growing evidence for their roles in the etiology of a wide range of neurodevelopmental and neurodegenerative disorders. In this Review, we discuss recent findings revealing the unique and diverse functions of these paralog proteins in neurodevelopment, excitatory synaptic transmission, and plasticity. We additionally survey the growing literature implicating these proteins in various neurological disorders.
Collapse
Affiliation(s)
- Jeremiah D Paskus
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Bruce E Herring
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
27
|
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
|
28
|
Maksoud E, Liao EH, Haghighi AP. A Neuron-Glial Trans-Signaling Cascade Mediates LRRK2-Induced Neurodegeneration. Cell Rep 2020; 26:1774-1786.e4. [PMID: 30759389 PMCID: PMC6474846 DOI: 10.1016/j.celrep.2019.01.077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/19/2018] [Accepted: 01/19/2019] [Indexed: 12/19/2022] Open
Abstract
Pathogenic mutations in leucine-rich repeat kinase 2 (LRRK2) induce an age-dependent loss of dopaminergic (DA) neurons. We have identified Furin 1, a pro-protein convertase, as a translational target of LRRK2 in DA neurons. Transgenic knockdown of Furin1 or its substrate the bone morphogenic protein (BMP) ligand glass bottom boat (Gbb) protects against LRRK2-induced loss of DA neurons. LRRK2 enhances the accumulation of phosphorylated Mad (pMad) in the nuclei of glial cells in the vicinity of DA neurons but not in DA neurons. Consistently, exposure to paraquat enhances Furin 1 levels in DA neurons and induces BMP signaling in glia. In support of a neuron-glial signaling model, knocking down BMP pathway members only in glia, but not in neurons, can protect against paraquat toxicity. We propose that a neuron-glial BMP-signaling cascade is critical for mediating age-dependent neurodegeneration in two models of Parkinson's disease, thus opening avenues for future therapeutic interventions.
Collapse
Affiliation(s)
- Elie Maksoud
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Edward H Liao
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | |
Collapse
|
29
|
McNeill EM, Thompson C, Berke B, Chou VT, Rusch J, Duckworth A, DeProto J, Taylor A, Gates J, Gertler F, Keshishian H, Van Vactor D. Drosophila enabled promotes synapse morphogenesis and regulates active zone form and function. Neural Dev 2020; 15:4. [PMID: 32183907 PMCID: PMC7076993 DOI: 10.1186/s13064-020-00141-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 02/25/2020] [Indexed: 11/10/2022] Open
Abstract
Background Recent studies of synapse form and function highlight the importance of the actin cytoskeleton in regulating multiple aspects of morphogenesis, neurotransmission, and neural plasticity. The conserved actin-associated protein Enabled (Ena) is known to regulate development of the Drosophila larval neuromuscular junction through a postsynaptic mechanism. However, the functions and regulation of Ena within the presynaptic terminal has not been determined. Methods Here, we use a conditional genetic approach to address a presynaptic role for Ena on presynaptic morphology and ultrastructure, and also examine the pathway in which Ena functions through epistasis experiments. Results We find that Ena is required to promote the morphogenesis of presynaptic boutons and branches, in contrast to its inhibitory role in muscle. Moreover, while postsynaptic Ena is regulated by microRNA-mediated mechanisms, presynaptic Ena relays the output of the highly conserved receptor protein tyrosine phosphatase Dlar and associated proteins including the heparan sulfate proteoglycan Syndecan, and the non-receptor Abelson tyrosine kinase to regulate addition of presynaptic varicosities. Interestingly, Ena also influences active zones, where it restricts active zone size, regulates the recruitment of synaptic vesicles, and controls the amplitude and frequency of spontaneous glutamate release. Conclusion We thus show that Ena, under control of the Dlar pathway, is required for presynaptic terminal morphogenesis and bouton addition and that Ena has active zone and neurotransmission phenotypes. Notably, in contrast to Dlar, Ena appears to integrate multiple pathways that regulate synapse form and function.
Collapse
Affiliation(s)
- Elizabeth M McNeill
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.
| | - Cheryl Thompson
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Brett Berke
- Department of Biology, Yale University, New Haven, CT, USA
| | - Vivian T Chou
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Jannette Rusch
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - April Duckworth
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jamin DeProto
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Alicia Taylor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA.,Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Julie Gates
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - Frank Gertler
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, England
| | | | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Atkins M, Gasmi L, Bercier V, Revenu C, Del Bene F, Hazan J, Fassier C. FIGNL1 associates with KIF1Bβ and BICD1 to restrict dynein transport velocity during axon navigation. J Cell Biol 2019; 218:3290-3306. [PMID: 31541015 PMCID: PMC6781435 DOI: 10.1083/jcb.201805128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 05/30/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023] Open
Abstract
Atkins et al. identify a new role for Fidgetin-like 1 in motor axon navigation via its regulation of bidirectional axonal transport. They show that Fidgetin-like 1 binds Kif1bβ and the opposed polarity-directed motor dynein/dynactin in a molecular complex and controls circuit wiring by reducing dynein velocity in developing motor axons. Neuronal connectivity relies on molecular motor-based axonal transport of diverse cargoes. Yet the precise players and regulatory mechanisms orchestrating such trafficking events remain largely unknown. We here report the ATPase Fignl1 as a novel regulator of bidirectional transport during axon navigation. Using a yeast two-hybrid screen and coimmunoprecipitation assays, we showed that Fignl1 binds the kinesin Kif1bβ and the dynein/dynactin adaptor Bicaudal D-1 (Bicd1) in a molecular complex including the dynactin subunit dynactin 1. Fignl1 colocalized with Kif1bβ and showed bidirectional mobility in zebrafish axons. Notably, Kif1bβ and Fignl1 loss of function similarly altered zebrafish motor axon pathfinding and increased dynein-based transport velocity of Rab3 vesicles in these navigating axons, pinpointing Fignl1/Kif1bβ as a dynein speed limiter complex. Accordingly, disrupting dynein/dynactin activity or Bicd1/Fignl1 interaction induced motor axon pathfinding defects characteristic of Fignl1 gain or loss of function, respectively. Finally, pharmacological inhibition of dynein activity partially rescued the axon pathfinding defects of Fignl1-depleted larvae. Together, our results identify Fignl1 as a key dynein regulator required for motor circuit wiring.
