1
|
Kang CJ, Guzmán-Clavel LE, Lei K, Koo M, To S, Roche JP. The exocyst subunit Sec15 is critical for proper synaptic development and function at the Drosophila NMJ. Mol Cell Neurosci 2024; 128:103914. [PMID: 38086519 DOI: 10.1016/j.mcn.2023.103914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
The exocyst protein complex is important for targeted vesicle fusion in a variety of cell types, however, its function in neurons is still not entirely known. We found that presynaptic knockdown (KD) of the exocyst component sec15 by transgenic RNAi expression caused a number of unexpected morphological and physiological defects in the synapse. These include the development of active zones (AZ) devoid of essential presynaptic proteins, an increase in the branching of the presynaptic arbor, the appearance of satellite boutons, and a decrease in the amplitude of stimulated postsynaptic currents as well as a decrease in the frequency of spontaneous synaptic vesicle release. We also found the release of extracellular vesicles from the presynaptic neuron was greatly diminished in the Sec15 KDs. These effects were mimicked by presynaptic knockdown of Rab11, a protein known to interact with the exocyst. sec15 RNAi expression caused an increase in phosphorylated Mothers against decapentaplegic (pMad) in the presynaptic terminal, an indication of enhanced bone morphogenic protein (BMP) signaling. Some morphological phenotypes caused by Sec15 knockdown were reduced by attenuation of BMP signaling through knockdown of wishful thinking (Wit), while other phenotypes were unaffected. Individual knockdown of multiple proteins of the exocyst complex also displayed a morphological phenotype similar to Sec15 KD. We conclude that Sec15, functioning as part of the exocyst complex, is critically important for proper formation and function of neuronal synapses. We propose a model in which Sec15 is involved in the trafficking of vesicles from the recycling endosome to the cell membrane as well as possibly trafficking extracellular vesicles for presynaptic release and these processes are necessary for the correct structure and function of the synapse.
Collapse
Affiliation(s)
- Chris J Kang
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America
| | - Luis E Guzmán-Clavel
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America
| | - Katherine Lei
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America
| | - Martin Koo
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America
| | - Steven To
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America
| | - John P Roche
- Neuroscience Program, Amherst College, Amherst, MA 01002, United States of America; Department of Biology, Amherst College, Amherst, MA 01002, United States of America.
| |
Collapse
|
2
|
Ciampelli C, Galleri G, Puggioni S, Fais M, Iannotta L, Galioto M, Becciu M, Greggio E, Bernardoni R, Crosio C, Iaccarino C. Inhibition of the Exocyst Complex Attenuates the LRRK2 Pathological Effects. Int J Mol Sci 2023; 24:12656. [PMID: 37628835 PMCID: PMC10454163 DOI: 10.3390/ijms241612656] [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: 06/09/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Pathological mutations in leucine-rich repeat kinase 2 (LRRK2) gene are the major genetic cause of Parkinson's disease (PD). Multiple lines of evidence link LRRK2 to the control of vesicle dynamics through phosphorylation of a subset of RAB proteins. However, the molecular mechanisms underlying these processes are not fully elucidated. We have previously demonstrated that LRRK2 increases the exocyst complex assembly by Sec8 interaction, one of the eight members of the exocyst complex, and that Sec8 over-expression mitigates the LRRK2 pathological effect in PC12 cells. Here, we extend this analysis using LRRK2 drosophila models and show that the LRRK2-dependent exocyst complex assembly increase is downstream of RAB phosphorylation. Moreover, exocyst complex inhibition rescues mutant LRRK2 pathogenic phenotype in cellular and drosophila models. Finally, prolonged exocyst inhibition leads to a significant reduction in the LRRK2 protein level, overall supporting the role of the exocyst complex in the LRRK2 pathway. Taken together, our study suggests that modulation of the exocyst complex may represent a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Cristina Ciampelli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Grazia Galleri
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Silvia Puggioni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Milena Fais
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Lucia Iannotta
- Department of Biology, University of Padova, 35131 Padova, Italy; (L.I.); (E.G.)
| | - Manuela Galioto
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Marta Becciu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Elisa Greggio
- Department of Biology, University of Padova, 35131 Padova, Italy; (L.I.); (E.G.)
| | - Roberto Bernardoni
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy;
| | - Claudia Crosio
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| | - Ciro Iaccarino
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.C.); (G.G.); (S.P.); (M.F.); (M.G.); (M.B.); (C.C.)
| |
Collapse
|
3
|
Cullell N, Soriano-Tárraga C, Gallego-Fábrega C, Cárcel-Márquez J, Muiño E, Llucià-Carol L, Lledós M, Esteller M, de Moura MC, Montaner J, Rosell A, Delgado P, Martí-Fábregas J, Krupinski J, Roquer J, Jiménez-Conde J, Fernández-Cadenas I. Altered methylation pattern in EXOC4 is associated with stroke outcome: an epigenome-wide association study. Clin Epigenetics 2022; 14:124. [PMID: 36180927 PMCID: PMC9526296 DOI: 10.1186/s13148-022-01340-5] [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: 05/20/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND AND PURPOSE The neurological course after stroke is highly variable and is determined by demographic, clinical and genetic factors. However, other heritable factors such as epigenetic DNA methylation could play a role in neurological changes after stroke. METHODS We performed a three-stage epigenome-wide association study to evaluate DNA methylation associated with the difference between the National Institutes of Health Stroke Scale (NIHSS) at baseline and at discharge (ΔNIHSS) in ischaemic stroke patients. DNA methylation data in the Discovery (n = 643) and Replication (n = 62) Cohorts were interrogated with the 450 K and EPIC BeadChip. Nominal CpG sites from the Discovery (p value < 10-06) were also evaluated in a meta-analysis of the Discovery and Replication cohorts, using a random-fixed effect model. Metabolic pathway enrichment was calculated with methylGSA. We integrated the methylation data with 1305 plasma protein expression levels measured by SOMAscan in 46 subjects and measured RNA expression with RT-PCR in a subgroup of 13 subjects. Specific cell-type methylation was assessed using EpiDISH. RESULTS The meta-analysis revealed an epigenome-wide significant association in EXOC4 (p value = 8.4 × 10-08) and in MERTK (p value = 1.56 × 10-07). Only the methylation in EXOC4 was also associated in the Discovery and in the Replication Cohorts (p value = 1.14 × 10-06 and p value = 1.3 × 10-02, respectively). EXOC4 methylation negatively correlated with the long-term outcome (coefficient = - 4.91) and showed a tendency towards a decrease in EXOC4 expression (rho = - 0.469, p value = 0.091). Pathway enrichment from the meta-analysis revealed significant associations related to the endocytosis and deubiquitination processes. Seventy-nine plasma proteins were differentially expressed in association with EXOC4 methylation. Pathway analysis of these proteins showed an enrichment in natural killer (NK) cell activation. The cell-type methylation analysis in blood also revealed a differential methylation in NK cells. CONCLUSIONS DNA methylation of EXOC4 is associated with a worse neurological course after stroke. The results indicate a potential modulation of pathways involving endocytosis and NK cells regulation.
