1
|
Swaim GL, Glomb OV, Xie Y, Emerson C, Li Z, Beaudet D, Hendricks AG, Yogev S. Axonal Mechanotransduction Drives Cytoskeletal Responses to Physiological Mechanical Forces. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637689. [PMID: 39990487 PMCID: PMC11844441 DOI: 10.1101/2025.02.11.637689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Axons experience strong mechanical forces due to animal movement. While these forces serve as sensory cues in mechanosensory neurons, their impact on other neuron types remains poorly defined. Here, we uncover signaling that controls an axonal cytoskeletal response to external physiological forces and plays a key role in axonal integrity. Live imaging of microtubules at single-polymer resolution in a C. elegans motor neuron reveals local oscillatory movements that fine-tune polymer positioning. Combining cell-specific chemogenetic silencing with targeted degradation alleles to distinguish neuron-intrinsic from extrinsic regulators of these movements, we find that they are driven by muscle contractions and require the mechanosensitive protein Talin, the small GTPase RhoA, and actomyosin activity in the axon. Genetic perturbation of the axon's ability to buffer tension by disrupting the spectrin-based membrane-associated skeleton leads to RhoA hyperactivation, actomyosin relocalization to foci at microtubule ends, and converts local oscillations into processive bidirectional movements. This results in large gaps between microtubules, disrupting coverage of the axon and leading to its breakage and degeneration. Notably, hyperpolarizing muscle or degrading components of the mechanotransduction signaling pathway in the axon rescues cytoskeletal defects in spectrin-deficient axons. These results identify mechanisms of an axonal cytoskeletal response to physiological forces and highlight the importance of force-buffering and mechanotransduction signaling for axonal integrity.
Collapse
Affiliation(s)
- Grace L Swaim
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510
- These authors contributed equally
| | - Oliver V. Glomb
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- These authors contributed equally
| | - Yi Xie
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Chloe Emerson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510
| | - Zhuoyuan Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Daniel Beaudet
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9
| | - Adam G. Hendricks
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9
| | - Shaul Yogev
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
2
|
Mallik B, Frank CA. Mitochondrial Complex I and ROS control synapse function through opposing pre- and postsynaptic mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630694. [PMID: 39803545 PMCID: PMC11722341 DOI: 10.1101/2024.12.30.630694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Neurons require high amounts energy, and mitochondria help to fulfill this requirement. Dysfunctional mitochondria trigger problems in various neuronal tasks. Using the Drosophila neuromuscular junction (NMJ) as a model synapse, we previously reported that Mitochondrial Complex I (MCI) subunits were required for maintaining NMJ function and growth. Here we report tissue-specific adaptations at the NMJ when MCI is depleted. In Drosophila motor neurons, MCI depletion causes profound cytological defects and increased mitochondrial reactive oxygen species (ROS). But instead of diminishing synapse function, neuronal ROS triggers a homeostatic signaling process that maintains normal NMJ excitation. We identify molecules mediating this compensatory response. MCI depletion in muscles also enhances local ROS. But high levels of muscle ROS cause destructive responses: synapse degeneration, mitochondrial fragmentation, and impaired neurotransmission. In humans, mutations affecting MCI subunits cause severe neurological and neuromuscular diseases. The tissue-level effects that we describe in the Drosophila system are potentially relevant to forms of mitochondrial pathogenesis.
Collapse
Affiliation(s)
- Bhagaban Mallik
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| |
Collapse
|
3
|
Kim S, Shahab J, Vogelsang E, Wodarz A. Re-assessment of the subcellular localization of Bazooka/Par-3 in Drosophila: no evidence for localization to the nucleus and the neuromuscular junction. Biol Open 2024; 13:bio060544. [PMID: 38841912 PMCID: PMC11225583 DOI: 10.1242/bio.060544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024] Open
Abstract
Bazooka/Par-3 (Baz) is an evolutionarily conserved scaffold protein that functions as a master regulator for the establishment and maintenance of cell polarity in many different cell types. In the vast majority of published research papers Baz has been reported to localize at the cell cortex and at intercellular junctions. However, there have also been several reports showing localization and function of Baz at additional subcellular sites, in particular the nuclear envelope and the neuromuscular junction. In this study we have re-assessed the localization of Baz to these subcellular sites in a systematic manner. We used antibodies raised in different host animals against different epitopes of Baz for confocal imaging of Drosophila tissues. We tested the specificity of these antisera by mosaic analysis with null mutant baz alleles and tissue-specific RNAi against baz. In addition, we used a GFP-tagged gene trap line for Baz and a bacterial artificial chromosome (BAC) expressing GFP-tagged Baz under control of its endogenous promoter in a baz mutant background to compare the subcellular localization of the GFP-Baz fusion proteins to the staining with anti-Baz antisera. Together, these experiments did not provide evidence for specific localization of Baz to the nucleus or the neuromuscular junction.
Collapse
Affiliation(s)
- Soya Kim
- Molecular Cell Biology, Center for Anatomy, University of Cologne and University Hospital Cologne, Weyertal 115c, 50931 Köln, Germany
| | - Jaffer Shahab
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August-University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Elisabeth Vogelsang
- Molecular Cell Biology, Center for Anatomy, University of Cologne and University Hospital Cologne, Weyertal 115c, 50931 Köln, Germany
- Center for Molecular Medicine Cologne, University of Cologne and University Hospital Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Andreas Wodarz
- Molecular Cell Biology, Center for Anatomy, University of Cologne and University Hospital Cologne, Weyertal 115c, 50931 Köln, Germany
- Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August-University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
- Center for Molecular Medicine Cologne, University of Cologne and University Hospital Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Cluster of Excellence - Cellular stress response in aging-associated diseases (CECAD), University of Cologne and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
4
|
Sert O, Ding X, Zhang C, Mi R, Hoke A, Rasband MN. Postsynaptic β1 spectrin maintains Na + channels at the neuromuscular junction. J Physiol 2024; 602:1127-1145. [PMID: 38441922 PMCID: PMC10942750 DOI: 10.1113/jp285894] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/13/2024] [Indexed: 03/16/2024] Open
Abstract
Spectrins function together with actin as obligatory subunits of the submembranous cytoskeleton. Spectrins maintain cell shape, resist mechanical forces, and stabilize ion channel and transporter protein complexes through binding to scaffolding proteins. Recently, pathogenic variants of SPTBN4 (β4 spectrin) were reported to cause both neuropathy and myopathy. Although the role of β4 spectrin in neurons is mostly understood, its function in skeletal muscle, another excitable tissue subject to large forces, is unknown. Here, using a muscle specific β4 spectrin conditional knockout mouse, we show that β4 spectrin does not contribute to muscle function. In addition, we show β4 spectrin is not present in muscle, indicating the previously reported myopathy associated with pathogenic SPTBN4 variants is neurogenic in origin. More broadly, we show that α2, β1 and β2 spectrins are found in skeletal muscle, with α2 and β1 spectrins being enriched at the postsynaptic neuromuscular junction (NMJ). Surprisingly, using muscle specific conditional knockout mice, we show that loss of α2 and β2 spectrins had no effect on muscle health, function or the enrichment of β1 spectrin at the NMJ. Muscle specific deletion of β1 spectrin also had no effect on muscle health, but, with increasing age, resulted in the loss of clustered NMJ Na+ channels. Together, our results suggest that muscle β1 spectrin functions independently of an associated α spectrin to maintain Na+ channel clustering at the postsynaptic NMJ. Furthermore, despite repeated exposure to strong forces and in contrast to neurons, muscles do not require spectrin cytoskeletons to maintain cell shape or integrity. KEY POINTS: The myopathy found in pathogenic human SPTBN4 variants (where SPTBN4 is the gene encoding β4 spectrin) is neurogenic in origin. β1 spectrin plays essential roles in maintaining the density of neuromuscular junction Nav1.4 Na+ channels. By contrast to the canonical view of spectrin organization and function, we show that β1 spectrin can function independently of an associated α spectrin. Despite the large mechanical forces experienced by muscle, we show that spectrins are not required for muscle cell integrity. This is in stark contrast to red blood cells and the axons of neurons.
Collapse
Affiliation(s)
- Ozlem Sert
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Chuansheng Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| | - Ruifa Mi
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Ahmet Hoke
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA 77030
| |
Collapse
|
5
|
Sun Y, Zhao Y, Johnson TK, Xie W. Immunohistochemical Analysis of the Drosophila Larval Neuromuscular Junction. Methods Mol Biol 2024; 2746:201-211. [PMID: 38070091 DOI: 10.1007/978-1-0716-3585-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Synapses are specialized junctions between cells that mediate neurotransmission to modify brain activity and body function. Studies on synapse structure and function play an important role in understanding how neurons communicate and the consequences of their dysfunction in neurological disorders. The Drosophila larval neuromuscular junction is an excellent model for dissecting the cellular and molecular mechanisms of the synapse, with its large size, accessibility, and well-characterized genetics. This protocol describes the steps required for morphological and immunohistochemical analysis of the Drosophila larval neuromuscular junction including its dissection and multiplex labeling of synaptic proteins. This technique can be used to assess the impact of genetic manipulations on synaptic development, integrity, and plasticity, thus providing a valuable tool for probing complex neurological processes in a whole animal system.
Collapse
Affiliation(s)
- Yichen Sun
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
- School of Biological Sciences, Monash University, Clayton, VIC, Australia.
| | - Yu Zhao
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Chemistry, and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Wei Xie
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| |
Collapse
|
6
|
Cuentas-Condori A, Chen S, Krout M, Gallik KL, Tipps J, Gailey C, Flautt L, Kim H, Mulcahy B, Zhen M, Richmond JE, Miller DM. The epithelial Na + channel UNC-8 promotes an endocytic mechanism that recycles presynaptic components to new boutons in remodeling neurons. Cell Rep 2023; 42:113327. [PMID: 37906594 PMCID: PMC10921563 DOI: 10.1016/j.celrep.2023.113327] [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: 08/18/2022] [Revised: 06/01/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023] Open
Abstract
Circuit refinement involves the formation of new presynaptic boutons as others are dismantled. Nascent presynaptic sites can incorporate material from recently eliminated synapses, but the recycling mechanisms remain elusive. In early-stage C. elegans larvae, the presynaptic boutons of GABAergic DD neurons are removed and new outputs established at alternative sites. Here, we show that developmentally regulated expression of the epithelial Na+ channel (ENaC) UNC-8 in remodeling DD neurons promotes a Ca2+ and actin-dependent mechanism, involving activity-dependent bulk endocytosis (ADBE), that recycles presynaptic material for reassembly at nascent DD synapses. ADBE normally functions in highly active neurons to accelerate local recycling of synaptic vesicles. In contrast, we find that an ADBE-like mechanism results in the distal recycling of synaptic material from old to new synapses. Thus, our findings suggest that a native mechanism (ADBE) can be repurposed to dismantle presynaptic terminals for reassembly at new, distant locations.