Collapse
Affiliation(s)
- Melody Atkins
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Laïla Gasmi
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Valérie Bercier
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Céline Revenu
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Filippo Del Bene
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
| | - Jamilé Hazan
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| | - Coralie Fassier
- Sorbonne Université, University Pierre and Marie Curie-Université Paris 6, Institut de Biologie Paris Seine, Unité de Neuroscience Paris Seine, Centre National de la Recherche Scientifique, Unité Mixte Recherche 8246, Institut National de la Santé et de la Recherche Médicale U1130, Paris, France
| |
Collapse
|
33
|
Vuilleumier R, Lian T, Flibotte S, Khan ZN, Fuchs A, Pyrowolakis G, Allan DW. Retrograde BMP signaling activates neuronal gene expression through widespread deployment of a conserved BMP-responsive cis-regulatory activation element. Nucleic Acids Res 2019; 47:679-699. [PMID: 30476189 PMCID: PMC6344883 DOI: 10.1093/nar/gky1135] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 10/25/2018] [Indexed: 12/29/2022] Open
Abstract
Retrograde Bone Morphogenetic Protein (BMP) signaling in neurons is essential for the differentiation and synaptic function of many neuronal subtypes. BMP signaling regulates these processes via Smad transcription factor activity, yet the scope and nature of Smad-dependent gene regulation in neurons are mostly unknown. Here, we applied a computational approach to predict Smad-binding cis-regulatory BMP-Activating Elements (BMP-AEs) in Drosophila, followed by transgenic in vivo reporter analysis to test their neuronal subtype enhancer activity in the larval central nervous system (CNS). We identified 34 BMP-AE-containing genomic fragments that are responsive to BMP signaling in neurons, and showed that the embedded BMP-AEs are required for this activity. RNA-seq analysis identified BMP-responsive genes in the CNS and revealed that BMP-AEs selectively enrich near BMP-activated genes. These data suggest that functional BMP-AEs control nearby BMP-activated genes, which we validated experimentally. Finally, we demonstrated that the BMP-AE motif mediates a conserved Smad-responsive function in the Drosophila and vertebrate CNS. Our results provide evidence that BMP signaling controls neuronal function by directly coordinating the expression of a battery of genes through widespread deployment of a conserved Smad-responsive cis-regulatory motif.
Collapse
Affiliation(s)
- Robin Vuilleumier
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephane Flibotte
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zaynah N Khan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alisa Fuchs
- BIOSS, Centre for Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Albert-Ludwigs University of Freiburg, Freiburg, Germany.,Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - George Pyrowolakis
- BIOSS, Centre for Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Albert-Ludwigs University of Freiburg, Freiburg, Germany
| | - Douglas W Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
Tenedini FM, Sáez González M, Hu C, Pedersen LH, Petruzzi MM, Spitzweck B, Wang D, Richter M, Petersen M, Szpotowicz E, Schweizer M, Sigrist SJ, Calderon de Anda F, Soba P. Maintenance of cell type-specific connectivity and circuit function requires Tao kinase. Nat Commun 2019; 10:3506. [PMID: 31383864 PMCID: PMC6683158 DOI: 10.1038/s41467-019-11408-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/13/2019] [Indexed: 01/05/2023] Open
Abstract
Sensory circuits are typically established during early development, yet how circuit specificity and function are maintained during organismal growth has not been elucidated. To gain insight we quantitatively investigated synaptic growth and connectivity in the Drosophila nociceptive network during larval development. We show that connectivity between primary nociceptors and their downstream neurons scales with animal size. We further identified the conserved Ste20-like kinase Tao as a negative regulator of synaptic growth required for maintenance of circuit specificity and connectivity. Loss of Tao kinase resulted in exuberant postsynaptic specializations and aberrant connectivity during larval growth. Using functional imaging and behavioral analysis we show that loss of Tao-induced ectopic synapses with inappropriate partner neurons are functional and alter behavioral responses in a connection-specific manner. Our data show that fine-tuning of synaptic growth by Tao kinase is required for maintaining specificity and behavioral output of the neuronal network during animal growth.