Collapse
Affiliation(s)
- Natalia Cullell
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
- Neurology, Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MutuaTerrassa, Terrassa, Spain
- Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Carolina Soriano-Tárraga
- Neurology, Hospital del Mar, Neurovascular Research Group, IMIM, Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, Missouri, USA
- NeuroGenomics and Informatics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Cristina Gallego-Fábrega
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
| | - Jara Cárcel-Márquez
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
| | - Elena Muiño
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
| | - Laia Llucià-Carol
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
| | - Miquel Lledós
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics & Biology Program (PEBC), L'Hospitalet, Spain
- Department of Physiological Sciences II, School of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | | | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Department of Neurology, Hospital Universitario Virgen Macarena Sevilla, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | | | - Jerzy Krupinski
- Neurology, Hospital Universitari MútuaTerrassa/Fundacio Docència i Recerca MutuaTerrassa, Terrassa, Spain
- Centre for Bioscience, School of HealthCare Science, Manchester Metropolitan University, Manchester, UK
| | - Jaume Roquer
- Neurology, Hospital del Mar, Neurovascular Research Group, IMIM, Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Jordi Jiménez-Conde
- Neurology, Hospital del Mar, Neurovascular Research Group, IMIM, Universitat Autònoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Israel Fernández-Cadenas
- Stroke Pharmacogenomics and Genetics, IIB-Sant Pau, Institut de Recerca de Sant Pau, Hospital Sant Pau, C/Sant Antoni Mª Claret,167, 08025, Barcelona, Spain.
| |
Collapse
|
4
|
Peppercorn K, Kleffmann T, Jones O, Hughes S, Tate W. Secreted Amyloid Precursor Protein Alpha, a Neuroprotective Protein in the Brain Has Widespread Effects on the Transcriptome and Proteome of Human Inducible Pluripotent Stem Cell-Derived Glutamatergic Neurons Related to Memory Mechanisms. Front Neurosci 2022; 16:858524. [PMID: 35692428 PMCID: PMC9179159 DOI: 10.3389/fnins.2022.858524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/14/2022] [Indexed: 11/18/2022] Open
Abstract
Secreted amyloid precursor protein alpha (sAPPα) processed from a parent human brain protein, APP, can modulate learning and memory. It has potential for development as a therapy preventing, delaying, or even reversing Alzheimer’s disease. In this study a comprehensive analysis to understand how it affects the transcriptome and proteome of the human neuron was undertaken. Human inducible pluripotent stem cell (iPSC)-derived glutamatergic neurons in culture were exposed to 1 nM sAPPα over a time course and changes in the transcriptome and proteome were identified with RNA sequencing and Sequential Window Acquisition of All THeoretical Fragment Ion Spectra-Mass Spectrometry (SWATH-MS), respectively. A large subset (∼30%) of differentially expressed transcripts and proteins were functionally involved with the molecular biology of learning and memory, consistent with reported links of sAPPα to memory enhancement, as well as neurogenic, neurotrophic, and neuroprotective phenotypes in previous studies. Differentially regulated proteins included those encoded in previously identified Alzheimer’s risk genes, APP processing related proteins, proteins involved in synaptogenesis, neurotransmitters, receptors, synaptic vesicle proteins, cytoskeletal proteins, proteins involved in protein and organelle trafficking, and proteins important for cell signalling, transcriptional splicing, and functions of the proteasome and lysosome. We have identified a complex set of genes affected by sAPPα, which may aid further investigation into the mechanism of how this neuroprotective protein affects memory formation and how it might be used as an Alzheimer’s disease therapy.
Collapse
Affiliation(s)
- Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Torsten Kleffmann
- Division of Health Sciences, Research Infrastructure Centre, University of Otago, Dunedin, New Zealand
| | - Owen Jones
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Stephanie Hughes
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Warren Tate
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- *Correspondence: Warren Tate,
| |
Collapse
|
5
|
Bis-Brewer DM, Gan-Or Z, Sleiman P, Hakonarson H, Fazal S, Courel S, Cintra V, Tao F, Estiar MA, Tarnopolsky M, Boycott KM, Yoon G, Suchowersky O, Dupré N, Cheng A, Lloyd TE, Rouleau G, Schüle R, Züchner S. Assessing non-Mendelian inheritance in inherited axonopathies. Genet Med 2020; 22:2114-2119. [PMID: 32741968 PMCID: PMC7710562 DOI: 10.1038/s41436-020-0924-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Inherited axonopathies (IA) are rare, clinically and genetically heterogeneous diseases that lead to length-dependent degeneration of the long axons in central (hereditary spastic paraplegia [HSP]) and peripheral (Charcot-Marie-Tooth type 2 [CMT2]) nervous systems. Mendelian high-penetrance alleles in over 100 different genes have been shown to cause IA; however, about 50% of IA cases do not receive a genetic diagnosis. A more comprehensive spectrum of causative genes and alleles is warranted, including causative and risk alleles, as well as oligogenic multilocus inheritance. METHODS Through international collaboration, IA exome studies are beginning to be sufficiently powered to perform a pilot rare variant burden analysis. After extensive quality control, our cohort contained 343 CMT cases, 515 HSP cases, and 935 non-neurological controls. We assessed the cumulative mutational burden across disease genes, explored the evidence for multilocus inheritance, and performed an exome-wide rare variant burden analysis. RESULTS We replicated the previously described mutational burden in a much larger cohort of CMT cases, and observed the same effect in HSP cases. We identified a preliminary risk allele for CMT in the EXOC4 gene (p value= 6.9 × 10-6, odds ratio [OR] = 2.1) and explored the possibility of multilocus inheritance in IA. CONCLUSION Our results support the continuing emergence of complex inheritance mechanisms in historically Mendelian disorders.
Collapse
Affiliation(s)
- Dana M Bis-Brewer
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Patrick Sleiman
- Center for Applied Genomics, The Children's Hospital of Philadelphia; Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia; Division of Human Genetics, Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Fazal
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Steve Courel
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vivian Cintra
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Feifei Tao
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mehrdad A Estiar
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| | - Mark Tarnopolsky
- Neuromuscular and Neurometabolics Division, Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Grace Yoon
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Oksana Suchowersky
- Department of Medicine, Medical Genetics and Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Nicolas Dupré
- Division of Neurosciences, CHU de Québec, Université Laval, Québec City, QC, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Andrew Cheng
- Department of Neurology and Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Thomas E Lloyd
- Department of Neurology and Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Guy Rouleau
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Rebecca Schüle
- Center for Neurology and Hertie Institute für Clinical Brain Research, University of Tübingen, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
6
|
Silva-Rodrigues JF, Patrício-Rodrigues CF, de Sousa-Xavier V, Augusto PM, Fernandes AC, Farinho AR, Martins JP, Teodoro RO. Peripheral axonal ensheathment is regulated by RalA GTPase and the exocyst complex. Development 2020; 147:dev.174540. [PMID: 31969325 DOI: 10.1242/dev.174540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/14/2020] [Indexed: 12/21/2022]
Abstract
Axon ensheathment is fundamental for fast impulse conduction and the normal physiological functioning of the nervous system. Defects in axonal insulation lead to debilitating conditions, but, despite its importance, the molecular players responsible are poorly defined. Here, we identify RalA GTPase as a key player in axon ensheathment in Drosophila larval peripheral nerves. We demonstrate through genetic analysis that RalA action through the exocyst complex is required in wrapping glial cells to regulate their growth and development. We suggest that the RalA-exocyst pathway controls the targeting of secretory vesicles for membrane growth or for the secretion of a wrapping glia-derived factor that itself regulates growth. In summary, our findings provide a new molecular understanding of the process by which axons are ensheathed in vivo, a process that is crucial for normal neuronal function.