Collapse
Affiliation(s)
- Andrea Cuentas-Condori
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Siqi Chen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Mia Krout
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kristin L Gallik
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - John Tipps
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Casey Gailey
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Leah Flautt
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Hongkyun Kim
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Ben Mulcahy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Mei Zhen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - David M Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA; Neurosience Program, Vanderbilt University, Nashville, TN 37240, USA.
| |
Collapse
|
7
|
Glomb O, Swaim G, Munoz LLancao P, Lovejoy C, Sutradhar S, Park J, Wu Y, Cason SE, Holzbaur ELF, Hammarlund M, Howard J, Ferguson SM, Gramlich MW, Yogev S. A kinesin-1 adaptor complex controls bimodal slow axonal transport of spectrin in Caenorhabditis elegans. Dev Cell 2023; 58:1847-1863.e12. [PMID: 37751746 PMCID: PMC10574138 DOI: 10.1016/j.devcel.2023.08.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/18/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
An actin-spectrin lattice, the membrane periodic skeleton (MPS), protects axons from breakage. MPS integrity relies on spectrin delivery via slow axonal transport, a process that remains poorly understood. We designed a probe to visualize endogenous spectrin dynamics at single-axon resolution in vivo. Surprisingly, spectrin transport is bimodal, comprising fast runs and movements that are 100-fold slower than previously reported. Modeling and genetic analysis suggest that the two rates are independent, yet both require kinesin-1 and the coiled-coil proteins UNC-76/FEZ1 and UNC-69/SCOC, which we identify as spectrin-kinesin adaptors. Knockdown of either protein led to disrupted spectrin motility and reduced distal MPS, and UNC-76 overexpression instructed excessive transport of spectrin. Artificially linking spectrin to kinesin-1 drove robust motility but inefficient MPS assembly, whereas impairing MPS assembly led to excessive spectrin transport, suggesting a balance between transport and assembly. These results provide insight into slow axonal transport and MPS integrity.
Collapse
Affiliation(s)
- Oliver Glomb
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Grace Swaim
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Pablo Munoz LLancao
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christopher Lovejoy
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Junhyun Park
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Youjun Wu
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sydney E Cason
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marc Hammarlund
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Quantitative Biology Institute, Yale University, New Haven, CT 06510, USA
| | - Shawn M Ferguson
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Shaul Yogev
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
8
|
Guss EJ, Akbergenova Y, Cunningham KL, Littleton JT. Loss of the extracellular matrix protein Perlecan disrupts axonal and synaptic stability during Drosophila development. eLife 2023; 12:RP88273. [PMID: 37368474 PMCID: PMC10328508 DOI: 10.7554/elife.88273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) form essential components of the extracellular matrix (ECM) and basement membrane (BM) and have both structural and signaling roles. Perlecan is a secreted ECM-localized HSPG that contributes to tissue integrity and cell-cell communication. Although a core component of the ECM, the role of Perlecan in neuronal structure and function is less understood. Here, we identify a role for Drosophila Perlecan in the maintenance of larval motoneuron axonal and synaptic stability. Loss of Perlecan causes alterations in the axonal cytoskeleton, followed by axonal breakage and synaptic retraction of neuromuscular junctions. These phenotypes are not prevented by blocking Wallerian degeneration and are independent of Perlecan's role in Wingless signaling. Expression of Perlecan solely in motoneurons cannot rescue synaptic retraction phenotypes. Similarly, removing Perlecan specifically from neurons, glia, or muscle does not cause synaptic retraction, indicating the protein is secreted from multiple cell types and functions non-cell autonomously. Within the peripheral nervous system, Perlecan predominantly localizes to the neural lamella, a specialized ECM surrounding nerve bundles. Indeed, the neural lamella is disrupted in the absence of Perlecan, with axons occasionally exiting their usual boundary in the nerve bundle. In addition, entire nerve bundles degenerate in a temporally coordinated manner across individual hemi-segments throughout larval development. These observations indicate disruption of neural lamella ECM function triggers axonal destabilization and synaptic retraction of motoneurons, revealing a role for Perlecan in axonal and synaptic integrity during nervous system development.
Collapse
Affiliation(s)
- Ellen J Guss
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Karen L Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
9
|
Mushtaq Z, Aavula K, Lasser DA, Kieweg ID, Lion LM, Kins S, Pielage J. Madm/NRBP1 mediates synaptic maintenance and neurodegeneration-induced presynaptic homeostatic potentiation. Cell Rep 2022; 41:111710. [PMID: 36450258 DOI: 10.1016/j.celrep.2022.111710] [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: 08/31/2022] [Revised: 10/05/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
The precise regulation of synaptic connectivity and function is essential to maintain neuronal circuits. Here, we show that the Drosophila pseudo-kinase Madm/NRBP1 (Mlf-1-adapter-molecule/nuclear-receptor-binding protein 1) is required presynaptically to maintain synaptic stability and to coordinate synaptic growth and function. Presynaptic Madm mediates these functions by controlling cap-dependent translation via the target of rapamycin (TOR) effector 4E-BP/Thor (eukaryotic initiation factor 4E binding protein/Thor). Strikingly, at degenerating neuromuscular synapses, postsynaptic Madm induces a compensatory, transsynaptic signal that utilizes the presynaptic homeostatic potentiation (PHP) machinery to offset synaptic release deficits and to delay synaptic degeneration. Madm is not required for canonical PHP but induces a neurodegeneration-specific form of PHP and acts via the regulation of the cap-dependent translation regulators 4E-BP/Thor and S6-kinase. Consistently, postsynaptic induction of canonical PHP or TOR activation can compensate for postsynaptic Madm to alleviate functional and structural synaptic defects. Our results provide insights into the molecular mechanisms underlying neurodegeneration-induced PHP with potential neurotherapeutic applications.
Collapse
Affiliation(s)
- Zeeshan Mushtaq
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Kumar Aavula
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany; Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| | - Dario A Lasser
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Ingrid D Kieweg
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Lena M Lion
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Jan Pielage
- Department of Zoology and Neurobiology, University of Kaiserslautern, 67663 Kaiserslautern, Germany.
| |
Collapse
|
10
|
Orr BO, Fetter RD, Davis GW. Activation and expansion of presynaptic signaling foci drives presynaptic homeostatic plasticity. Neuron 2022; 110:3743-3759.e6. [PMID: 36087584 PMCID: PMC9671843 DOI: 10.1016/j.neuron.2022.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 06/07/2022] [Accepted: 08/11/2022] [Indexed: 12/15/2022]
Abstract
Presynaptic homeostatic plasticity (PHP) adaptively regulates synaptic transmission in health and disease. Despite identification of numerous genes that are essential for PHP, we lack a dynamic framework to explain how PHP is initiated, potentiated, and limited to achieve precise control of vesicle fusion. Here, utilizing both mice and Drosophila, we demonstrate that PHP progresses through the assembly and physical expansion of presynaptic signaling foci where activated integrins biochemically converge with trans-synaptic Semaphorin2b/PlexinB signaling. Each component of the identified signaling complexes, including alpha/beta-integrin, Semaphorin2b, PlexinB, talin, and focal adhesion kinase (FAK), and their biochemical interactions, are essential for PHP. Complex integrity requires the Sema2b ligand and complex expansion includes a ∼2.5-fold expansion of active-zone associated puncta composed of the actin-binding protein talin. Finally, complex pre-expansion is sufficient to accelerate the rate and extent of PHP. A working model is proposed incorporating signal convergence with dynamic molecular assemblies that instruct PHP.
Collapse
Affiliation(s)
- Brian O Orr
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158 USA.
| |
Collapse
|
11
|
Han Y, Chien C, Goel P, He K, Pinales C, Buser C, Dickman D. Botulinum neurotoxin accurately separates tonic vs. phasic transmission and reveals heterosynaptic plasticity rules in Drosophila. eLife 2022; 11:e77924. [PMID: 35993544 PMCID: PMC9439677 DOI: 10.7554/elife.77924] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/20/2022] [Indexed: 11/13/2022] Open
Abstract
In developing and mature nervous systems, diverse neuronal subtypes innervate common targets to establish, maintain, and modify neural circuit function. A major challenge towards understanding the structural and functional architecture of neural circuits is to separate these inputs and determine their intrinsic and heterosynaptic relationships. The Drosophila larval neuromuscular junction is a powerful model system to study these questions, where two glutamatergic motor neurons, the strong phasic-like Is and weak tonic-like Ib, co-innervate individual muscle targets to coordinate locomotor behavior. However, complete neurotransmission from each input has never been electrophysiologically separated. We have employed a botulinum neurotoxin, BoNT-C, that eliminates both spontaneous and evoked neurotransmission without perturbing synaptic growth or structure, enabling the first approach that accurately isolates input-specific neurotransmission. Selective expression of BoNT-C in Is or Ib motor neurons disambiguates the functional properties of each input. Importantly, the blended values of Is+Ib neurotransmission can be fully recapitulated by isolated physiology from each input. Finally, selective silencing by BoNT-C does not induce heterosynaptic structural or functional plasticity at the convergent input. Thus, BoNT-C establishes the first approach to accurately separate neurotransmission between tonic vs. phasic neurons and defines heterosynaptic plasticity rules in a powerful model glutamatergic circuit.