Collapse
Affiliation(s)
- Federico Marcello Tenedini
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Maria Sáez González
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Chun Hu
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Lisa Hedegaard Pedersen
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Mabel Matamala Petruzzi
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Bettina Spitzweck
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Denan Wang
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Melanie Richter
- Neuronal Development laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Meike Petersen
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Emanuela Szpotowicz
- Electron microscopy unit, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Michaela Schweizer
- Electron microscopy unit, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Stephan J Sigrist
- Institute of Biology, Free University Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Froylan Calderon de Anda
- Neuronal Development laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Peter Soba
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany.
| |
Collapse
|
35
|
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
|
36
|
BMP-dependent synaptic development requires Abi-Abl-Rac signaling of BMP receptor macropinocytosis. Nat Commun 2019; 10:684. [PMID: 30737382 PMCID: PMC6368546 DOI: 10.1038/s41467-019-08533-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 01/09/2019] [Indexed: 12/01/2022] Open
Abstract
Retrograde BMP trans-synaptic signaling is essential for synaptic development. Despite the importance of endocytosis-regulated BMP receptor (BMPR) control of this developmental signaling, the mechanism remains unknown. Here, we provide evidence that Abelson interactor (Abi), a substrate for Abl kinase and component of the SCAR/WAVE complex, links Abl and Rac1 GTPase signaling to BMPR macropinocytosis to restrain BMP-mediated synaptic development. We find that Abi acts downstream of Abl and Rac1, and that BMP ligand Glass bottom boat (Gbb) induces macropinocytosis dependent on Rac1/SCAR signaling, Abl-mediated Abi phosphorylation, and BMPR activation. Macropinocytosis acts as the major internalization route for BMPRs at the synapse in a process driven by Gbb activation and resulting in receptor degradation. Key regulators of macropinocytosis (Rabankyrin and CtBP) control BMPR trafficking to limit BMP trans-synaptic signaling. We conclude that BMP-induced macropinocytosis acts as a BMPR homeostatic mechanism to regulate BMP-mediated synaptic development. BMP ligands act as retrograde signalling molecules to regulate presynaptic development, and regulation of BMP receptors by endocytosis may be an important component of this signalling pathway. Here, the authors show that Abi-mediated macropinocytosis of BMP receptors in Drosophila larva and contributes to neuromuscular development.
Collapse
|
37
|
Ou M, Wang S, Sun M, An J, Lv H, Zeng X, Hou SX, Xie W. The PDZ-GEF Gef26 regulates synapse development and function via FasII and Rap1 at the Drosophila neuromuscular junction. Exp Cell Res 2018; 374:342-352. [PMID: 30553967 DOI: 10.1016/j.yexcr.2018.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/09/2018] [Accepted: 12/12/2018] [Indexed: 12/29/2022]
Abstract
Guanine nucleotide exchange factors (GEFs) are essential for small G proteins to activate their downstream signaling pathways, which are involved in morphogenesis, cell adhesion, and migration. Mutants of Gef26, a PDZ-GEF (PDZ domain-containing guanine nucleotide exchange factor) in Drosophila, exhibit strong defects in wings, eyes, and the reproductive and nervous systems. However, the precise roles of Gef26 in development remain unclear. In the present study, we analyzed the role of Gef26 in synaptic development and function. We identified significant decreases in bouton number and branch length at larval neuromuscular junctions (NMJs) in Gef26 mutants, and these defects were fully rescued by restoring Gef26 expression, indicating that Gef26 plays an important role in NMJ morphogenesis. In addition to the observed defects in NMJ morphology, electrophysiological analyses revealed functional defects at NMJs, and locomotor deficiency appeared in Gef26 mutant larvae. Furthermore, Gef26 regulated NMJ morphogenesis by regulating the level of synaptic Fasciclin II (FasII), a well-studied cell adhesion molecule that functions in NMJ development and remodeling. Finally, our data demonstrate that Gef26-specific small G protein Rap1 worked downstream of Gef26 to regulate the level of FasII at NMJs, possibly through a βPS integrin-mediated signaling pathway. Taken together, our findings define a novel role of Gef26 in regulating NMJ development and function.
Collapse
Affiliation(s)
- Mengzhu Ou
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Su Wang
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Mingkuan Sun
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Jinsong An
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Huihui Lv
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Xiankun Zeng
- Basic Research Laboratory, National Cancer Institute at Frederick, NIH, Frederick, MD 21702, USA
| | - Steven X Hou
- Basic Research Laboratory, National Cancer Institute at Frederick, NIH, Frederick, MD 21702, USA.
| | - Wei Xie
- The Key Laboratory of Development Genes and Human Diseases, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China.