Collapse
Affiliation(s)
- Joana F Silva-Rodrigues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Cátia F Patrício-Rodrigues
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Vicente de Sousa-Xavier
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Pedro M Augusto
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Ana C Fernandes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Ana R Farinho
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - João P Martins
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rita O Teodoro
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School - Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| |
Collapse
|
7
|
Drosophila Exo70 Is Essential for Neurite Extension and Survival under Thermal Stress. J Neurosci 2018; 38:8071-8086. [PMID: 30209205 DOI: 10.1523/jneurosci.0620-18.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 11/21/2022] Open
Abstract
The octomeric exocyst complex governs the final step of exocytosis in both plants and animals. Its roles, however, extend beyond exocytosis and include organelle biogenesis, ciliogenesis, cell migration, and cell growth. Exo70 is a conserved component of the exocyst whose function in Drosophila is unclear. In this study, we characterized two mutant alleles of Drosophila exo70. exo70 mutants exhibit reduced synaptic growth, locomotor activity, glutamate receptor density, and mEPSP amplitude. We found that presynaptic Exo70 is necessary for normal synaptic growth at the neuromuscular junction (NMJ). At the neuromuscular junction, exo70 genetically interacts with the small GTPase ralA to regulate synaptic growth. Loss of Exo70 leads to the blockage of JNK signaling-, activity-, and temperature-induced synaptic outgrowths. We showed that this phenotype is associated with an impairment of integral membrane protein transport to the cell surface at synaptic terminals. In octopaminergic motor neurons, Exo70 is detected in synaptic varicosities, as well as the regions of membrane extensions in response to activity stimulation. Strikingly, mild thermal stress causes severe neurite outgrowth defects and pharate adult lethality in exo70 mutants. exo70 mutants also display defective locomotor activity in response to starvation stress. These results demonstrated that Exo70 is an important regulator of induced synaptic growth and is crucial for an organism's adaptation to environmental changes.SIGNIFICANCE STATEMENT The exocyst complex is a conserved protein complex directing secretory vesicles to the site of membrane fusion during exocytosis, which is essential for transporting proteins and membranes to the cell surface. Exo70 is a subunit of the exocyst complex whose roles in neurons remain elusive, and its function in Drosophila is unclear. In Drosophila, Exo70 is expressed in both glutamatergic and octopaminergic neurons, and presynaptic Exo70 regulates synaptic outgrowth. Moreover, exo70 mutants have impaired integral membrane transport to the cell surface at synaptic terminals and block several kinds of induced synaptic growth. Remarkably, elevated temperature causes severe arborization defects and lethality in exo70 mutants, thus underpinning the importance of Exo70 functions in development and adaptation to the environment.
Collapse
|
8
|
Bodaleo FJ, Gonzalez-Billault C. The Presynaptic Microtubule Cytoskeleton in Physiological and Pathological Conditions: Lessons from Drosophila Fragile X Syndrome and Hereditary Spastic Paraplegias. Front Mol Neurosci 2016; 9:60. [PMID: 27504085 PMCID: PMC4958632 DOI: 10.3389/fnmol.2016.00060] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/11/2016] [Indexed: 11/21/2022] Open
Abstract
The capacity of the nervous system to generate neuronal networks relies on the establishment and maintenance of synaptic contacts. Synapses are composed of functionally different presynaptic and postsynaptic compartments. An appropriate synaptic architecture is required to provide the structural basis that supports synaptic transmission, a process involving changes in cytoskeletal dynamics. Actin microfilaments are the main cytoskeletal components present at both presynaptic and postsynaptic terminals in glutamatergic synapses. However, in the last few years it has been demonstrated that microtubules (MTs) transiently invade dendritic spines, promoting their maturation. Nevertheless, the presence and functions of MTs at the presynaptic site are still a matter of debate. Early electron microscopy (EM) studies revealed that MTs are present in the presynaptic terminals of the central nervous system (CNS) where they interact with synaptic vesicles (SVs) and reach the active zone. These observations have been reproduced by several EM protocols; however, there is empirical heterogeneity in detecting presynaptic MTs, since they appear to be both labile and unstable. Moreover, increasing evidence derived from studies in the fruit fly neuromuscular junction proposes different roles for MTs in regulating presynaptic function in physiological and pathological conditions. In this review, we summarize the main findings that support the presence and roles of MTs at presynaptic terminals, integrating descriptive and biochemical analyses, and studies performed in invertebrate genetic models.
Collapse
Affiliation(s)
- Felipe J Bodaleo
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de ChileSantiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO)Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de ChileSantiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO)Santiago, Chile; The Buck Institute for Research on Aging, NovatoCA, USA
| |
Collapse
|
9
|
Hussein NA, Delaney TL, Tounsel BL, Liebl FLW. The Extracellular-Regulated Kinase Effector Lk6 is Required for Glutamate Receptor Localization at the Drosophila Neuromuscular Junction. J Exp Neurosci 2016; 10:77-91. [PMID: 27199570 PMCID: PMC4866800 DOI: 10.4137/jen.s32840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/16/2022] Open
Abstract
The proper localization and synthesis of postsynaptic glutamate receptors are essential for synaptic plasticity. Synaptic translation initiation is thought to occur via the target of rapamycin (TOR) and mitogen-activated protein kinase signal-integrating kinase (Mnk) signaling pathways, which is downstream of extracellular-regulated kinase (ERK). We used the model glutamatergic synapse, the Drosophila neuromuscular junction, to better understand the roles of the Mnk and TOR signaling pathways in synapse development. These synapses contain non-NMDA receptors that are most similar to AMPA receptors. Our data show that Lk6, the Drosophila homolog of Mnk1 and Mnk2, is required in either presynaptic neurons or postsynaptic muscle for the proper localization of the GluRIIA glutamate receptor subunit. Lk6 may signal through eukaryotic initiation factor (eIF) 4E to regulate the synaptic levels of GluRIIA as either interfering with eIF4E binding to eIF4G or expression of a nonphosphorylatable isoform of eIF4E resulted in a significant reduction in GluRIIA at the synapse. We also find that Lk6 and TOR may independently regulate synaptic levels of GluRIIA.