Collapse
Affiliation(s)
- Yifu Han
- Department of Neurobiology, University of Southern CaliforniaLos AngelesUnited States
| | - Chun Chien
- Department of Neurobiology, University of Southern CaliforniaLos AngelesUnited States
| | - Pragya Goel
- Department of Neurobiology, University of Southern CaliforniaLos AngelesUnited States
| | - Kaikai He
- Department of Neurobiology, University of Southern CaliforniaLos AngelesUnited States
| | | | | | - Dion Dickman
- Department of Neurobiology, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
12
|
Li H, Datunashvili M, Reyes RC, Voglmaier SM. Inositol hexakisphosphate kinases differentially regulate trafficking of vesicular glutamate transporters 1 and 2. Front Cell Neurosci 2022; 16:926794. [PMID: 35936490 PMCID: PMC9355605 DOI: 10.3389/fncel.2022.926794] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Inositol pyrophosphates have been implicated in cellular signaling and membrane trafficking, including synaptic vesicle (SV) recycling. Inositol hexakisphosphate kinases (IP6Ks) and their product, diphosphoinositol pentakisphosphate (PP-IP5 or IP7), directly and indirectly regulate proteins important in vesicle recycling by the activity-dependent bulk endocytosis pathway (ADBE). In the present study, we show that two isoforms, IP6K1 and IP6K3, are expressed in axons. The role of the kinases in SV recycling are investigated using pharmacologic inhibition, shRNA knockdown, and IP6K1 and IP6K3 knockout mice. Live-cell imaging experiments use optical reporters of SV recycling based on vesicular glutamate transporter isoforms, VGLUT1- and VGLUT2-pHluorins (pH), which recycle differently. VGLUT1-pH recycles by classical AP-2 dependent endocytosis under moderate stimulation conditions, while VGLUT2-pH recycles using AP-1 and AP-3 adaptor proteins as well. Using a short stimulus to release the readily releasable pool (RRP), we show that IP6K1 KO increases exocytosis of both VGLUT1-and VGLUT2-pH, while IP6K3 KO decreases the amount of both transporters in the RRP. In electrophysiological experiments we measure glutamate signaling with short stimuli and under the intense stimulation conditions that trigger bulk endocytosis. IP6K1 KO increases synaptic facilitation and IP6K3 KO decreases facilitation compared to wild type in CA1 hippocampal Schaffer collateral synapses. After intense stimulation, the rate of endocytosis of VGLUT2-pH, but not VGLUT1-pH, is increased by knockout, knockdown, and pharmacologic inhibition of IP6Ks. Thus IP6Ks differentially affect the endocytosis of two SV protein cargos that use different endocytic pathways. However, while IP6K1 KO and IP6K3 KO exert similar effects on endocytosis after stimulation, the isoforms exert different effects on exocytosis earlier in the stimulus and on the early phase of glutamate release. Taken together, the data indicate a role for IP6Ks both in exocytosis early in the stimulation period and in endocytosis, particularly under conditions that may utilize AP-1/3 adaptors.
Collapse
|
13
|
Duhart JC, Mosca TJ. Genetic regulation of central synapse formation and organization in Drosophila melanogaster. Genetics 2022; 221:6597078. [PMID: 35652253 DOI: 10.1093/genetics/iyac078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/29/2022] [Indexed: 01/04/2023] Open
Abstract
A goal of modern neuroscience involves understanding how connections in the brain form and function. Such a knowledge is essential to inform how defects in the exquisite complexity of nervous system growth influence neurological disease. Studies of the nervous system in the fruit fly Drosophila melanogaster enabled the discovery of a wealth of molecular and genetic mechanisms underlying development of synapses-the specialized cell-to-cell connections that comprise the essential substrate for information flow and processing in the nervous system. For years, the major driver of knowledge was the neuromuscular junction due to its ease of examination. Analogous studies in the central nervous system lagged due to a lack of genetic accessibility of specific neuron classes, synaptic labels compatible with cell-type-specific access, and high resolution, quantitative imaging strategies. However, understanding how central synapses form remains a prerequisite to understanding brain development. In the last decade, a host of new tools and techniques extended genetic studies of synapse organization into central circuits to enhance our understanding of synapse formation, organization, and maturation. In this review, we consider the current state-of-the-field. We first discuss the tools, technologies, and strategies developed to visualize and quantify synapses in vivo in genetically identifiable neurons of the Drosophila central nervous system. Second, we explore how these tools enabled a clearer understanding of synaptic development and organization in the fly brain and the underlying molecular mechanisms of synapse formation. These studies establish the fly as a powerful in vivo genetic model that offers novel insights into neural development.
Collapse
Affiliation(s)
- Juan Carlos Duhart
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Timothy J Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
14
|
Restrepo LJ, DePew AT, Moese ER, Tymanskyj SR, Parisi MJ, Aimino MA, Duhart JC, Fei H, Mosca TJ. γ-secretase promotes Drosophila postsynaptic development through the cleavage of a Wnt receptor. Dev Cell 2022; 57:1643-1660.e7. [PMID: 35654038 DOI: 10.1016/j.devcel.2022.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 04/06/2022] [Accepted: 05/10/2022] [Indexed: 12/27/2022]
Abstract
Developing synapses mature through the recruitment of specific proteins that stabilize presynaptic and postsynaptic structure and function. Wnt ligands signaling via Frizzled (Fz) receptors play many crucial roles in neuronal and synaptic development, but whether and how Wnt and Fz influence synaptic maturation is incompletely understood. Here, we show that Fz2 receptor cleavage via the γ-secretase complex is required for postsynaptic development and maturation. In the absence of γ-secretase, Drosophila neuromuscular synapses fail to recruit postsynaptic scaffolding and cytoskeletal proteins, leading to behavioral deficits. Introducing presenilin mutations linked to familial early-onset Alzheimer's disease into flies leads to synaptic maturation phenotypes that are identical to those seen in null alleles. This conserved role for γ-secretase in synaptic maturation and postsynaptic development highlights the importance of Fz2 cleavage and suggests that receptor processing by proteins linked to neurodegeneration may be a shared mechanism with aspects of synaptic development.
Collapse
Affiliation(s)
- Lucas J Restrepo
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Alison T DePew
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Elizabeth R Moese
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Stephen R Tymanskyj
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael J Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael A Aimino
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Juan Carlos Duhart
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Hong Fei
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Timothy J Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA.
| |
Collapse
|
15
|
Weber JJ, Haas E, Maringer Y, Hauser S, Casadei NLP, Chishti AH, Riess O, Hübener-Schmid J. Calpain-1 ablation partially rescues disease-associated hallmarks in models of Machado-Joseph disease. Hum Mol Genet 2021; 29:892-906. [PMID: 31960910 DOI: 10.1093/hmg/ddaa010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
Proteolytic fragmentation of polyglutamine-expanded ataxin-3 is a concomitant and modifier of the molecular pathogenesis of Machado-Joseph disease (MJD), the most common autosomal dominant cerebellar ataxia. Calpains, a group of calcium-dependent cysteine proteases, are important mediators of ataxin-3 cleavage and implicated in multiple neurodegenerative conditions. Pharmacologic and genetic approaches lowering calpain activity showed beneficial effects on molecular and behavioural disease characteristics in MJD model organisms. However, specifically targeting one of the calpain isoforms by genetic means has not yet been evaluated as a potential therapeutic strategy. In our study, we tested whether calpains are overactivated in the MJD context and if reduction or ablation of calpain-1 expression ameliorates the disease-associated phenotype in MJD cells and mice. In all analysed MJD models, we detected an elevated calpain activity at baseline. Lowering or removal of calpain-1 in cells or mice counteracted calpain system overactivation and led to reduced cleavage of ataxin-3 without affecting its aggregation. Moreover, calpain-1 knockout in YAC84Q mice alleviated excessive fragmentation of important synaptic proteins. Despite worsening some motor characteristics, YAC84Q mice showed a rescue of body weight loss and extended survival upon calpain-1 knockout. Together, our findings emphasize the general potential of calpains as a therapeutic target in MJD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonasz J Weber
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany.,Department of Human Genetics, Ruhr-University Bochum, Bochum 44801, Germany
| | - Eva Haas
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany
| | - Yacine Maringer
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany
| | - Stefan Hauser
- German Center for Neurodegenerative Diseases, Tübingen 72076, Germany
| | - Nicolas L P Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany
| | - Athar H Chishti
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany
| | - Jeannette Hübener-Schmid
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen 72076, Germany.,Centre for Rare Diseases, University of Tübingen, Tübingen 72076, Germany
| |
Collapse
|
16
|
Johnson AE, Orr BO, Fetter RD, Moughamian AJ, Primeaux LA, Geier EG, Yokoyama JS, Miller BL, Davis GW. SVIP is a molecular determinant of lysosomal dynamic stability, neurodegeneration and lifespan. Nat Commun 2021; 12:513. [PMID: 33479240 PMCID: PMC7820495 DOI: 10.1038/s41467-020-20796-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Missense mutations in Valosin-Containing Protein (VCP) are linked to diverse degenerative diseases including IBMPFD, amyotrophic lateral sclerosis (ALS), muscular dystrophy and Parkinson's disease. Here, we characterize a VCP-binding co-factor (SVIP) that specifically recruits VCP to lysosomes. SVIP is essential for lysosomal dynamic stability and autophagosomal-lysosomal fusion. SVIP mutations cause muscle wasting and neuromuscular degeneration while muscle-specific SVIP over-expression increases lysosomal abundance and is sufficient to extend lifespan in a context, stress-dependent manner. We also establish multiple links between SVIP and VCP-dependent disease in our Drosophila model system. A biochemical screen identifies a disease-causing VCP mutation that prevents SVIP binding. Conversely, over-expression of an SVIP mutation that prevents VCP binding is deleterious. Finally, we identify a human SVIP mutation and confirm the pathogenicity of this mutation in our Drosophila model. We propose a model for VCP disease based on the differential, co-factor-dependent recruitment of VCP to intracellular organelles.
Collapse
Affiliation(s)
- Alyssa E Johnson
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Brian O Orr
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Armen J Moughamian
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Logan A Primeaux
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Ethan G Geier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
17
|
Yang P, Yang Y, Sun P, Tian Y, Gao F, Wang C, Zong T, Li M, Zhang Y, Yu T, Jiang Z. βII spectrin (SPTBN1): biological function and clinical potential in cancer and other diseases. Int J Biol Sci 2021; 17:32-49. [PMID: 33390831 PMCID: PMC7757025 DOI: 10.7150/ijbs.52375] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022] Open
Abstract
βII spectrin, the most common isoform of non-erythrocyte spectrin, is a cytoskeleton protein present in all nucleated cells. Interestingly, βII spectrin is essential for the development of various organs such as nerve, epithelium, inner ear, liver and heart. The functions of βII spectrin include not only establishing and maintaining the cell structure but also regulating a variety of cellular functions, such as cell apoptosis, cell adhesion, cell spreading and cell cycle regulation. Notably, βII spectrin dysfunction is associated with embryonic lethality and the DNA damage response. More recently, the detection of altered βII spectrin expression in tumors indicated that βII spectrin might be involved in the development and progression of cancer. Its mutations and disorders could result in developmental disabilities and various diseases. The versatile roles of βII spectrin in disease have been examined in an increasing number of studies; nonetheless, the exact mechanisms of βII spectrin are still poorly understood. Thus, we summarize the structural features and biological roles of βII spectrin and discuss its molecular mechanisms and functions in development, homeostasis, regeneration and differentiation. This review highlight the potential effects of βII spectrin dysfunction in cancer and other diseases, outstanding questions for the future investigation of therapeutic targets. The investigation of the regulatory mechanism of βII spectrin signal inactivation and recovery may bring hope for future therapy of related diseases.