| |
Collapse
|
38
|
Banerjee S, Riordan M. Coordinated Regulation of Axonal Microtubule Organization and Transport by Drosophila Neurexin and BMP Pathway. Sci Rep 2018; 8:17337. [PMID: 30478335 PMCID: PMC6255869 DOI: 10.1038/s41598-018-35618-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/07/2018] [Indexed: 01/23/2023] Open
Abstract
Neurexins are well known trans-synaptic cell adhesion molecules that are required for proper synaptic development and function across species. Beyond synapse organization and function, little is known about other roles Neurexins might have in the nervous system. Here we report novel phenotypic consequences of mutations in Drosophila neurexin (dnrx), which alters axonal microtubule organization and transport. We show that dnrx mutants display phenotypic similarities with the BMP receptor wishful thinking (wit) and one of the downstream effectors, futsch, which is a known regulator of microtubule organization and stability. dnrx has genetic interactions with wit and futsch. Loss of Dnrx also results in reduced levels of other downstream effectors of BMP signaling, phosphorylated-Mad and Trio. Interestingly, postsynaptic overexpression of the BMP ligand, Glass bottom boat, in dnrx mutants partially rescues the axonal transport defects but not the synapse undergrowth at the neuromuscular junctions. These data suggest that Dnrx and BMP signaling are involved in many diverse functions and that regulation of axonal MT organization and transport might be distinct from regulation of synaptic growth in dnrx mutants. Together, our work uncovers a novel function of Drosophila Neurexin and may provide insights into functions of Neurexins in vertebrates.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | - Maeveen Riordan
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.,University of Colorado School of Medicine, 12631 E. 17th Avenue B177, Aurora, CO, 80045, USA
| |
Collapse
|
39
|
Akbergenova Y, Cunningham KL, Zhang YV, Weiss S, Littleton JT. Characterization of developmental and molecular factors underlying release heterogeneity at Drosophila synapses. eLife 2018; 7:38268. [PMID: 29989549 PMCID: PMC6075867 DOI: 10.7554/elife.38268] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/30/2018] [Indexed: 12/14/2022] Open
Abstract
Neurons communicate through neurotransmitter release at specialized synaptic regions known as active zones (AZs). Using biosensors to visualize single synaptic vesicle fusion events at Drosophila neuromuscular junctions, we analyzed the developmental and molecular determinants of release probability (Pr) for a defined connection with ~300 AZs. Pr was heterogeneous but represented a stable feature of each AZ. Pr remained stable during high frequency stimulation and retained heterogeneity in mutants lacking the Ca2+ sensor Synaptotagmin 1. Pr correlated with both presynaptic Ca2+ channel abundance and Ca2+ influx at individual release sites. Pr heterogeneity also correlated with glutamate receptor abundance, with high Pr connections developing receptor subtype segregation. Intravital imaging throughout development revealed that AZs acquire high Pr during a multi-day maturation period, with Pr heterogeneity largely reflecting AZ age. The rate of synapse maturation was activity-dependent, as both increases and decreases in neuronal activity modulated glutamate receptor field size and segregation. To send a message to its neighbor, a neuron releases chemicals called neurotransmitters into the gap – or synapse – between them. The neurotransmitter molecules bind to proteins on the receiver neuron called receptors. But what causes the sender neuron to release neurotransmitter in the first place? The process starts when an electrical impulse called an action potential arrives at the sender cell. Its arrival causes channels in the membrane of the sender neuron to open, so that calcium ions flood into the cell. The calcium ions interact with packages of neurotransmitter molecules, known as synaptic vesicles. This causes some of the vesicles to empty their contents into the synapse. But this process is not particularly reliable. Only a small fraction of action potentials cause vesicles to fuse with the synaptic membrane. How likely this is to occur varies greatly between neurons, and even between synapses formed by the same neuron. Synapses that are likely to release neurotransmitter are said to be strong. They are good at passing messages from the sender neuron to the receiver. Synapses with a low probability of release are said to be weak. But what exactly differs between strong and weak synapses? Akbergenova et al. studied synapses between motor neurons and muscle cells in the fruit fly Drosophila. Each motor neuron forms several hundred synapses. Some of these synapses are 50 times more likely to release neurotransmitter than others. Using calcium imaging and genetics, Akbergenova et al. showed that sender cells at strong synapses have more calcium channels than sender cells at weak synapses. The subtypes and arrangement of receptor proteins also differ between the receiver neurons of strong versus weak synapses. Finally, studies in larvae revealed that newly formed synapses all start out weak and then gradually become stronger. How fast this strengthening occurs depends on how active the neuron at the synapse is. This study has shown, in unprecedented detail, key molecular factors that make some fruit fly synapses more likely to release neurotransmitter than others. Many proteins at synapses of mammals resemble those at fruit fly synapses. This means that similar factors may also explain differences in synaptic strength in the mammalian brain. Changes in the strength of synapses underlie the ability to learn. Furthermore, many neurological and psychiatric disorders result from disruption of synapses. Understanding the molecular basis of synapses will thus provide clues to the origins of certain brain diseases.