Collapse
Affiliation(s)
- Nizar A Hussein
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Taylor L Delaney
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Brittany L Tounsel
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
10
|
Kim YJ, Igiesuorobo O, Ramos CI, Bao H, Zhang B, Serpe M. Prodomain removal enables neto to stabilize glutamate receptors at the Drosophila neuromuscular junction. PLoS Genet 2015; 11:e1004988. [PMID: 25723514 PMCID: PMC4344203 DOI: 10.1371/journal.pgen.1004988] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/06/2015] [Indexed: 11/24/2022] Open
Abstract
Stabilization of neurotransmitter receptors at postsynaptic specializations is a key step in the assembly of functional synapses. Drosophila Neto (Neuropillin and Tolloid-like protein) is an essential auxiliary subunit of ionotropic glutamate receptor (iGluR) complexes required for the iGluRs clustering at the neuromuscular junction (NMJ). Here we show that optimal levels of Neto are crucial for stabilization of iGluRs at synaptic sites and proper NMJ development. Genetic manipulations of Neto levels shifted iGluRs distribution to extrajunctional locations. Perturbations in Neto levels also produced small NMJs with reduced synaptic transmission, but only Neto-depleted NMJs showed diminished postsynaptic components. Drosophila Neto contains an inhibitory prodomain that is processed by Furin1-mediated limited proteolysis. neto null mutants rescued with a Neto variant that cannot be processed have severely impaired NMJs and reduced iGluRs synaptic clusters. Unprocessed Neto retains the ability to engage iGluRs in vivo and to form complexes with normal synaptic transmission. However, Neto prodomain must be removed to enable iGluRs synaptic stabilization and proper postsynaptic differentiation. Synapse development is initiated by genetic programs, but is coordinated by neuronal activity, by communication between the pre- and postsynaptic compartments, and by cellular signals that integrate the status of the whole organisms and its developmental progression. The molecular mechanisms underlining these processes are poorly understood. In particular, how neurotransmitter receptors are recruited and stabilized at central synapses remain the subject of intense research. The Drosophila NMJ is a glutamatergic synapse similar in composition and physiology with mammalian central excitatory synapses. Like mammals, Drosophila utilizes auxiliary subunit(s) to modulate the formation and function of glutamatergic synapses. We have previously reported that Neto is an auxiliary protein essential for functional glutamate receptors and for organization of postsynaptic specializations. Here we report that synapse assembly and NMJ development are exquisitely sensitive to postsynaptic Neto levels. Furthermore, we show that Neto activity is controlled by Furin-type proteases, which regulate the processing and maturation of many developmentally important proteins, from growth factors and neuropeptides to extracellular matrix components. Such concerted control may serve to coordinate synapse assembly with synapse growth and developmental progression.
Collapse
Affiliation(s)
- Young-Jun Kim
- Program in Cellular Regulation and Metabolism, NICHD, NIH, Bethesda, Maryland, United States of America
| | - Oghomwen Igiesuorobo
- Program in Cellular Regulation and Metabolism, NICHD, NIH, Bethesda, Maryland, United States of America
| | - Cathy I. Ramos
- Program in Cellular Regulation and Metabolism, NICHD, NIH, Bethesda, Maryland, United States of America
| | - Hong Bao
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Bing Zhang
- Division of Biological Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Mihaela Serpe
- Program in Cellular Regulation and Metabolism, NICHD, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
11
|
Gardiol A, St Johnston D. Staufen targets coracle mRNA to Drosophila neuromuscular junctions and regulates GluRIIA synaptic accumulation and bouton number. Dev Biol 2014; 392:153-67. [PMID: 24951879 PMCID: PMC4111903 DOI: 10.1016/j.ydbio.2014.06.007] [Citation(s) in RCA: 17] [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: 03/24/2014] [Revised: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 11/28/2022]
Abstract
The post-synaptic translation of localised mRNAs has been postulated to underlie several forms of plasticity at vertebrate synapses, but the mechanisms that target mRNAs to these postsynaptic sites are not well understood. Here we show that the evolutionary conserved dsRNA binding protein, Staufen, localises to the postsynaptic side of the Drosophila neuromuscular junction (NMJ), where it is required for the localisation of coracle mRNA and protein. Staufen plays a well-characterised role in the localisation of oskar mRNA to the oocyte posterior, where Staufen dsRNA-binding domain 5 is specifically required for its translation. Removal of Staufen dsRNA-binding domain 5, disrupts the postsynaptic accumulation of Coracle protein without affecting the localisation of cora mRNA, suggesting that Staufen similarly regulates Coracle translation. Tropomyosin II, which functions with Staufen in oskar mRNA localisation, is also required for coracle mRNA localisation, suggesting that similar mechanisms target mRNAs to the NMJ and the oocyte posterior. Coracle, the orthologue of vertebrate band 4.1, functions in the anchoring of the glutamate receptor IIA subunit (GluRIIA) at the synapse. Consistent with this, staufen mutant larvae show reduced accumulation of GluRIIA at synapses. The NMJs of staufen mutant larvae have also a reduced number of synaptic boutons. Altogether, this suggests that this novel Staufen-dependent mRNA localisation and local translation pathway may play a role in the developmentally regulated growth of the NMJ.
Collapse
Affiliation(s)
- Alejandra Gardiol
- The WellcomeCRUK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, United Kingdom
| | - Daniel St Johnston
- The WellcomeCRUK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, United Kingdom.
| |
Collapse
|
12
|
Long JB, Van Vactor D. Embryonic and larval neural connectivity: progressive changes in synapse form and function at the neuromuscular junction mediated by cytoskeletal regulation. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2013; 2:747-65. [PMID: 24123935 DOI: 10.1002/wdev.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
During development, precise formation of millions of synaptic connections is critical for the formation of a functional nervous system. Synaptogenesis is a complex multistep process in which axons follow gradients of secreted and cell surface guidance cues to reach their target area, at which point they must accurately distinguish their specific target. Upon target recognition, the axonal growth cone undergoes rapid growth and morphological changes, ultimately forming a functional synapse that continues to remodel during activity-dependent plasticity. Significant evidence suggests that the underlying actin and microtubule (MT) cytoskeletons are key effectors throughout synaptogenesis downstream of numerous receptors and signaling pathways. An increasing number of cytoskeletal-associated proteins have been shown to influence actin and MT stability and dynamics and many of these regulators have been implicated during synaptic morphogenesis using both mammalian and invertebrate model systems. In this review, we present an overview of the role cytoskeletal regulators play during the formation of the Drosophila neuromuscular junction.