Collapse
Affiliation(s)
- Panyu Yang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People's Republic of China
| | - Pin Sun
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Yu Tian
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Fang Gao
- Department of Physical Medicine and Rehabiliation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chen Wang
- Department of Physical Medicine and Rehabiliation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Tingyu Zong
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Min Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Ying Zhang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.,Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China
| | - Zhirong Jiang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
18
|
Chou VT, Johnson SA, Van Vactor D. Synapse development and maturation at the drosophila neuromuscular junction. Neural Dev 2020; 15:11. [PMID: 32741370 PMCID: PMC7397595 DOI: 10.1186/s13064-020-00147-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Synapses are the sites of neuron-to-neuron communication and form the basis of the neural circuits that underlie all animal cognition and behavior. Chemical synapses are specialized asymmetric junctions between a presynaptic neuron and a postsynaptic target that form through a series of diverse cellular and subcellular events under the control of complex signaling networks. Once established, the synapse facilitates neurotransmission by mediating the organization and fusion of synaptic vesicles and must also retain the ability to undergo plastic changes. In recent years, synaptic genes have been implicated in a wide array of neurodevelopmental disorders; the individual and societal burdens imposed by these disorders, as well as the lack of effective therapies, motivates continued work on fundamental synapse biology. The properties and functions of the nervous system are remarkably conserved across animal phyla, and many insights into the synapses of the vertebrate central nervous system have been derived from studies of invertebrate models. A prominent model synapse is the Drosophila melanogaster larval neuromuscular junction, which bears striking similarities to the glutamatergic synapses of the vertebrate brain and spine; further advantages include the simplicity and experimental versatility of the fly, as well as its century-long history as a model organism. Here, we survey findings on the major events in synaptogenesis, including target specification, morphogenesis, and the assembly and maturation of synaptic specializations, with a emphasis on work conducted at the Drosophila neuromuscular junction.
Collapse
Affiliation(s)
- Vivian T Chou
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Seth A Johnson
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
19
|
Beijer D, Deconinck T, De Bleecker JL, Dotti MT, Malandrini A, Urtizberea JA, Zulaica M, López de Munain A, Asselbergh B, De Jonghe P, Baets J. Nonsense mutations in alpha-II spectrin in three families with juvenile onset hereditary motor neuropathy. Brain 2020; 142:2605-2616. [PMID: 31332438 DOI: 10.1093/brain/awz216] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/25/2019] [Accepted: 05/28/2019] [Indexed: 01/09/2023] Open
Abstract
Distal hereditary motor neuropathies are a rare subgroup of inherited peripheral neuropathies hallmarked by a length-dependent axonal degeneration of lower motor neurons without significant involvement of sensory neurons. We identified patients with heterozygous nonsense mutations in the αII-spectrin gene, SPTAN1, in three separate dominant hereditary motor neuropathy families via next-generation sequencing. Variable penetrance was noted for these mutations in two of three families, and phenotype severity differs greatly between patients. The mutant mRNA containing nonsense mutations is broken down by nonsense-mediated decay and leads to reduced protein levels in patient cells. Previously, dominant-negative αII-spectrin gene mutations were described as causal in a spectrum of epilepsy phenotypes.
Collapse
Affiliation(s)
- Danique Beijer
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium
| | - Tine Deconinck
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium
| | | | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neuroscience, University of Siena, Italy
| | | | | | - Miren Zulaica
- Neuroscience Area, Institute Biodonostia, Hospital Universitario Donostia, San Sebastian, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, Madrid, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Institute Biodonostia, Hospital Universitario Donostia, San Sebastian, Spain.,Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, Madrid, Spain
| | - Bob Asselbergh
- VIB-UAntwerp Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Peter De Jonghe
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Belgium
| | - Jonathan Baets
- Neurogenetics Group, Center for Molecular Neurology, University of Antwerp, Belgium.,Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Belgium
| |
Collapse
|
20
|
Presynaptic Homeostasis Opposes Disease Progression in Mouse Models of ALS-Like Degeneration: Evidence for Homeostatic Neuroprotection. Neuron 2020; 107:95-111.e6. [PMID: 32380032 DOI: 10.1016/j.neuron.2020.04.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Progressive synapse loss is an inevitable and insidious part of age-related neurodegenerative disease. Typically, synapse loss precedes symptoms of cognitive and motor decline. This suggests the existence of compensatory mechanisms that can temporarily counteract the effects of ongoing neurodegeneration. Here, we demonstrate that presynaptic homeostatic plasticity (PHP) is induced at degenerating neuromuscular junctions, mediated by an evolutionarily conserved activity of presynaptic ENaC channels in both Drosophila and mouse. To assess the consequence of eliminating PHP in a mouse model of ALS-like degeneration, we generated a motoneuron-specific deletion of Scnn1a, encoding the ENaC channel alpha subunit. We show that Scnn1a is essential for PHP without adversely affecting baseline neural function or lifespan. However, Scnn1a knockout in a degeneration-causing mutant background accelerated motoneuron loss and disease progression to twice the rate observed in littermate controls with intact PHP. We propose a model of neuroprotective homeostatic plasticity, extending organismal lifespan and health span.
Collapse
|
21
|
Weber T, Stephan R, Moreno E, Pielage J. The Ankyrin Repeat Domain Controls Presynaptic Localization of Drosophila Ankyrin2 and Is Essential for Synaptic Stability. Front Cell Dev Biol 2019; 7:148. [PMID: 31475145 PMCID: PMC6703079 DOI: 10.3389/fcell.2019.00148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/16/2019] [Indexed: 01/24/2023] Open
Abstract
The structural integrity of synaptic connections critically depends on the interaction between synaptic cell adhesion molecules (CAMs) and the underlying actin and microtubule cytoskeleton. This interaction is mediated by giant Ankyrins, that act as specialized adaptors to establish and maintain axonal and synaptic compartments. In Drosophila, two giant isoforms of Ankyrin2 (Ank2) control synapse stability and organization at the larval neuromuscular junction (NMJ). Both Ank2-L and Ank2-XL are highly abundant in motoneuron axons and within the presynaptic terminal, where they control synaptic CAMs distribution and organization of microtubules. Here, we address the role of the conserved N-terminal ankyrin repeat domain (ARD) for subcellular localization and function of these giant Ankyrins in vivo. We used a P[acman] based rescue approach to generate deletions of ARD subdomains, that contain putative binding sites of interacting transmembrane proteins. We show that specific subdomains control synaptic but not axonal localization of Ank2-L. These domains contain binding sites to L1-family member CAMs, and we demonstrate that these regions are necessary for the organization of synaptic CAMs and for the control of synaptic stability. In contrast, presynaptic Ank2-XL localization only partially depends on the ARD but strictly requires the presynaptic presence of Ank2-L demonstrating a critical co-dependence of the two isoforms at the NMJ. Ank2-XL dependent control of microtubule organization correlates with presynaptic abundance of the protein and is thus only partially affected by ARD deletions. Together, our data provides novel insights into the synaptic targeting of giant Ankyrins with relevance for the control of synaptic plasticity and maintenance.
Collapse
Affiliation(s)
- Tobias Weber
- Department of Zoology and Neurobiology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Raiko Stephan
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Eliza Moreno
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jan Pielage
- Department of Zoology and Neurobiology, University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
22
|
Wang G, Simon DJ, Wu Z, Belsky DM, Heller E, O'Rourke MK, Hertz NT, Molina H, Zhong G, Tessier-Lavigne M, Zhuang X. Structural plasticity of actin-spectrin membrane skeleton and functional role of actin and spectrin in axon degeneration. eLife 2019; 8:e38730. [PMID: 31042147 PMCID: PMC6494423 DOI: 10.7554/elife.38730] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 03/30/2019] [Indexed: 01/08/2023] Open
Abstract
Axon degeneration sculpts neuronal connectivity patterns during development and is an early hallmark of several adult-onset neurodegenerative disorders. Substantial progress has been made in identifying effector mechanisms driving axon fragmentation, but less is known about the upstream signaling pathways that initiate this process. Here, we investigate the behavior of the actin-spectrin-based Membrane-associated Periodic Skeleton (MPS), and effects of actin and spectrin manipulations in sensory axon degeneration. We show that trophic deprivation (TD) of mouse sensory neurons causes a rapid disassembly of the axonal MPS, which occurs prior to protein loss and independently of caspase activation. Actin destabilization initiates TD-related retrograde signaling needed for degeneration; actin stabilization prevents MPS disassembly and retrograde signaling during TD. Depletion of βII-spectrin, a key component of the MPS, suppresses retrograde signaling and protects axons against degeneration. These data demonstrate structural plasticity of the MPS and suggest its potential role in early steps of axon degeneration.
Collapse
Affiliation(s)
- Guiping Wang
- Department of Chemistry and Chemical BiologyHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
- Department of PhysicsHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
| | - David J Simon
- Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
- Department of BiologyStanford UniversityStanfordUnited States
| | - Zhuhao Wu
- Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
| | - Deanna M Belsky
- Department of BiologyStanford UniversityStanfordUnited States
| | - Evan Heller
- Department of Chemistry and Chemical BiologyHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
- Department of PhysicsHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
| | | | - Nicholas T Hertz
- Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
- Department of BiologyStanford UniversityStanfordUnited States
| | - Henrik Molina
- Proteomics Resource CenterThe Rockefeller UniversityNew YorkUnited States
| | - Guisheng Zhong
- Department of Chemistry and Chemical BiologyHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
- Department of PhysicsHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and RepairThe Rockefeller UniversityNew YorkUnited States
- Department of BiologyStanford UniversityStanfordUnited States
| | - Xiaowei Zhuang
- Department of Chemistry and Chemical BiologyHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
- Department of PhysicsHoward Hughes Medical Institute, Harvard UniversityCambridgeUnited States
| |
Collapse
|
23
|
A Screen for Synaptic Growth Mutants Reveals Mechanisms That Stabilize Synaptic Strength. J Neurosci 2019; 39:4051-4065. [PMID: 30902873 DOI: 10.1523/jneurosci.2601-18.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 03/12/2019] [Accepted: 03/14/2019] [Indexed: 01/28/2023] Open
Abstract
Synapses grow, prune, and remodel throughout development, experience, and disease. This structural plasticity can destabilize information transfer in the nervous system. However, neural activity remains stable throughout life, implying that adaptive countermeasures exist that maintain neurotransmission within proper physiological ranges. Aberrant synaptic structure and function have been associated with a variety of neural diseases, including Fragile X syndrome, autism, and intellectual disability. We have screened 300 mutants in Drosophila larvae of both sexes for defects in synaptic growth at the neuromuscular junction, identifying 12 mutants with severe reductions or enhancements in synaptic growth. Remarkably, electrophysiological recordings revealed that synaptic strength was unchanged in all but one of these mutants compared with WT. We used a combination of genetic, anatomical, and electrophysiological analyses to illuminate three mechanisms that stabilize synaptic strength despite major disparities in synaptic growth. These include compensatory changes in (1) postsynaptic neurotransmitter receptor abundance, (2) presynaptic morphology, and (3) active zone structure. Together, this characterization identifies new mutants with defects in synaptic growth and the adaptive strategies used by synapses to homeostatically stabilize neurotransmission in response.SIGNIFICANCE STATEMENT This study reveals compensatory mechanisms used by synapses to ensure stable functionality during severe alterations in synaptic growth using the neuromuscular junction of Drosophila melanogaster as a model system. Through a forward genetic screen, we identify mutants that exhibit dramatic undergrown or overgrown synapses yet express stable levels of synaptic strength, with three specific compensatory mechanisms discovered. Thus, this study reveals novel insights into the adaptive strategies that constrain neurotransmission within narrow physiological ranges while allowing considerable flexibility in overall synapse number. More broadly, these findings provide insights into how stable synaptic function may be maintained in the nervous system during periods of intensive synaptic growth, pruning, and remodeling.