Collapse
Affiliation(s)
- Yulia Akbergenova
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Karen L Cunningham
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Yao V Zhang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Shirley Weiss
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
40
|
Lee PT, Zirin J, Kanca O, Lin WW, Schulze KL, Li-Kroeger D, Tao R, Devereaux C, Hu Y, Chung V, Fang Y, He Y, Pan H, Ge M, Zuo Z, Housden BE, Mohr SE, Yamamoto S, Levis RW, Spradling AC, Perrimon N, Bellen HJ. A gene-specific T2A-GAL4 library for Drosophila. eLife 2018; 7:35574. [PMID: 29565247 PMCID: PMC5898912 DOI: 10.7554/elife.35574] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
We generated a library of ~1000 Drosophila stocks in which we inserted a construct in the intron of genes allowing expression of GAL4 under control of endogenous promoters while arresting transcription with a polyadenylation signal 3’ of the GAL4. This allows numerous applications. First, ~90% of insertions in essential genes cause a severe loss-of-function phenotype, an effective way to mutagenize genes. Interestingly, 12/14 chromosomes engineered through CRISPR do not carry second-site lethal mutations. Second, 26/36 (70%) of lethal insertions tested are rescued with a single UAS-cDNA construct. Third, loss-of-function phenotypes associated with many GAL4 insertions can be reverted by excision with UAS-flippase. Fourth, GAL4 driven UAS-GFP/RFP reports tissue and cell-type specificity of gene expression with high sensitivity. We report the expression of hundreds of genes not previously reported. Finally, inserted cassettes can be replaced with GFP or any DNA. These stocks comprise a powerful resource for assessing gene function. Determining what role newly discovered genes play in the body is an important part of genetics. This task requires a lot of extra information about each gene, such as the specific cells where the gene is active, or what happens when the gene is deleted. To answer these questions, researchers need tools and methods to manipulate genes within a living organism. The fruit fly Drosophila is useful for such experiments because a toolbox of genetic techniques is already available. Gene editing in fruit flies allows small pieces of genetic information to be removed from or added to anywhere in the animal’s DNA. Another tool, known as GAL4-UAS, is a two-part system used to study gene activity. The GAL4 component is a protein that switches on genes. GAL4 alone does very little in Drosophila cells because it only recognizes a DNA sequence called UAS. However, if a GAL4-producing cell is also engineered to contain a UAS-controlled gene, GAL4 will switch the gene on. Lee et al. used gene editing to insert a small piece of DNA, containing the GAL4 sequence followed by a ‘stop’ signal, into many different fly genes. The insertion made the cells where each gene was normally active produce GAL4, but – thanks to the stop signal – rendered the rest of the original gene non-functional. This effectively deleted the proteins encoded by each gene, giving information about the biological processes they normally control. Lee et al. went on to use their insertion approach to make a Drosophila genetic library. This is a collection of around 1,000 different strains of fly, each carrying the GAL4/stop combination in a single gene. The library allows any gene in the collection to be studied in detail simply by combining the GAL4 with different UAS-controlled genetic tools. For example, introducing a UAS-controlled marker would pinpoint where in the body the original gene was active. Alternatively, adding UAS-controlled human versions of the gene would create humanized flies, which are a valuable tool to study potential disease-causing genes in humans. This Drosophila library is a resource that contributes new experimental tools to fly genetics. Insights gained from flies can also be applied to more complex animals like humans, especially since around 65% of genes are similar across humans and Drosophila. As such, Lee et al. hope that this resource will help other researchers shed new light on the role of many different genes in health and disease.
Collapse
Affiliation(s)
- Pei-Tseng Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Jonathan Zirin
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Wen-Wen Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Karen L Schulze
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - David Li-Kroeger
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Rong Tao
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Colby Devereaux
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Verena Chung
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Ying Fang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Yuchun He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Hongling Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Ming Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | | | - Stephanie E Mohr
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Robert W Levis
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, Baltimore, United States
| | - Allan C Spradling
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, Baltimore, United States
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
41
|
Xing G, Li M, Sun Y, Rui M, Zhuang Y, Lv H, Han J, Jia Z, Xie W. Neurexin-Neuroligin 1 regulates synaptic morphology and functions via the WAVE regulatory complex in Drosophila neuromuscular junction. eLife 2018. [PMID: 29537369 PMCID: PMC5873926 DOI: 10.7554/elife.30457] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuroligins are postsynaptic adhesion molecules that are essential for postsynaptic specialization and synaptic function. But the underlying molecular mechanisms of neuroligin functions remain unclear. We found that Drosophila Neuroligin 1 (DNlg1) regulates synaptic structure and function through WAVE regulatory complex (WRC)-mediated postsynaptic actin reorganization. The disruption of DNlg1, DNlg2, or their presynaptic partner neurexin (DNrx) led to a dramatic decrease in the amount of F-actin. Further study showed that DNlg1, but not DNlg2 or DNlg3, directly interacts with the WRC via its C-terminal interacting receptor sequence. That interaction is required to recruit WRC to the postsynaptic membrane to promote F-actin assembly. Furthermore, the interaction between DNlg1 and the WRC is essential for DNlg1 to rescue the morphological and electrophysiological defects in dnlg1 mutants. Our results reveal a novel mechanism by which the DNrx-DNlg1 trans-synaptic interaction coordinates structural and functional properties at the neuromuscular junction.
Collapse
Affiliation(s)
- Guanglin Xing
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Moyi Li
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yichen Sun
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Menglong Rui
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Yan Zhuang
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China
| | - Huihui Lv
- The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Junhai Han
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhengping Jia
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,Neurosciences and Mental Health Program, The Hospital for Sick Children, University of Toronto, Ontario, Canada
| | - Wei Xie
- Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, China.,The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
42
|
Liao EH, Gray L, Tsurudome K, El-Mounzer W, Elazzouzi F, Baim C, Farzin S, Calderon MR, Kauwe G, Haghighi AP. Kinesin Khc-73/KIF13B modulates retrograde BMP signaling by influencing endosomal dynamics at the Drosophila neuromuscular junction. PLoS Genet 2018; 14:e1007184. [PMID: 29373576 PMCID: PMC5802963 DOI: 10.1371/journal.pgen.1007184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 02/07/2018] [Accepted: 01/03/2018] [Indexed: 11/18/2022] Open
Abstract
Retrograde signaling is essential for neuronal growth, function and survival; however, we know little about how signaling endosomes might be directed from synaptic terminals onto retrograde axonal pathways. We have identified Khc-73, a plus-end directed microtubule motor protein, as a regulator of sorting of endosomes in Drosophila larval motor neurons. The number of synaptic boutons and the amount of neurotransmitter release at the Khc-73 mutant larval neuromuscular junction (NMJ) are normal, but we find a significant decrease in the number of presynaptic release sites. This defect in Khc-73 mutant larvae can be genetically enhanced by a partial genetic loss of Bone Morphogenic Protein (BMP) signaling or suppressed by activation of BMP signaling in motoneurons. Consistently, activation of BMP signaling that normally enhances the accumulation of phosphorylated form of BMP transcription factor Mad in the nuclei, can be suppressed by genetic removal of Khc-73. Using a number of assays including live imaging in larval motor neurons, we show that loss of Khc-73 curbs the ability of retrograde-bound endosomes to leave the synaptic area and join the retrograde axonal pathway. Our findings identify Khc-73 as a regulator of endosomal traffic at the synapse and modulator of retrograde BMP signaling in motoneurons.