Collapse
Affiliation(s)
- Jennifer B Long
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
13
|
Teodoro RO, Pekkurnaz G, Nasser A, Higashi-Kovtun ME, Balakireva M, McLachlan IG, Camonis J, Schwarz TL. Ral mediates activity-dependent growth of postsynaptic membranes via recruitment of the exocyst. EMBO J 2013; 32:2039-55. [PMID: 23812009 DOI: 10.1038/emboj.2013.147] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 05/28/2013] [Indexed: 11/09/2022] Open
Abstract
Remodelling neuronal connections by synaptic activity requires membrane trafficking. We present evidence for a signalling pathway by which synaptic activity and its consequent Ca(2+) influx activate the small GTPase Ral and thereby recruit exocyst proteins to postsynaptic zones. In accord with the ability of the exocyst to direct delivery of post-Golgi vesicles, constitutively active Ral expressed in Drosophila muscle causes the exocyst to be concentrated in the region surrounding synaptic boutons and consequently enlarges the membrane folds of the postsynaptic plasma membrane (the subsynaptic reticulum, SSR). SSR growth requires Ral and the exocyst component Sec5 and Ral-induced enlargement of these membrane folds does not occur in sec5(-/-) muscles. Chronic changes in synaptic activity influence the plastic growth of this membrane in a manner consistent with activity-dependent activation of Ral. Thus, Ral regulation of the exocyst represents a control point for postsynaptic plasticity. This pathway may also function in mammals as expression of activated RalA in hippocampal neurons increases dendritic spine density in an exocyst-dependent manner and increases Sec5 in spines.
Collapse
Affiliation(s)
- Rita O Teodoro
- The F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
The cytoskeleton forms the backbone of neuronal architecture, sustaining its form and size, subcellular compartments and cargo logistics. The synaptic cytoskeleton can be categorized in the microtubule-based core cytoskeleton and the cortical membrane skeleton. While central microtubules form the fundamental basis for the construction of elaborate neuronal processes, including axons and synapses, cortical actin filaments are generally considered to function as mediators of synapse dynamics and plasticity. More recently, the submembranous network of spectrin and ankyrin molecules has been involved in the regulation of synaptic stability and maintenance. Disruption of the synaptic cytoskeleton primarily affects the stability and maturation of synapses but also secondarily disturbs neuronal communication. Consequently, a variety of inherited diseases are accompanied by cytoskeletal malfunctions, including spastic paraplegias, spinocerebellar ataxias, and mental retardation. Since the primary reasons for many of these diseases are still unknown model organisms with a conserved repertoire of cytoskeletal elements help to understand the underlying biological mechanisms. The astonishing technical as well as genetic accessibility of synapses in Drosophila has shown that loss of the cytoskeletal architecture leads to axonal transport defects, synaptic maturation deficits, and retraction of synaptic boutons, before synaptic terminals finally detach from their target cells, suggesting that similar processes could be involved in human neuronal diseases.
Collapse
Affiliation(s)
- Bernd Goellner
- Heinrich-Heine-University Düsseldorf, Functional Cell Morphology Lab, Düsseldorf, Germany
| | | |
Collapse
|
15
|
Ganesan S, Karr JE, Featherstone DE. Drosophila glutamate receptor mRNA expression and mRNP particles. RNA Biol 2011; 8:771-81. [PMID: 21743295 PMCID: PMC3256355 DOI: 10.4161/rna.8.5.16014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 03/31/2011] [Indexed: 02/07/2023] Open
Abstract
The processes controlling glutamate receptor expression early in synaptogenesis are poorly understood. Here, we examine glutamate receptor (GluR) subunit mRNA expression and localization in Drosophila embryonic/larval neuromuscular junctions (NMJs). We show that postsynaptic GluR subunit gene expression is triggered by contact from the presynaptic nerve, approximately halfway through embryogenesis. After contact, GluRIIA and GluRIIB mRNA abundance rises quickly approximately 20-fold, then falls within a few hours back to very low levels. Protein abundance, however, gradually increases throughout development. At the same time that mRNA levels decrease following their initial spike, GluRIIA, GluRIIB, and GluRIIC subunit mRNA aggregates become visible in the cytoplasm of postsynaptic muscle cells. These mRNA aggregates do not colocalize with eIF4E, but nevertheless presumably represent mRNP particles of unknown function. Multiplex FISH shows that different GluR subunit mRNAs are found in different mRNPs. GluRIIC mRNPs are most common, followed by GluRIIA and then GluRIIB mRNPs. GluR mRNP density is not increased near NMJs, for any subunit; if anything, GluR mRNP density is highest away from NMJs and near nuclei. These results reveal some of the earliest events in postsynaptic development and provide a foundation for future studies of GluR mRNA biology.
Collapse
Affiliation(s)
- Subhashree Ganesan
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
16
|
Ganesan S, Karr JE, Featherstone DE. Drosophila glutamate receptor mRNA expression and mRNP particles. RNA Biol 2011; 8. [PMID: 21743295 PMCID: PMC3256355 DOI: 10.4161/rna.8.6.16014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The processes controlling glutamate receptor expression early in synaptogenesis are poorly understood. Here, we examine glutamate receptor (GluR) subunit mRNA expression and localization in Drosophila embryonic/larval neuromuscular junctions (NMJs). We show that postsynaptic GluR subunit gene expression is triggered by contact from the presynaptic nerve, approximately halfway through embryogenesis. After contact, GluRIIA and GluRIIB mRNA abundance rises quickly approximately 20-fold, then falls within a few hours back to very low levels. Protein abundance, however, gradually increases throughout development. At the same time that mRNA levels decrease following their initial spike, GluRIIA, GluRIIB, and GluRIIC subunit mRNA aggregates become visible in the cytoplasm of postsynaptic muscle cells. These mRNA aggregates do not colocalize with eIF4E, but nevertheless presumably represent mRNP particles of unknown function. Multiplex FISH shows that different GluR subunit mRNAs are found in different mRNPs. GluRIIC mRNPs are most common, followed by GluRIIA and then GluRIIB mRNPs. GluR mRNP density is not increased near NMJs, for any subunit; if anything, GluR mRNP density is highest away from NMJs and near nuclei. These results reveal some of the earliest events in postsynaptic development and provide a foundation for future studies of GluR mRNA biology.
Collapse
|
17
|
Nahm M, Long AA, Paik SK, Kim S, Bae YC, Broadie K, Lee S. The Cdc42-selective GAP rich regulates postsynaptic development and retrograde BMP transsynaptic signaling. ACTA ACUST UNITED AC 2010; 191:661-75. [PMID: 21041451 PMCID: PMC3003324 DOI: 10.1083/jcb.201007086] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inhibition of Cdc42 by dRich induces postsynaptic release of the BMP ligand Glass bottom boat. Retrograde bone morphogenetic protein signaling mediated by the Glass bottom boat (Gbb) ligand modulates structural and functional synaptogenesis at the Drosophila melanogaster neuromuscular junction. However, the molecular mechanisms regulating postsynaptic Gbb release are poorly understood. In this study, we show that Drosophila Rich (dRich), a conserved Cdc42-selective guanosine triphosphatase–activating protein (GAP), inhibits the Cdc42–Wsp pathway to stimulate postsynaptic Gbb release. Loss of dRich causes synaptic undergrowth and strongly impairs neurotransmitter release. These presynaptic defects are rescued by targeted postsynaptic expression of wild-type dRich but not a GAP-deficient mutant. dRich inhibits the postsynaptic localization of the Cdc42 effector Wsp (Drosophila orthologue of mammalian Wiskott-Aldrich syndrome protein, WASp), and manifestation of synaptogenesis defects in drich mutants requires Wsp signaling. In addition, dRich regulates postsynaptic organization independently of Cdc42. Importantly, dRich increases Gbb release and elevates presynaptic phosphorylated Mad levels. We propose that dRich coordinates the Gbb-dependent modulation of synaptic growth and function with postsynaptic development.