Collapse
|
24
|
DePew AT, Aimino MA, Mosca TJ. The Tenets of Teneurin: Conserved Mechanisms Regulate Diverse Developmental Processes in the Drosophila Nervous System. Front Neurosci 2019; 13:27. [PMID: 30760977 PMCID: PMC6363694 DOI: 10.3389/fnins.2019.00027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/11/2019] [Indexed: 01/26/2023] Open
Abstract
To successfully integrate a neuron into a circuit, a myriad of developmental events must occur correctly and in the correct order. Neurons must be born and grow out toward a destination, responding to guidance cues to direct their path. Once arrived, each neuron must segregate to the correct sub-region before sorting through a milieu of incorrect partners to identify the correct partner with which they can connect. Finally, the neuron must make a synaptic connection with their correct partner; a connection that needs to be broadly maintained throughout the life of the animal while remaining responsive to modes of plasticity and pruning. Though many intricate molecular mechanisms have been discovered to regulate each step, recent work showed that a single family of proteins, the Teneurins, regulates a host of these developmental steps in Drosophila – an example of biological adaptive reuse. Teneurins first influence axon guidance during early development. Once neurons arrive in their target regions, Teneurins enable partner matching and synapse formation in both the central and peripheral nervous systems. Despite these diverse processes and systems, the Teneurins use conserved mechanisms to achieve these goals, as defined by three tenets: (1) transsynaptic interactions with each other, (2) membrane stabilization via an interaction with and regulation of the cytoskeleton, and (3) a role for presynaptic Ten-a in regulating synaptic function. These processes are further distinguished by (1) the nature of the transsynaptic interaction – homophilic interactions (between the same Teneurins) to engage partner matching and heterophilic interactions (between different Teneurins) to enable synaptic connectivity and the proper apposition of pre- and postsynaptic sites and (2) the location of cytoskeletal regulation (presynaptic cytoskeletal regulation in the CNS and postsynaptic regulation of the cytoskeleton at the NMJ). Thus, both the roles and the mechanisms governing them are conserved across processes and synapses. Here, we will highlight the contributions of Drosophila synaptic biology to our understanding of the Teneurins, discuss the mechanistic conservation that allows the Teneurins to achieve common neurodevelopmental goals, and present new data in support of these points. Finally, we will posit the next steps for understanding how this remarkably versatile family of proteins functions to control multiple distinct events in the creation of a nervous system.
Collapse
Affiliation(s)
- Alison T DePew
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Michael A Aimino
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| | - Timothy J Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
25
|
Borgen MA, Giles AC, Wang D, Grill B. Synapse maintenance is impacted by ATAT-2 tubulin acetyltransferase activity and the RPM-1 signaling hub. eLife 2019; 8:44040. [PMID: 30652969 PMCID: PMC6355192 DOI: 10.7554/elife.44040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022] Open
Abstract
Synapse formation is comprised of target cell recognition, synapse assembly, and synapse maintenance. Maintaining established synaptic connections is essential for generating functional circuitry and synapse instability is a hallmark of neurodegenerative disease. While many molecules impact synapse formation generally, we know little about molecules that affect synapse maintenance in vivo. Using genetics and developmental time course analysis in C.elegans, we show that the α-tubulin acetyltransferase ATAT-2 and the signaling hub RPM-1 are required presynaptically to maintain stable synapses. Importantly, the enzymatic acetyltransferase activity of ATAT-2 is required for synapse maintenance. Our analysis revealed that RPM-1 is a hub in a genetic network composed of ATAT-2, PTRN-1 and DLK-1. In this network, ATAT-2 functions independent of the DLK-1 MAPK and likely acts downstream of RPM-1. Thus, our study reveals an important role for tubulin acetyltransferase activity in presynaptic maintenance, which occurs via the RPM-1/ATAT-2 pathway.
Collapse
Affiliation(s)
- Melissa A Borgen
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Andrew C Giles
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Dandan Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Brock Grill
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| |
Collapse
|
26
|
Wang Q, Han TH, Nguyen P, Jarnik M, Serpe M. Tenectin recruits integrin to stabilize bouton architecture and regulate vesicle release at the Drosophila neuromuscular junction. eLife 2018; 7:35518. [PMID: 29901439 PMCID: PMC6040883 DOI: 10.7554/elife.35518] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 06/13/2018] [Indexed: 11/15/2022] Open
Abstract
Assembly, maintenance and function of synaptic junctions depend on extracellular matrix (ECM) proteins and their receptors. Here we report that Tenectin (Tnc), a Mucin-type protein with RGD motifs, is an ECM component required for the structural and functional integrity of synaptic specializations at the neuromuscular junction (NMJ) in Drosophila. Using genetics, biochemistry, electrophysiology, histology and electron microscopy, we show that Tnc is secreted from motor neurons and striated muscles and accumulates in the synaptic cleft. Tnc selectively recruits αPS2/βPS integrin at synaptic terminals, but only the cis Tnc/integrin complexes appear to be biologically active. These complexes have distinct pre- and postsynaptic functions, mediated at least in part through the local engagement of the spectrin-based membrane skeleton: the presynaptic complexes control neurotransmitter release, while postsynaptic complexes ensure the size and architectural integrity of synaptic boutons. Our study reveals an unprecedented role for integrin in the synaptic recruitment of spectrin-based membrane skeleton. Nerve cells or neurons can communicate with each other by releasing chemical messengers into the gap between them, the synapse. Both neurons and synapses are surrounded by a network of proteins called the extracellular matrix, which anchors, protects and supports the synapse. The matrix also helps to regulate the dynamic communication across the synapses and consequently neurons. Little is known about the proteins of the extracellular matrix, in particular about the ones involved in structural support. This is especially important for the so-called neuromuscular junctions, where neurons stimulate muscle contraction and trigger vigorous movement. Receptor proteins on cell surfaces, such as integrins, can bind to the extracellular matrix proteins to anchor the cells and are important for all cell junctions, including synaptic junctions. But because of their many essential roles during development, it was unclear how integrins modulate the activity of the synapse. To investigate this further, Wang et al. studied the neuromuscular junctions of fruit flies. The experiments revealed that both muscle and neurons secrete a large protein called Tenectin, which accumulates into the small space between the neuron and the muscle, the synaptic cleft. This protein can bind to integrin and is necessary to support the neuromuscular junction structurally and functionally. Wang et al. discovered that Tenectin works by gathering integrins on the surface of the neuron and the muscle. In the neuron, Tenectin forms complexes with integrin to regulate the release of neurotransmitters. In the muscle, the complexes provide support to the synaptic structures. However, when Tenectin was experimentally removed, it only disrupted the integrins at the neuromuscular junction, without affecting integrins in other regions of the cells, such as the site where the muscle uses integrins to attach to the tendon. Moreover, without Tenectin an important intracellular scaffolding meshwork that lines up and reinforces cell membranes was no longer organized properly at the synapse. A next step will be to identify the missing components between Tenectin/integrin complexes on the surface of neurons and the neurotransmitter release machinery inside the cells. The extracellular matrix and its receptors play fundamental roles in the development and function of the nervous system. A better knowledge of the underlying mechanisms will help us to better understand the complex interplay between the synapse and the extracellular matrix.
Collapse
Affiliation(s)
- Qi Wang
- Section on Cellular Communication, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Tae Hee Han
- Section on Cellular Communication, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Peter Nguyen
- Section on Cellular Communication, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Michal Jarnik
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Mihaela Serpe
- Section on Cellular Communication, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| |
Collapse
|
27
|
Li X, Goel P, Chen C, Angajala V, Chen X, Dickman DK. Synapse-specific and compartmentalized expression of presynaptic homeostatic potentiation. eLife 2018; 7:34338. [PMID: 29620520 PMCID: PMC5927770 DOI: 10.7554/elife.34338] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/04/2018] [Indexed: 01/23/2023] Open
Abstract
Postsynaptic compartments can be specifically modulated during various forms of synaptic plasticity, but it is unclear whether this precision is shared at presynaptic terminals. Presynaptic homeostatic plasticity (PHP) stabilizes neurotransmission at the Drosophila neuromuscular junction, where a retrograde enhancement of presynaptic neurotransmitter release compensates for diminished postsynaptic receptor functionality. To test the specificity of PHP induction and expression, we have developed a genetic manipulation to reduce postsynaptic receptor expression at one of the two muscles innervated by a single motor neuron. We find that PHP can be induced and expressed at a subset of synapses, over both acute and chronic time scales, without influencing transmission at adjacent release sites. Further, homeostatic modulations to CaMKII, vesicle pools, and functional release sites are compartmentalized and do not spread to neighboring pre- or post-synaptic structures. Thus, both PHP induction and expression mechanisms are locally transmitted and restricted to specific synaptic compartments. Everything we think and do is the result of communication between neurons. This communication takes place at junctions called synapses. When two nerve cells or neurons communicate at a synapse, the output terminal of the first cell releases a chemical called a neurotransmitter. This binds to receiver proteins, or receptors, on the second cell. When this communication is interrupted, synapses can adapt to maintain a stable dialogue between them. This can occur in two ways. Either the first neuron starts to release more neurotransmitter from its output terminal, or the second neuron produces extra receptors with which to detect the neurotransmitter. But how specific are these changes? The brain contains far more synapses than neurons because each neuron can form synapses with many other cells. Can a neuron adjust how much of the neurotransmitter it releases at some of its synapses while leaving the others unchanged? Li et al. have now addressed this question by studying a special type of synapse that forms between neurons and muscles, known as a neuromuscular junction. At one particular neuromuscular junction in fruit flies, a single neuron splits into two output terminals, each of which forms a synapse with a different muscle. Li et al. show that when the number of neurotransmitter receptors in one of the muscles is artificially reduced, the associated output terminal compensates by increasing its neurotransmitter release. By contrast, the other output terminal remains unaffected. This suggests that a neuron can induce remarkably specific changes in a subset of its synapses. This discovery paves the way towards identifying the smallest possible unit of change that can occur in the neurons’ ability to communicate. This unit may in turn be the smallest change that can support learning. Such knowledge will help us understand how the nervous system processes and stabilizes information transfer, both in health and after injury or disease.