Collapse
Affiliation(s)
- Edward H. Liao
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Lindsay Gray
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - Kazuya Tsurudome
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | | | - Fatima Elazzouzi
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Christopher Baim
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Sarah Farzin
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Mario R. Calderon
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA, United States of America
| | - A. Pejmun Haghighi
- Buck Institute for Research on Aging, Novato, CA, United States of America
- Department of Physiology, McGill University, Montreal, QC, Canada
- * E-mail:
| |
Collapse
|
43
|
Heo K, Nahm M, Lee MJ, Kim YE, Ki CS, Kim SH, Lee S. The Rap activator Gef26 regulates synaptic growth and neuronal survival via inhibition of BMP signaling. Mol Brain 2017; 10:62. [PMID: 29282074 PMCID: PMC5745669 DOI: 10.1186/s13041-017-0342-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/12/2017] [Indexed: 12/24/2022] Open
Abstract
In Drosophila, precise regulation of BMP signaling is essential for normal synaptic growth at the larval neuromuscular junction (NMJ) and neuronal survival in the adult brain. However, the molecular mechanisms underlying fine-tuning of BMP signaling in neurons remain poorly understood. We show that loss of the Drosophila PDZ guanine nucleotide exchange factor Gef26 significantly increases synaptic growth at the NMJ and enhances BMP signaling in motor neurons. We further show that Gef26 functions upstream of Rap1 in motor neurons to restrain synaptic growth. Synaptic overgrowth in gef26 or rap1 mutants requires BMP signaling, indicating that Gef26 and Rap1 regulate synaptic growth via inhibition of BMP signaling. We also show that Gef26 is involved in the endocytic downregulation of surface expression of the BMP receptors thickveins (Tkv) and wishful thinking (Wit). Finally, we demonstrate that loss of Gef26 also induces progressive brain neurodegeneration through Rap1- and BMP signaling-dependent mechanisms. Taken together, these results suggest that the Gef26-Rap1 signaling pathway regulates both synaptic growth and neuronal survival by controlling BMP signaling.
Collapse
Affiliation(s)
- Keunjung Heo
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea.,Department of Cell & Developmental Biology, Dental Research Institute, Seoul National University, Seoul, 03080, South Korea
| | - Minyeop Nahm
- Department of Neurology, Hanyang University College of Medicine, Seoul, 04763, South Korea
| | - Min-Jung Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea.,Department of Cell & Developmental Biology, Dental Research Institute, Seoul National University, Seoul, 03080, South Korea
| | - Young-Eun Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea
| | - Chang-Seok Ki
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University College of Medicine, Seoul, 04763, South Korea
| | - Seungbok Lee
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
44
|
Drosophila Syd-1 Has RhoGAP Activity That Is Required for Presynaptic Clustering of Bruchpilot/ELKS but Not Neurexin-1. Genetics 2017; 208:705-716. [PMID: 29217522 DOI: 10.1534/genetics.117.300538] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/22/2017] [Indexed: 12/23/2022] Open
Abstract
Syd-1 proteins are required for presynaptic development in worm, fly, and mouse. Syd-1 proteins in all three species contain a Rho GTPase activating protein (GAP)-like domain of unclear significance: invertebrate Syd-1s are thought to lack GAP activity, and mouse mSYD1A has GAP activity that is thought to be dispensable for its function. Here, we show that Drosophila melanogaster Syd-1 can interact with all six fly Rhos and has GAP activity toward Rac1 and Cdc42. During development, fly Syd-1 clusters multiple presynaptic proteins at the neuromuscular junction (NMJ), including the cell adhesion molecule Neurexin (Nrx-1) and the active zone (AZ) component Bruchpilot (Brp), both of which Syd-1 binds directly. We show that a mutant form of Syd-1 that specifically lacks GAP activity localizes normally to presynaptic sites and is sufficient to recruit Nrx-1 but fails to cluster Brp normally. We provide evidence that Syd-1 participates with Rac1 in two separate functions: (1) together with the Rac guanine exchange factor (RacGEF) Trio, GAP-active Syd-1 is required to regulate the nucleotide-bound state of Rac1, thereby promoting Brp clustering; and (2) Syd-1, independent of its GAP activity, is required for the recruitment of Nrx-1 to boutons, including the recruitment of Nrx-1 that is promoted by GTP-bound Rac1. We conclude that, contrary to current models, the GAP domain of fly Syd-1 is active and required for presynaptic development; we suggest that the same may be true of vertebrate Syd-1 proteins. In addition, our data provide new molecular insight into the ability of Rac1 to promote presynaptic development.