Collapse
Affiliation(s)
- Minyeop Nahm
- Interdisplinary Program in Brain Science, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
18
|
Franco M, Seyfried NT, Brand AH, Peng J, Mayor U. A novel strategy to isolate ubiquitin conjugates reveals wide role for ubiquitination during neural development. Mol Cell Proteomics 2010; 10:M110.002188. [PMID: 20861518 PMCID: PMC3098581 DOI: 10.1074/mcp.m110.002188] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ubiquitination has essential roles in neuronal development and function. Ubiquitin proteomics studies on yeast and HeLa cells have proven very informative, but there still is a gap regarding neuronal tissue-specific ubiquitination. In an organism context, direct evidence for the ubiquitination of neuronal proteins is even scarcer. Here, we report a novel proteomics strategy based on the in vivo biotinylation of ubiquitin to isolate ubiquitin conjugates from the neurons of Drosophila melanogaster embryos. We confidently identified 48 neuronal ubiquitin substrates, none of which was yet known to be ubiquitinated. Earlier proteomics and biochemical studies in non-neuronal cell types had identified orthologs to some of those but not to others. The identification here of novel ubiquitin substrates, those with no known ubiquitinated ortholog, suggests that proteomics studies must be performed on neuronal cells to identify ubiquitination pathways not shared by other cell types. Importantly, several of those newly found neuronal ubiquitin substrates are key players in synaptogenesis. Mass spectrometry results were validated by Western blotting to confirm that those proteins are indeed ubiquitinated in the Drosophila embryonic nervous system and to elucidate whether they are mono- or polyubiquitinated. In addition to the ubiquitin substrates, we also identified the ubiquitin carriers that are active during synaptogenesis. Identifying endogenously ubiquitinated proteins in specific cell types, at specific developmental stages, and within the context of a living organism will allow understanding how the tissue-specific function of those proteins is regulated by the ubiquitin system.
Collapse
Affiliation(s)
- Maribel Franco
- CIC Biogune, Bizkaia Teknologi Parkea, 48160 Derio, Spain
| | | | | | | | | |
Collapse
|
19
|
Liebl FLW, McKeown C, Yao Y, Hing HK. Mutations in Wnt2 alter presynaptic motor neuron morphology and presynaptic protein localization at the Drosophila neuromuscular junction. PLoS One 2010; 5:e12778. [PMID: 20856675 PMCID: PMC2939895 DOI: 10.1371/journal.pone.0012778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Accepted: 08/24/2010] [Indexed: 01/02/2023] Open
Abstract
Wnt proteins are secreted proteins involved in a number of developmental processes including neural development and synaptogenesis. We sought to determine the role of the Drosophila Wnt7b ortholog, Wnt2, using the neuromuscular junction (NMJ). Mutations in wnt2 produce an increase in the number of presynaptic branches and a reduction in immunolabeling of the active zone proteins, Bruchpilot and synaptobrevin, at the NMJ. There was no change, however, in immunolabeling for the presynaptic proteins cysteine-string protein (CSP) and synaptotagmin, nor the postsynaptic proteins GluRIIA and DLG at the NMJ. Consistent with the presynaptic defects, wnt2 mutants exhibit approximately a 50% reduction in evoked excitatory junctional currents. Rescue, RNAi, and tissue-specific qRT-PCR experiments indicate that Wnt2 is expressed by the postsynaptic cell where it may serve as a retrograde signal that regulates presynaptic morphology and the localization of presynaptic proteins.
Collapse
Affiliation(s)
- Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America.
| | | | | | | |
Collapse
|
20
|
Owald D, Fouquet W, Schmidt M, Wichmann C, Mertel S, Depner H, Christiansen F, Zube C, Quentin C, Körner J, Urlaub H, Mechtler K, Sigrist SJ. A Syd-1 homologue regulates pre- and postsynaptic maturation in Drosophila. ACTA ACUST UNITED AC 2010; 188:565-79. [PMID: 20176924 PMCID: PMC2828917 DOI: 10.1083/jcb.200908055] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A proteomics approach identifies Drosophila Syd-1 as a Bruchpilot binding partner that controls maturation on both sides of the neuromuscular junction. Active zones (AZs) are presynaptic membrane domains mediating synaptic vesicle fusion opposite postsynaptic densities (PSDs). At the Drosophila neuromuscular junction, the ELKS family member Bruchpilot (BRP) is essential for dense body formation and functional maturation of AZs. Using a proteomics approach, we identified Drosophila Syd-1 (DSyd-1) as a BRP binding partner. In vivo imaging shows that DSyd-1 arrives early at nascent AZs together with DLiprin-α, and both proteins localize to the AZ edge as the AZ matures. Mutants in dsyd-1 form smaller terminals with fewer release sites, and release less neurotransmitter. The remaining AZs are often large and misshapen, and ectopic, electron-dense accumulations of BRP form in boutons and axons. Furthermore, glutamate receptor content at PSDs increases because of excessive DGluRIIA accumulation. The AZ protein DSyd-1 is needed to properly localize DLiprin-α at AZs, and seems to control effective nucleation of newly forming AZs together with DLiprin-α. DSyd-1 also organizes trans-synaptic signaling to control maturation of PSD composition independently of DLiprin-α.
Collapse
Affiliation(s)
- David Owald
- Department of Genetics, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Andersen NJ, Yeaman C. Sec3-containing exocyst complex is required for desmosome assembly in mammalian epithelial cells. Mol Biol Cell 2009; 21:152-64. [PMID: 19889837 PMCID: PMC2801709 DOI: 10.1091/mbc.e09-06-0459] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In epithelial cells, Sec3 associates with Exocyst complexes enriched at desmosomes and centrosomes, distinct from Sec6/8 complexes at the apical junctional complex. RNAi-mediated suppression of Sec3 alters trafficking of desmosomal cadherins and impairs desmosome morphology and function, without noticeable effect on adherens junctions. The Exocyst is a conserved multisubunit complex involved in the docking of post-Golgi transport vesicles to sites of membrane remodeling during cellular processes such as polarization, migration, and division. In mammalian epithelial cells, Exocyst complexes are recruited to nascent sites of cell–cell contact in response to E-cadherin–mediated adhesive interactions, and this event is an important early step in the assembly of intercellular junctions. Sec3 has been hypothesized to function as a spatial landmark for the development of polarity in budding yeast, but its role in epithelial cells has not been investigated. Here, we provide evidence in support of a function for a Sec3-containing Exocyst complex in the assembly or maintenance of desmosomes, adhesive junctions that link intermediate filament networks to sites of strong intercellular adhesion. We show that Sec3 associates with a subset of Exocyst complexes that are enriched at desmosomes. Moreover, we found that membrane recruitment of Sec3 is dependent on cadherin-mediated adhesion but occurs later than that of the known Exocyst components Sec6 and Sec8 that are recruited to adherens junctions. RNA interference-mediated suppression of Sec3 expression led to specific impairment of both the morphology and function of desmosomes, without noticeable effect on adherens junctions. These results suggest that two different exocyst complexes may function in basal–lateral membrane trafficking and will enable us to better understand how exocytosis is spatially organized during development of epithelial plasma membrane domains.