Collapse
Affiliation(s)
- Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Neuroscience Graduate Program, University of Southern California, California, United States
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Graduate Program in Molecular and Computational Biology, University of Southern California, California, United States
| | - Catherine Chen
- Department of Neurobiology, University of Southern California, Los Angeles, United States
| | | | - Xun Chen
- Neuroscience Graduate Program, University of Southern California, California, United States
| | | |
Collapse
|
28
|
Migh E, Götz T, Földi I, Szikora S, Gombos R, Darula Z, Medzihradszky KF, Maléth J, Hegyi P, Sigrist S, Mihály J. Microtubule organization in presynaptic boutons relies on the formin DAAM. Development 2018; 145:dev158519. [PMID: 29487108 DOI: 10.1242/dev.158519] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 02/14/2018] [Indexed: 02/02/2023]
Abstract
Regulation of the cytoskeleton is fundamental to the development and function of synaptic terminals, such as neuromuscular junctions. Despite the identification of numerous proteins that regulate synaptic actin and microtubule dynamics, the mechanisms of cytoskeletal control during terminal arbor formation have remained largely elusive. Here, we show that DAAM, a member of the formin family of cytoskeleton organizing factors, is an important presynaptic regulator of neuromuscular junction development in Drosophila We demonstrate that the actin filament assembly activity of DAAM plays a negligible role in terminal formation; rather, DAAM is necessary for synaptic microtubule organization. Genetic interaction studies consistently link DAAM with the Wg/Ank2/Futsch module of microtubule regulation and bouton formation. Finally, we provide evidence that DAAM is tightly associated with the synaptic active zone scaffold, and electrophysiological data point to a role in the modulation of synaptic vesicle release. Based on these results, we propose that DAAM is an important cytoskeletal effector element of the Wg/Ank2 pathway involved in the determination of basic synaptic structures, and, additionally, that DAAM may couple the active zone scaffold to the presynaptic cytoskeleton.
Collapse
Affiliation(s)
- Ede Migh
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Torsten Götz
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - István Földi
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Rita Gombos
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Zsuzsanna Darula
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - Katalin F Medzihradszky
- Laboratory of Proteomics Research, Biological Research Centre, Hungarian Academy of Sciences, Szeged H-6726, Hungary
| | - József Maléth
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
| | - Péter Hegyi
- MTA-SZTE Translational Gastroenterology Research Group, Szeged H-6725, Hungary
- Institute for Translational Medicine, University of Pecs, Pécs H-7624, Hungary
| | - Stephan Sigrist
- Institut für Biologie/Genetik and NeuroCure, Freie Universitat Berlin, Takustrasse 6, D-14195 Berlin, Germany
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
29
|
Hauswirth AG, Ford KJ, Wang T, Fetter RD, Tong A, Davis GW. A postsynaptic PI3K-cII dependent signaling controller for presynaptic homeostatic plasticity. eLife 2018; 7:31535. [PMID: 29303480 PMCID: PMC5773188 DOI: 10.7554/elife.31535] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 01/04/2018] [Indexed: 01/29/2023] Open
Abstract
Presynaptic homeostatic plasticity stabilizes information transfer at synaptic connections in organisms ranging from insect to human. By analogy with principles of engineering and control theory, the molecular implementation of PHP is thought to require postsynaptic signaling modules that encode homeostatic sensors, a set point, and a controller that regulates transsynaptic negative feedback. The molecular basis for these postsynaptic, homeostatic signaling elements remains unknown. Here, an electrophysiology-based screen of the Drosophila kinome and phosphatome defines a postsynaptic signaling platform that includes a required function for PI3K-cII, PI3K-cIII and the small GTPase Rab11 during the rapid and sustained expression of PHP. We present evidence that PI3K-cII localizes to Golgi-derived, clathrin-positive vesicles and is necessary to generate an endosomal pool of PI(3)P that recruits Rab11 to recycling endosomal membranes. A morphologically distinct subdivision of this platform concentrates postsynaptically where we propose it functions as a homeostatic controller for retrograde, trans-synaptic signaling.
Collapse
Affiliation(s)
- Anna G Hauswirth
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Kevin J Ford
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Tingting Wang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Amy Tong
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, United States.,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
30
|
Manole A, Jaunmuktane Z, Hargreaves I, Ludtmann MHR, Salpietro V, Bello OD, Pope S, Pandraud A, Horga A, Scalco RS, Li A, Ashokkumar B, Lourenço CM, Heales S, Horvath R, Chinnery PF, Toro C, Singleton AB, Jacques TS, Abramov AY, Muntoni F, Hanna MG, Reilly MM, Revesz T, Kullmann DM, Jepson JEC, Houlden H. Clinical, pathological and functional characterization of riboflavin-responsive neuropathy. Brain 2017; 140:2820-2837. [PMID: 29053833 PMCID: PMC5808726 DOI: 10.1093/brain/awx231] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 01/02/2023] Open
Abstract
Brown-Vialetto-Van Laere syndrome represents a phenotypic spectrum of motor, sensory, and cranial nerve neuropathy, often with ataxia, optic atrophy and respiratory problems leading to ventilator-dependence. Loss-of-function mutations in two riboflavin transporter genes, SLC52A2 and SLC52A3, have recently been linked to Brown-Vialetto-Van Laere syndrome. However, the genetic frequency, neuropathology and downstream consequences of riboflavin transporter mutations are unclear. By screening a large cohort of 132 patients with early-onset severe sensory, motor and cranial nerve neuropathy we confirmed the strong genetic link between riboflavin transporter mutations and Brown-Vialetto-Van Laere syndrome, identifying 22 pathogenic mutations in SLC52A2 and SLC52A3, 14 of which were novel. Brain and spinal cord neuropathological examination of two cases with SLC52A3 mutations showed classical symmetrical brainstem lesions resembling pathology seen in mitochondrial disease, including severe neuronal loss in the lower cranial nerve nuclei, anterior horns and corresponding nerves, atrophy of the spinothalamic and spinocerebellar tracts and posterior column-medial lemniscus pathways. Mitochondrial dysfunction has previously been implicated in an array of neurodegenerative disorders. Since riboflavin metabolites are critical components of the mitochondrial electron transport chain, we hypothesized that reduced riboflavin transport would result in impaired mitochondrial activity, and confirmed this using in vitro and in vivo models. Electron transport chain complex I and complex II activity were decreased in SLC52A2 patient fibroblasts, while global knockdown of the single Drosophila melanogaster riboflavin transporter homologue revealed reduced levels of riboflavin, downstream metabolites, and electron transport chain complex I activity. This in turn led to abnormal mitochondrial membrane potential, respiratory chain activity and morphology. Riboflavin transporter knockdown in Drosophila also resulted in severely impaired locomotor activity and reduced lifespan, mirroring patient pathology, and these phenotypes could be partially rescued using a novel esterified derivative of riboflavin. Our findings expand the genetic, clinical and neuropathological features of Brown-Vialetto-Van Laere syndrome, implicate mitochondrial dysfunction as a downstream consequence of riboflavin transporter gene defects, and validate riboflavin esters as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Andreea Manole
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Zane Jaunmuktane
- Division of Neuropathology and Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Iain Hargreaves
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, London, WC1N 3BG, UK
| | - Marthe H R Ludtmann
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Vincenzo Salpietro
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Oscar D Bello
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Simon Pope
- Neurometabolic Unit, National Hospital for Neurology and Neurosurgery, London, WC1N 3BG, UK
| | - Amelie Pandraud
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Alejandro Horga
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Renata S Scalco
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Abi Li
- Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, Queen Square, London WC1N 3BG, UK
| | - Balasubramaniem Ashokkumar
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Charles M Lourenço
- Departamento de Neurociências e Ciências do Comportamento, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Simon Heales
- Chemical Pathology, Great Ormond Street Children’s Hospital, London, UK
| | - Rita Horvath
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- MRC Mitochondrial Biology Unit, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Camilo Toro
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, Bethesda, MD, USA
| | | | - Thomas S Jacques
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Andrey Y Abramov
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, UK
| | - Michael G Hanna
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Mary M Reilly
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Tamas Revesz
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Reta Lila Weston Institute of Neurological Studies and Queen Square Brain Bank for Neurological Disorders, Queen Square, London WC1N 3BG, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - James E C Jepson
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Henry Houlden
- Department of Molecular Neuroscience and Neurogenetics Laboratory, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| |
Collapse
|
31
|
β-III-spectrin spinocerebellar ataxia type 5 mutation reveals a dominant cytoskeletal mechanism that underlies dendritic arborization. Proc Natl Acad Sci U S A 2017; 114:E9376-E9385. [PMID: 29078305 DOI: 10.1073/pnas.1707108114] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A spinocerebellar ataxia type 5 (SCA5) L253P mutation in the actin-binding domain (ABD) of β-III-spectrin causes high-affinity actin binding and decreased thermal stability in vitro. Here we show in mammalian cells, at physiological temperature, that the mutant ABD retains high-affinity actin binding. Significantly, we provide evidence that the mutation alters the mobility and recruitment of β-III-spectrin in mammalian cells, pointing to a potential disease mechanism. To explore this mechanism, we developed a Drosophila SCA5 model in which an equivalent mutant Drosophila β-spectrin is expressed in neurons that extend complex dendritic arbors, such as Purkinje cells, targeted in SCA5 pathogenesis. The mutation causes a proximal shift in arborization coincident with decreased β-spectrin localization in distal dendrites. We show that SCA5 β-spectrin dominantly mislocalizes α-spectrin and ankyrin-2, components of the endogenous spectrin cytoskeleton. Our data suggest that high-affinity actin binding by SCA5 β-spectrin interferes with spectrin-actin cytoskeleton dynamics, leading to a loss of a cytoskeletal mechanism in distal dendrites required for dendrite stabilization and arbor outgrowth.