Collapse
|
45
|
An autism spectrum disorder-related de novo mutation hotspot discovered in the GEF1 domain of Trio. Nat Commun 2017; 8:601. [PMID: 28928363 PMCID: PMC5605661 DOI: 10.1038/s41467-017-00472-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/30/2017] [Indexed: 12/24/2022] Open
Abstract
The Rho guanine nucleotide exchange factor (RhoGEF) Trio promotes actin polymerization by directly activating the small GTPase Rac1. Recent studies suggest that autism spectrum disorder (ASD)-related behavioral phenotypes in animal models of ASD can be produced by dysregulation of Rac1’s control of actin polymerization at glutamatergic synapses. Here, in humans, we discover a large cluster of ASD-related de novo mutations in Trio’s Rac1 activating domain, GEF1. Our study reveals that these mutations produce either hypofunctional or hyperfunctional forms of Trio in rodent neurons in vitro. In accordance with pathological increases or decreases in glutamatergic neurotransmission observed in animal models of ASD, we find that these mutations result in either reduced synaptic AMPA receptor expression or enhanced glutamatergic synaptogenesis. Together, our findings implicate both excessive and reduced Trio activity and the resulting synaptic dysfunction in ASD-related pathogenesis, and point to the Trio-Rac1 pathway at glutamatergic synapses as a possible key point of convergence of many ASD-related genes. Trio is a RhoGEF protein that promotes actin polymerization and is implicated in the regulation of glutamatergic synapses in autism spectrum disorder (ASD). Here the authors identify a large cluster of de novo mutations in the GEF1 domain of Trio in whole-exome sequencing data from individuals with ASD, and confirm that some of these mutations lead to glutamatergic dysregulation in vitro.
Collapse
|
46
|
Zhang X, Rui M, Gan G, Huang C, Yi J, Lv H, Xie W. Neuroligin 4 regulates synaptic growth via the bone morphogenetic protein (BMP) signaling pathway at the Drosophila neuromuscular junction. J Biol Chem 2017; 292:17991-18005. [PMID: 28912273 PMCID: PMC5672027 DOI: 10.1074/jbc.m117.810242] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/12/2017] [Indexed: 01/26/2023] Open
Abstract
The neuroligin (Nlg) family of neural cell adhesion molecules is thought to be required for synapse formation and development and has been linked to the development of autism spectrum disorders in humans. In Drosophila melanogaster, mutations in the neuroligin 1–3 genes have been reported to induce synapse developmental defects at neuromuscular junctions (NMJs), but the role of neuroligin 4 (dnlg4) in synapse development has not been determined. Here, we report that the Drosophila neuroligin 4 (DNlg4) is different from DNlg1–3 in that it presynaptically regulates NMJ synapse development. Loss of dnlg4 results in reduced growth of NMJs with fewer synaptic boutons. The morphological defects caused by dnlg4 mutant are associated with a corresponding decrease in synaptic transmission efficacy. All of these defects could only be rescued when DNlg4 was expressed in the presynapse of NMJs. To understand the basis of DNlg4 function, we looked for genetic interactions and found connections with the components of the bone morphogenetic protein (BMP) signaling pathway. Immunostaining and Western blot analyses demonstrated that the regulation of NMJ growth by DNlg4 was due to the positive modulation of BMP signaling by DNlg4. Specifically, BMP type I receptor thickvein (Tkv) abundance was reduced in dnlg4 mutants, and immunoprecipitation assays showed that DNlg4 and Tkv physically interacted in vivo. Our study demonstrates that DNlg4 presynaptically regulates neuromuscular synaptic growth via the BMP signaling pathway by modulating Tkv.
Collapse
Affiliation(s)
- Xinwang Zhang
- From the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, Jiangsu 210096, China.,the Department of Biology, Basic Medical School of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Menglong Rui
- From the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, Jiangsu 210096, China.,Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing, Jiangsu 210096, China, and
| | - Guangmin Gan
- Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing, Jiangsu 210096, China, and
| | - Cong Huang
- From the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, Jiangsu 210096, China
| | - Jukang Yi
- Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing, Jiangsu 210096, China, and
| | - Huihui Lv
- From the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, Jiangsu 210096, China.,Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing, Jiangsu 210096, China, and
| | - Wei Xie
- From the Institute of Life Sciences, the Collaborative Innovation Center for Brain Science, Southeast University, Nanjing, Jiangsu 210096, China, .,Key Laboratory of Developmental Genes and Human Disease, Jiangsu Co-innovation Center of Neuroregeneration, Southeast University, Nanjing, Jiangsu 210096, China, and
| |
Collapse
|
47
|
Upadhyay A, Moss-Taylor L, Kim MJ, Ghosh AC, O'Connor MB. TGF-β Family Signaling in Drosophila. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022152. [PMID: 28130362 DOI: 10.1101/cshperspect.a022152] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The transforming growth factor β (TGF-β) family signaling pathway is conserved and ubiquitous in animals. In Drosophila, fewer representatives of each signaling component are present compared with vertebrates, simplifying mechanistic study of the pathway. Although there are fewer family members, the TGF-β family pathway still regulates multiple and diverse functions in Drosophila. In this review, we focus our attention on several of the classic and best-studied functions for TGF-β family signaling in regulating Drosophila developmental processes such as embryonic and imaginal disc patterning, but we also describe several recently discovered roles in regulating hormonal, physiological, neuronal, innate immunity, and tissue homeostatic processes.