Collapse
|
22
|
Karr J, Vagin V, Chen K, Ganesan S, Olenkina O, Gvozdev V, Featherstone DE. Regulation of glutamate receptor subunit availability by microRNAs. ACTA ACUST UNITED AC 2009; 185:685-97. [PMID: 19433455 PMCID: PMC2711579 DOI: 10.1083/jcb.200902062] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The efficacy of synaptic transmission depends, to a large extent, on postsynaptic receptor abundance. The molecular mechanisms controlling receptor abundance are poorly understood. We tested whether abundance of postsynaptic glutamate receptors (GluRs) in Drosophila neuromuscular junctions is controlled by microRNAs, and provide evidence that it is. We show here that postsynaptic knockdown of dicer-1, the endoribonuclease necessary for microRNA synthesis, leads to large increases in postsynaptic GluR subunit messenger RNA and protein. Specifically, we measured increases in GluRIIA and GluRIIB but not GluRIIC. Further, knockout of MiR-284, a microRNA predicted to bind to GluRIIA and GluRIIB but not GluRIIC, increases expression of GluRIIA and GluRIIB but not GluRIIC proportional to the number of predicted binding sites in each transcript. Most of the de-repressed GluR protein, however, does not appear to be incorporated into functional receptors, and only minor changes in synaptic strength are observed, which suggests that microRNAs primarily regulate Drosophila receptor subunit composition rather than overall receptor abundance or synaptic strength.
Collapse
Affiliation(s)
- Julie Karr
- Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Formin-dependent synaptic growth: evidence that Dlar signals via Diaphanous to modulate synaptic actin and dynamic pioneer microtubules. J Neurosci 2008; 28:11111-23. [PMID: 18971454 DOI: 10.1523/jneurosci.0833-08.2008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The diaphanous gene is the founding member of a family of Diaphanous-related formin proteins (DRFs). We identified diaphanous in a screen for genes that are necessary for the normal growth and stabilization of the Drosophila neuromuscular junction (NMJ). Here, we demonstrate that diaphanous mutations perturb synaptic growth at the NMJ. Diaphanous protein is present both presynaptically and postsynaptically. However, genetic rescue experiments in combination with additional genetic interaction experiments support the conclusion that dia is necessary presynaptically for normal NMJ growth. We then document defects in both the actin and microtubule cytoskeletons in dia mutant nerve terminals. In so doing, we define and characterize a population of dynamic pioneer microtubules within the NMJ that are distinct from the bundled core of microtubules identified by the MAP1b-like protein Futsch. Defects in both synaptic actin and dynamic pioneer microtubules are correlated with impaired synaptic growth in dia mutants. Finally, we present genetic evidence that Dia functions downstream of the presynaptic receptor tyrosine phosphatase Dlar and the Rho-type GEF (guanine nucleotide exchange factor) trio to control NMJ growth. Based on the established function of DRFs as Rho-GTPase-dependent regulators of the cell cytoskeleton, we propose a model in which Diaphanous links receptor tyrosine phosphatase signaling at the plasma membrane to growth-dependent modulation of the synaptic actin and microtubule cytoskeletons.
Collapse
|
24
|
Liebl FLW, Featherstone DE. Identification and investigation of Drosophila postsynaptic density homologs. Bioinform Biol Insights 2008; 2:369-81. [PMID: 19812789 PMCID: PMC2735971 DOI: 10.4137/bbi.s2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AMPA receptors are responsible for fast excitatory transmission in the CNS and the trafficking of these receptors has been implicated in LTP and learning and memory. These receptors reside in the postsynaptic density, a network of proteins that links the receptors to downstream signaling components and to the neuronal cytoskeleton. To determine whether the fruit fly, Drosophila melanogaster, possesses a similar array of proteins as are found at the mammalian PSD, we identified Drosophila homologs of 95.8% of mammalian PSD proteins. We investigated, for the first time, the role of one of these PSD proteins, Pod1 in GluR cluster formation at the Drosophila neuromuscular junction and found that mutations in pod1 resulted in a specific loss of A-type receptors at the synapse.
Collapse
Affiliation(s)
- Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA. fl
| | | |
Collapse
|
25
|
Chen K, Augustin H, Featherstone DE. Effect of ambient extracellular glutamate on Drosophila glutamate receptor trafficking and function. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2008; 195:21-9. [PMID: 18941757 DOI: 10.1007/s00359-008-0378-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 09/25/2008] [Accepted: 09/26/2008] [Indexed: 02/06/2023]
Abstract
Measurements suggest that the hemolymph glutamate concentrations in Drosophila are relatively high. This raises the possibility that extracellular glutamate could be an important regulator of glutamatergic transmission in vivo. Using voltage clamp electrophysiology, we found that synaptic currents in D. melanogaster larval neuromuscular junctions are reduced by extracellular glutamate (EC50: approximately 0.4 mM), such that only 10-30% of receptors were functionally available in 1 mM extracellular glutamate. The kinetics of synaptic currents were also slowed in a dose-dependent fashion (EC50: approximately 1 mM), consistent with the idea that extracellular glutamate preferentially removes the fastest-desensitizing receptors from the functional pool. Prolonged exposure (several hours) to extracellular glutamate also triggers loss of glutamate receptor immunoreactivity from neuromuscular junctions. To determine whether this receptor loss requires that glutamate bind directly to the lost receptors, we examined glutamate-dependent loss of receptor immunoreactivity in larvae with glutamate receptor ligand binding mutations. Our results suggest that glutamate-dependent receptor loss requires binding of glutamate directly to the lost receptors. To determine whether lost receptor protein is degraded or merely redistributed, we used immunoblots. Results suggest that glutamate receptor protein is redistributed, but not degraded, after prolonged exposure to high extracellular glutamate.