Collapse
|
32
|
Praschberger R, Lowe SA, Malintan NT, Giachello CNG, Patel N, Houlden H, Kullmann DM, Baines RA, Usowicz MM, Krishnakumar SS, Hodge JJL, Rothman JE, Jepson JEC. Mutations in Membrin/GOSR2 Reveal Stringent Secretory Pathway Demands of Dendritic Growth and Synaptic Integrity. Cell Rep 2017; 21:97-109. [PMID: 28978487 PMCID: PMC5640804 DOI: 10.1016/j.celrep.2017.09.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 11/16/2022] Open
Abstract
Mutations in the Golgi SNARE (SNAP [soluble NSF attachment protein] receptor) protein Membrin (encoded by the GOSR2 gene) cause progressive myoclonus epilepsy (PME). Membrin is a ubiquitous and essential protein mediating ER-to-Golgi membrane fusion. Thus, it is unclear how mutations in Membrin result in a disorder restricted to the nervous system. Here, we use a multi-layered strategy to elucidate the consequences of Membrin mutations from protein to neuron. We show that the pathogenic mutations cause partial reductions in SNARE-mediated membrane fusion. Importantly, these alterations were sufficient to profoundly impair dendritic growth in Drosophila models of GOSR2-PME. Furthermore, we show that Membrin mutations cause fragmentation of the presynaptic cytoskeleton coupled with transsynaptic instability and hyperactive neurotransmission. Our study highlights how dendritic growth is vulnerable even to subtle secretory pathway deficits, uncovers a role for Membrin in synaptic function, and provides a comprehensive explanatory basis for genotype-phenotype relationships in GOSR2-PME.
Collapse
Affiliation(s)
- Roman Praschberger
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Simon A Lowe
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | - Nancy T Malintan
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Carlo N G Giachello
- Faculty of Biology, Medicine, and Health, Division of Neuroscience & Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Nian Patel
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Henry Houlden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK
| | - Richard A Baines
- Faculty of Biology, Medicine, and Health, Division of Neuroscience & Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Maria M Usowicz
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | - Shyam S Krishnakumar
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK; Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - James J L Hodge
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, UK
| | - James E Rothman
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK; Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - James E C Jepson
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK.
| |
Collapse
|
33
|
Danella EB, Keller LC. A Simple Neuronal Mechanical Injury Methodology to Study Drosophila Motor Neuron Degeneration. J Vis Exp 2017. [PMID: 28745645 DOI: 10.3791/56128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The degeneration of neurons occurs during normal development and in response to injury, stress, and disease. The cellular hallmarks of neuronal degeneration are remarkably similar in humans and invertebrates as are the molecular mechanisms that drive these processes. The fruit fly, Drosophila melanogaster, provides a powerful yet simple genetic model organism to study the cellular complexities of neurodegenerative diseases. In fact, approximately 70% of disease-associated human genes have a Drosophila homolog and a plethora of tools and assays have been described using flies to study human neurodegenerative diseases. More specifically the neuromuscular junction (NMJ) in Drosophila has proven to be an effective system to study neuromuscular diseases because of the ability to analyze the structural connections between the neuron and the muscle. Here, we report on an in vivo motor neuron injury assay in Drosophila, which reproducibly induces neurodegeneration at the NMJ by 24 h. Using this methodology, we have described a temporal sequence of cellular events resulting in motor neuron degeneration. The injury method has diverse applications and has also been utilized to identify specific genes required for neurodegeneration and to dissect transcriptional responses to neuronal injury.
Collapse
Affiliation(s)
| | - Lani C Keller
- Department of Biological Sciences, Quinnipiac University;
| |
Collapse
|
34
|
Qu Y, Hahn I, Webb SED, Pearce SP, Prokop A. Periodic actin structures in neuronal axons are required to maintain microtubules. Mol Biol Cell 2016; 28:296-308. [PMID: 27881663 PMCID: PMC5231898 DOI: 10.1091/mbc.e16-10-0727] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/09/2016] [Accepted: 11/15/2016] [Indexed: 12/31/2022] Open
Abstract
Drosophila genetics is combined with high-resolution microscopy and a number of functional readouts to demonstrate key factors required for the presence of regularly spaced rings of cortical actin in axons. The data suggest important roles for the actin rings in microtubule regulation, most likely by sustaining their polymerization. Axons are cable-like neuronal processes wiring the nervous system. They contain parallel bundles of microtubules as structural backbones, surrounded by regularly spaced actin rings termed the periodic membrane skeleton (PMS). Despite being an evolutionarily conserved, ubiquitous, highly ordered feature of axons, the function of PMS is unknown. Here we studied PMS abundance, organization, and function, combining versatile Drosophila genetics with superresolution microscopy and various functional readouts. Analyses with 11 actin regulators and three actin-targeting drugs suggest that PMS contains short actin filaments that are depolymerization resistant and sensitive to spectrin, adducin, and nucleator deficiency, consistent with microscopy-derived models proposing PMS as specialized cortical actin. Upon actin removal, we observed gaps in microtubule bundles, reduced microtubule polymerization, and reduced axon numbers, suggesting a role of PMS in microtubule organization. These effects become strongly enhanced when carried out in neurons lacking the microtubule-stabilizing protein Short stop (Shot). Combining the aforementioned actin manipulations with Shot deficiency revealed a close correlation between PMS abundance and microtubule regulation, consistent with a model in which PMS-dependent microtubule polymerization contributes to their maintenance in axons. We discuss potential implications of this novel PMS function along axon shafts for axon maintenance and regeneration.
Collapse
Affiliation(s)
- Yue Qu
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Ines Hahn
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Stephen E D Webb
- Rutherford Appleton Laboratory, Science and Technology Facilities Council, Didcot OX11 0QX, United Kingdom
| | - Simon P Pearce
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom.,School of Mathematics, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Andreas Prokop
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
35
|
Schulte C, Ripamonti M, Maffioli E, Cappelluti MA, Nonnis S, Puricelli L, Lamanna J, Piazzoni C, Podestà A, Lenardi C, Tedeschi G, Malgaroli A, Milani P. Scale Invariant Disordered Nanotopography Promotes Hippocampal Neuron Development and Maturation with Involvement of Mechanotransductive Pathways. Front Cell Neurosci 2016; 10:267. [PMID: 27917111 PMCID: PMC5114288 DOI: 10.3389/fncel.2016.00267] [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: 07/27/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022] Open
Abstract
The identification of biomaterials which promote neuronal maturation up to the generation of integrated neural circuits is fundamental for modern neuroscience. The development of neural circuits arises from complex maturative processes regulated by poorly understood signaling events, often guided by the extracellular matrix (ECM). Here we report that nanostructured zirconia surfaces, produced by supersonic cluster beam deposition of zirconia nanoparticles and characterized by ECM-like nanotopographical features, can direct the maturation of neural networks. Hippocampal neurons cultured on such cluster-assembled surfaces displayed enhanced differentiation paralleled by functional changes. The latter was demonstrated by single-cell electrophysiology showing earlier action potential generation and increased spontaneous postsynaptic currents compared to the neurons grown on the featureless unnaturally flat standard control surfaces. Label-free shotgun proteomics broadly confirmed the functional changes and suggests furthermore a vast impact of the neuron/nanotopography interaction on mechanotransductive machinery components, known to control physiological in vivo ECM-regulated axon guidance and synaptic plasticity. Our results indicate a potential of cluster-assembled zirconia nanotopography exploitable for the creation of efficient neural tissue interfaces and cell culture devices promoting neurogenic events, but also for unveiling mechanotransductive aspects of neuronal development and maturation.
Collapse
Affiliation(s)
- Carsten Schulte
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di MilanoMilan, Italy; Fondazione FilareteMilan, Italy
| | - Maddalena Ripamonti
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Elisa Maffioli
- Fondazione FilareteMilan, Italy; Dipartimento di Medicina Veterinaria, Università degli Studi di MilanoMilan, Italy
| | - Martino A Cappelluti
- Fondazione FilareteMilan, Italy; SEMM - European School of Molecular MedicineMilan, Italy
| | - Simona Nonnis
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano Milan, Italy
| | - Luca Puricelli
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Jacopo Lamanna
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Claudio Piazzoni
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Alessandro Podestà
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Cristina Lenardi
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| | - Gabriella Tedeschi
- Fondazione FilareteMilan, Italy; Dipartimento di Medicina Veterinaria, Università degli Studi di MilanoMilan, Italy
| | - Antonio Malgaroli
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute San Raffaele, Università Vita-Salute San Raffaele Milan, Italy
| | - Paolo Milani
- Dipartimento di Fisica, Centro Interdisciplinare Materiali e Interfacce Nanostrutturate, Università degli Studi di Milano Milan, Italy
| |
Collapse
|
36
|
Banerjee S, Venkatesan A, Bhat MA. Neurexin, Neuroligin and Wishful Thinking coordinate synaptic cytoarchitecture and growth at neuromuscular junctions. Mol Cell Neurosci 2016; 78:9-24. [PMID: 27838296 DOI: 10.1016/j.mcn.2016.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/19/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022] Open
Abstract
Trans-synaptic interactions involving Neurexins and Neuroligins are thought to promote adhesive interactions for precise alignment of the pre- and postsynaptic compartments and organize synaptic macromolecular complexes across species. In Drosophila, while Neurexin (Dnrx) and Neuroligins (Dnlg) are emerging as central organizing molecules at synapses, very little is known of the spectrum of proteins that might be recruited to the Dnrx/Dnlg trans-synaptic interface for organization and growth of the synapses. Using full length and truncated forms of Dnrx and Dnlg1 together with cell biological analyses and genetic interactions, we report novel functions of Dnrx and Dnlg1 in clustering of pre- and postsynaptic proteins, coordination of synaptic growth and ultrastructural organization. We show that Dnrx and Dnlg1 extracellular and intracellular regions are required for proper synaptic growth and localization of Dnlg1 and Dnrx, respectively. dnrx and dnlg1 single and double mutants display altered subcellular distribution of Discs large (Dlg), which is the homolog of mammalian post-synaptic density protein, PSD95. dnrx and dnlg1 mutants also display ultrastructural defects ranging from abnormal active zones, misformed pre- and post-synaptic areas with underdeveloped subsynaptic reticulum. Interestingly, dnrx and dnlg1 mutants have reduced levels of the Bone Morphogenetic Protein (BMP) receptor Wishful thinking (Wit), and Dnrx and Dnlg1 are required for proper localization and stability of Wit. In addition, the synaptic overgrowth phenotype resulting from the overexpression of Dnrx fails to manifest in wit mutants. Phenotypic analyses of dnrx/wit and dnlg1/wit mutants indicate that Dnrx/Dnlg1/Wit coordinate synaptic growth and architecture at the NMJ. Our findings also demonstrate that loss of Dnrx and Dnlg1 leads to decreased levels of the BMP co-receptor, Thickveins and the downstream effector phosphorylated Mad at the Neuromuscular Junction (NMJ) synapses indicating that Dnrx/Dnlg1 regulate components of the BMP signaling pathway. Together our findings reveal that Dnrx/Dnlg are at the core of a highly orchestrated process that combines adhesive and signaling mechanisms to ensure proper synaptic organization and growth during NMJ development.