Collapse
Affiliation(s)
- Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Lindsay Moss-Taylor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Arpan C Ghosh
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
48
|
Shilts J, Broadie K. Secreted tissue inhibitor of matrix metalloproteinase restricts trans-synaptic signaling to coordinate synaptogenesis. J Cell Sci 2017; 130:2344-2358. [PMID: 28576972 DOI: 10.1242/jcs.200808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/29/2017] [Indexed: 12/20/2022] Open
Abstract
Synaptogenesis is coordinated by trans-synaptic signals that traverse the specialized synaptomatrix between presynaptic and postsynaptic cells. Matrix metalloproteinase (Mmp) activity sculpts this environment, balanced by secreted tissue inhibitors of Mmp (Timp). Here, we use the simplified Drosophila melanogaster matrix metalloproteome to test the consequences of eliminating all Timp regulatory control of Mmp activity at the neuromuscular junction (NMJ). Using in situ zymography, we find Timp limits Mmp activity at the NMJ terminal and shapes extracellular proteolytic dynamics surrounding individual synaptic boutons. In newly generated timp null mutants, NMJs exhibit architectural overelaboration with supernumerary synaptic boutons. With cell-targeted RNAi and rescue studies, we find that postsynaptic Timp limits presynaptic architecture. Functionally, timp null mutants exhibit compromised synaptic vesicle cycling, with activity that is lower in amplitude and fidelity. NMJ defects manifest in impaired locomotor function. Mechanistically, we find that Timp limits BMP trans-synaptic signaling and the downstream synapse-to-nucleus signal transduction. Pharmacologically restoring Mmp inhibition in timp null mutants corrects bone morphogenetic protein (BMP) signaling and synaptic properties. Genetically restoring BMP signaling in timp null mutants corrects NMJ structure and motor function. Thus, Timp inhibition of Mmp proteolytic activity restricts BMP trans-synaptic signaling to coordinate synaptogenesis.
Collapse
Affiliation(s)
- Jarrod Shilts
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
49
|
Van Vactor D, Sigrist SJ. Presynaptic morphogenesis, active zone organization and structural plasticity in Drosophila. Curr Opin Neurobiol 2017; 43:119-129. [PMID: 28388491 DOI: 10.1016/j.conb.2017.03.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/31/2022]
Abstract
Effective adaptation of neural circuit function to a changing environment requires many forms of plasticity. Among these, structural plasticity is one of the most durable, and is also an intrinsic part of the developmental logic for the formation and refinement of synaptic connectivity. Structural plasticity of presynaptic sites can involve the addition, remodeling, or removal of pre- and post-synaptic elements. However, this requires coordination of morphogenesis and assembly of the subcellular machinery for neurotransmitter release within the presynaptic neuron, as well as coordination of these events with the postsynaptic cell. While much progress has been made in revealing the cell biological mechanisms of postsynaptic structural plasticity, our understanding of presynaptic mechanisms is less complete.
Collapse
Affiliation(s)
- David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA; Okinawa Institute of Science and Technology, Graduate University, Tancha 1919-1, Onna-son, Okinawa, Japan.
| | - Stephan J Sigrist
- Institut für Biologie/Genetik and NeuroCure, Freie Universität Berlin, Takustrasse 6, D-14195 Berlin, Germany.
| |
Collapse
|
50
|
Terry-Lorenzo RT, Torres VI, Wagh D, Galaz J, Swanson SK, Florens L, Washburn MP, Waites CL, Gundelfinger ED, Reimer RJ, Garner CC. Trio, a Rho Family GEF, Interacts with the Presynaptic Active Zone Proteins Piccolo and Bassoon. PLoS One 2016; 11:e0167535. [PMID: 27907191 PMCID: PMC5132261 DOI: 10.1371/journal.pone.0167535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/15/2016] [Indexed: 12/17/2022] Open
Abstract
Synaptic vesicles (SVs) fuse with the plasma membrane at a precise location called the presynaptic active zone (AZ). This fusion is coordinated by proteins embedded within a cytoskeletal matrix assembled at the AZ (CAZ). In the present study, we have identified a novel binding partner for the CAZ proteins Piccolo and Bassoon. This interacting protein, Trio, is a member of the Dbl family of guanine nucleotide exchange factors (GEFs) known to regulate the dynamic assembly of actin and growth factor dependent axon guidance and synaptic growth. Trio was found to interact with the C-terminal PBH 9/10 domains of Piccolo and Bassoon via its own N-terminal Spectrin repeats, a domain that is also critical for its localization to the CAZ. Moreover, our data suggest that regions within the C-terminus of Trio negatively regulate its interactions with Piccolo/Bassoon. These findings provide a mechanism for the presynaptic targeting of Trio and support a model in which Piccolo and Bassoon play a role in regulating neurotransmission through interactions with proteins, including Trio, that modulate the dynamic assembly of F-actin during cycles of synaptic vesicle exo- and endocytosis.
Collapse
Affiliation(s)
- Ryan T. Terry-Lorenzo
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Viviana I. Torres
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
- Centro de Envejecimiento y Regeneración (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile, Alameda, Santiago, Chile
| | - Dhananjay Wagh
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Jose Galaz
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Selene K. Swanson
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Laurence Florens
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Michael P. Washburn
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Clarissa L. Waites
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
| | - Eckart D. Gundelfinger
- Dept. of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Richard J. Reimer
- Dept. of Neurology and Neurological Sciences Stanford University and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Craig C. Garner
- Dept. of Psychiatry and Behavioral Science, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California, United States of America
- German Centers for Neurodegenerative Diseases, Charité - Medical University, Berlin, Germany
- * E-mail:
| |
Collapse
|