Collapse
Affiliation(s)
- Kaiyun Chen
- Department of Biological Sciences, University of Illinois at Chicago, 840 W. Taylor Street (MC 067), Chicago, IL 60607, USA
| | | | | |
Collapse
|
26
|
Liebl FLW, Wu Y, Featherstone DE, Noordermeer JN, Fradkin L, Hing H. Derailed regulates development of the Drosophila neuromuscular junction. Dev Neurobiol 2008; 68:152-65. [PMID: 17963254 PMCID: PMC2279185 DOI: 10.1002/dneu.20562] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neural function is dependent upon the proper formation and development of synapses. We show here that Wnt5 regulates the growth of the Drosophila neuromuscular junction (NMJ) by signaling through the Derailed receptor. Mutations in both wnt5 and drl result in a significant reduction in the number of synaptic boutons. Cell-type specific rescue experiments show that wnt5 functions in the presynaptic motor neuron while drl likely functions in the postsynaptic muscle cell. Epistatic analyses indicate that drl acts downstream of wnt5 to promote synaptic growth. Structure-function analyses of the Drl protein indicate that normal synaptic growth requires the extracellular Wnt inhibitory factor domain and the intracellular domain, which includes an atypical kinase. Our findings reveal a novel signaling mechanism that regulates morphology of the Drosophila NMJ.
Collapse
Affiliation(s)
- Faith L W Liebl
- Cell and Developmental Biology Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | | | | | | | |
Collapse
|
27
|
Drosophila fragile X mental retardation protein and metabotropic glutamate receptor A convergently regulate the synaptic ratio of ionotropic glutamate receptor subclasses. J Neurosci 2007; 27:12378-89. [PMID: 17989302 DOI: 10.1523/jneurosci.2970-07.2007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A current hypothesis proposes that fragile X mental retardation protein (FMRP), an RNA-binding translational regulator, acts downstream of glutamatergic transmission, via metabotropic glutamate receptor (mGluR) G(q)-dependent signaling, to modulate protein synthesis critical for trafficking ionotropic glutamate receptors (iGluRs) at synapses. However, direct evidence linking FMRP and mGluR function with iGluR synaptic expression is limited. In this study, we use the Drosophila fragile X model to test this hypothesis at the well characterized glutamatergic neuromuscular junction (NMJ). Two iGluR classes reside at this synapse, each containing common GluRIIC (III), IID and IIE subunits, and variable GluRIIA (A-class) or GluRIIB (B-class) subunits. In Drosophila fragile X mental retardation 1 (dfmr1) null mutants, A-class GluRs accumulate and B-class GluRs are lost, whereas total GluR levels do not change, resulting in a striking change in GluR subclass ratio at individual synapses. The sole Drosophila mGluR, DmGluRA, is also expressed at the NMJ. In dmGluRA null mutants, both iGluR classes increase, resulting in an increase in total synaptic GluR content at individual synapses. Targeted postsynaptic dmGluRA overexpression causes the exact opposite GluR phenotype to the dfmr1 null, confirming postsynaptic GluR subtype-specific regulation. In dfmr1; dmGluRA double null mutants, there is an additive increase in A-class GluRs, and a similar additive impact on B-class GluRs, toward normal levels in the double mutants. These results show that both dFMRP and DmGluRA differentially regulate the abundance of different GluR subclasses in a convergent mechanism within individual postsynaptic domains.
Collapse
|
28
|
Augustin H, Grosjean Y, Chen K, Sheng Q, Featherstone DE. Nonvesicular release of glutamate by glial xCT transporters suppresses glutamate receptor clustering in vivo. J Neurosci 2007; 27:111-23. [PMID: 17202478 PMCID: PMC2193629 DOI: 10.1523/jneurosci.4770-06.2007] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We hypothesized that cystine/glutamate transporters (xCTs) might be critical regulators of ambient extracellular glutamate levels in the nervous system and that misregulation of this glutamate pool might have important neurophysiological and/or behavioral consequences. To test this idea, we identified and functionally characterized a novel Drosophila xCT gene, which we subsequently named "genderblind" (gb). Genderblind is expressed in a previously overlooked subset of peripheral and central glia. Genetic elimination of gb causes a 50% reduction in extracellular glutamate concentration, demonstrating that xCT transporters are important regulators of extracellular glutamate. Consistent with previous studies showing that extracellular glutamate regulates postsynaptic glutamate receptor clustering, gb mutants show a large (200-300%) increase in the number of postsynaptic glutamate receptors. This increase in postsynaptic receptor abundance is not accompanied by other obvious synaptic changes and is completely rescued when synapses are cultured in wild-type levels of glutamate. Additional in situ pharmacology suggests that glutamate-mediated suppression of glutamate receptor clustering depends on receptor desensitization. Together, our results suggest that (1) xCT transporters are critical for regulation of ambient extracellular glutamate in vivo; (2) ambient extracellular glutamate maintains some receptors constitutively desensitized in vivo; and (3) constitutive desensitization of ionotropic glutamate receptors suppresses their ability to cluster at synapses.
Collapse
Affiliation(s)
- Hrvoje Augustin
- Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | | | | | | | | |
Collapse
|
29
|
Schwarz TL. Transmitter release at the neuromuscular junction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2006; 75:105-44. [PMID: 17137926 DOI: 10.1016/s0074-7742(06)75006-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Affiliation(s)
- Thomas L Schwarz
- Program in Neurobiology, Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
30
|
Liebl FLW, Werner KM, Sheng Q, Karr JE, McCabe BD, Featherstone DE. Genome-wide P-element screen for Drosophila synaptogenesis mutants. ACTA ACUST UNITED AC 2006; 66:332-47. [PMID: 16408305 PMCID: PMC1626350 DOI: 10.1002/neu.20229] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A molecular understanding of synaptogenesis is a critical step toward the goal of understanding how brains "wire themselves up," and then "rewire" during development and experience. Recent genomic and molecular advances have made it possible to study synaptogenesis on a genomic scale. Here, we describe the results of a screen for genes involved in formation and development of the glutamatergic Drosophila neuromuscular junction (NMJ). We screened 2185 P-element transposon mutants representing insertions in approximately 16% of the entire Drosophila genome. We first identified recessive lethal mutants, based on the hypothesis that mutations causing severe disruptions in synaptogenesis are likely to be lethal. Two hundred twenty (10%) of all insertions were homozygous lethal. Two hundred five (93%) of these lethal mutants developed at least through late embryogenesis and formed neuromusculature. We examined embryonic/larval NMJs in 202 of these homozygous mutants using immunocytochemistry and confocal microscopy. We identified and classified 88 mutants with altered NMJ morphology. Insertion loci in these mutants encode several different types of proteins, including ATP- and GTPases, cytoskeletal regulators, cell adhesion molecules, kinases, phosphatases, RNA regulators, regulators of protein formation, transcription factors, and transporters. Thirteen percent of insertions are in genes that encode proteins of novel or unknown function. Complementation tests and RT-PCR assays suggest that approximately 51% of the insertion lines carry background mutations. Our results reveal that synaptogenesis requires the coordinated action of many different types of proteins--perhaps as much as 44% of the entire genome--and that transposon mutageneses carry important caveats that must be respected when interpreting results generated using this method.
Collapse
Affiliation(s)
- Faith L W Liebl
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | | | | | | | | | | |
Collapse
|