Collapse
Affiliation(s)
- Swati Banerjee
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| | - Anandakrishnan Venkatesan
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| |
Collapse
|
37
|
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
|
38
|
Multicolour Multilevel STED nanoscopy of Actin/Spectrin Organization at Synapses. Sci Rep 2016; 6:26725. [PMID: 27220554 PMCID: PMC4879624 DOI: 10.1038/srep26725] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 11/29/2022] Open
Abstract
Superresolution fluorescence microscopy of multiple fluorophores still requires development. Here we present simultaneous three-colour stimulated emission depletion (STED) nanoscopy relying on a single STED beam at 620 nm. Toggling the STED beam between two or more power levels (“multilevelSTED”) optimizes resolution and contrast in all colour channels, which are intrinsically co-aligned and well separated. Three-colour recording is demonstrated by imaging the nanoscale cytoskeletal organization in cultured hippocampal neurons. The down to ~35 nm resolution identified periodic actin/betaII spectrin lattices along dendrites and spines; however, at presynaptic and postsynaptic sites, these patterns were found to be absent. Both our multicolour scheme and the 620 nm STED line should be attractive for routine STED microscopy applications.
Collapse
|
39
|
Prevalent presence of periodic actin-spectrin-based membrane skeleton in a broad range of neuronal cell types and animal species. Proc Natl Acad Sci U S A 2016; 113:6029-34. [PMID: 27162329 DOI: 10.1073/pnas.1605707113] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Actin, spectrin, and associated molecules form a periodic, submembrane cytoskeleton in the axons of neurons. For a better understanding of this membrane-associated periodic skeleton (MPS), it is important to address how prevalent this structure is in different neuronal types, different subcellular compartments, and across different animal species. Here, we investigated the organization of spectrin in a variety of neuronal- and glial-cell types. We observed the presence of MPS in all of the tested neuronal types cultured from mouse central and peripheral nervous systems, including excitatory and inhibitory neurons from several brain regions, as well as sensory and motor neurons. Quantitative analyses show that MPS is preferentially formed in axons in all neuronal types tested here: Spectrin shows a long-range, periodic distribution throughout all axons but appears periodic only in a small fraction of dendrites, typically in the form of isolated patches in subregions of these dendrites. As in dendrites, we also observed patches of periodic spectrin structures in a small fraction of glial-cell processes in four types of glial cells cultured from rodent tissues. Interestingly, despite its strong presence in the axonal shaft, MPS is disrupted in most presynaptic boutons but is present in an appreciable fraction of dendritic spine necks, including some projecting from dendrites where such a periodic structure is not observed in the shaft. Finally, we found that spectrin is capable of adopting a similar periodic organization in neurons of a variety of animal species, including Caenorhabditis elegans, Drosophila, Gallus gallus, Mus musculus, and Homo sapiens.
Collapse
|
40
|
Perkins E, Suminaite D, Jackson M. Cerebellar ataxias: β-III spectrin's interactions suggest common pathogenic pathways. J Physiol 2016; 594:4661-76. [PMID: 26821241 PMCID: PMC4983618 DOI: 10.1113/jp271195] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/14/2015] [Indexed: 12/12/2022] Open
Abstract
Spinocerebellar ataxias (SCAs) are a genetically heterogeneous group of disorders all characterised by postural abnormalities, motor deficits and cerebellar degeneration. Animal and in vitro models have revealed β‐III spectrin, a cytoskeletal protein present throughout the soma and dendritic tree of cerebellar Purkinje cells, to be required for the maintenance of dendritic architecture and for the trafficking and/or stabilisation of several membrane proteins: ankyrin‐R, cell adhesion molecules, metabotropic glutamate receptor‐1 (mGluR1), voltage‐gated sodium channels (Nav) and glutamate transporters. This scaffold of interactions connects β‐III spectrin to a wide variety of proteins implicated in the pathology of many SCAs. Heterozygous mutations in the gene encoding β‐III spectrin (SPTBN2) underlie SCA type‐5 whereas homozygous mutations cause spectrin associated autosomal recessive ataxia type‐1 (SPARCA1), an infantile form of ataxia with cognitive impairment. Loss‐of β‐III spectrin function appears to underpin cerebellar dysfunction and degeneration in both diseases resulting in thinner dendrites, excessive dendritic protrusion with loss of planarity, reduced resurgent sodium currents and abnormal glutamatergic neurotransmission. The initial physiological consequences are a decrease in spontaneous activity and excessive excitation, likely to be offsetting each other, but eventually hyperexcitability gives rise to dark cell degeneration and reduced cerebellar output. Similar molecular mechanisms have been implicated for SCA1, 2, 3, 7, 13, 14, 19, 22, 27 and 28, highlighting alterations to intrinsic Purkinje cell activity, dendritic architecture and glutamatergic transmission as possible common mechanisms downstream of various loss‐of‐function primary genetic defects. A key question for future research is whether similar mechanisms underlie progressive cerebellar decline in normal ageing.
![]()
Collapse
Affiliation(s)
- Emma Perkins
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Daumante Suminaite
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Mandy Jackson
- Centre for Integrative Physiology, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| |
Collapse
|
41
|
Grill B, Murphey RK, Borgen MA. The PHR proteins: intracellular signaling hubs in neuronal development and axon degeneration. Neural Dev 2016; 11:8. [PMID: 27008623 PMCID: PMC4806438 DOI: 10.1186/s13064-016-0063-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/15/2016] [Indexed: 11/10/2022] Open
Abstract
During development, a coordinated and integrated series of events must be accomplished in order to generate functional neural circuits. Axons must navigate toward target cells, build synaptic connections, and terminate outgrowth. The PHR proteins (consisting of mammalian Phr1/MYCBP2, Drosophila Highwire and C. elegans RPM-1) function in each of these events in development. Here, we review PHR function across species, as well as the myriad of signaling pathways PHR proteins regulate. These findings collectively suggest that the PHR proteins are intracellular signaling hubs, a concept we explore in depth. Consistent with prominent developmental functions, genetic links have begun to emerge between PHR signaling networks and neurodevelopmental disorders, such as autism, schizophrenia and intellectual disability. Finally, we discuss the recent and important finding that PHR proteins regulate axon degeneration, which has further heightened interest in this fascinating group of molecules.
Collapse
Affiliation(s)
- Brock Grill
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, FL, 33458, USA.
| | - Rodney K Murphey
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Melissa A Borgen
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, FL, 33458, USA
| |
Collapse
|
42
|
Ng BF, Selvaraj GK, Santa-Cruz Mateos C, Grosheva I, Alvarez-Garcia I, Martín-Bermudo MD, Palacios IM. α-Spectrin and integrins act together to regulate actomyosin and columnarization, and to maintain a monolayered follicular epithelium. Development 2016; 143:1388-99. [PMID: 26952981 PMCID: PMC4852512 DOI: 10.1242/dev.130070] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/18/2016] [Indexed: 12/26/2022]
Abstract
The spectrin cytoskeleton crosslinks actin to the membrane, and although it has been greatly studied in erythrocytes, much is unknown about its function in epithelia. We have studied the role of spectrins during epithelia morphogenesis using the Drosophila follicular epithelium (FE). As previously described, we show that α-Spectrin and β-Spectrin are essential to maintain a monolayered FE, but, contrary to previous work, spectrins are not required to control proliferation. Furthermore, spectrin mutant cells show differentiation and polarity defects only in the ectopic layers of stratified epithelia, similar to integrin mutants. Our results identify α-Spectrin and integrins as novel regulators of apical constriction-independent cell elongation, as α-Spectrin and integrin mutant cells fail to columnarize. Finally, we show that increasing and reducing the activity of the Rho1-Myosin II pathway enhances and decreases multilayering of α-Spectrin cells, respectively. Similarly, higher Myosin II activity enhances the integrin multilayering phenotype. This work identifies a primary role for α-Spectrin in controlling cell shape, perhaps by modulating actomyosin. In summary, we suggest that a functional spectrin-integrin complex is essential to balance adequate forces, in order to maintain a monolayered epithelium.
Collapse
Affiliation(s)
- Bing Fu Ng
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Gokul Kannan Selvaraj
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | | | - Inna Grosheva
- Centro Andaluz de Biología del Desarrollo CSIC-Univ. Pablo de Olavide, Sevilla 41013, Spain
| | - Ines Alvarez-Garcia
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | | | - Isabel M Palacios
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| |
Collapse
|
43
|
An Adaptable Spectrin/Ankyrin-Based Mechanism for Long-Range Organization of Plasma Membranes in Vertebrate Tissues. CURRENT TOPICS IN MEMBRANES 2015; 77:143-84. [PMID: 26781832 DOI: 10.1016/bs.ctm.2015.10.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ankyrins are membrane-associated proteins that together with their spectrin partners are responsible for micron-scale organization of vertebrate plasma membranes, including those of erythrocytes, excitable membranes of neurons and heart, lateral membrane domains of columnar epithelial cells, and striated muscle. Ankyrins coordinate functionally related membrane transporters and cell adhesion proteins (15 protein families identified so far) within plasma membrane compartments through independently evolved interactions of intrinsically disordered sequences with a highly conserved peptide-binding groove formed by the ANK repeat solenoid. Ankyrins are coupled to spectrins, which are elongated organelle-sized proteins that form mechanically resilient arrays through cross-linking by specialized actin filaments. In addition to protein interactions, cellular targeting and assembly of spectrin/ankyrin domains also critically depend on palmitoylation of ankyrin-G by aspartate-histidine-histidine-cysteine 5/8 palmitoyltransferases, as well as interaction of beta-2 spectrin with phosphoinositide lipids. These lipid-dependent spectrin/ankyrin domains are not static but are locally dynamic and determine membrane identity through opposing endocytosis of bulk lipids as well as specific proteins. A partnership between spectrin, ankyrin, and cell adhesion molecules first emerged in bilaterians over 500 million years ago. Ankyrin and spectrin may have been recruited to plasma membranes from more ancient roles in organelle transport. The basic bilaterian spectrin-ankyrin toolkit markedly expanded in vertebrates through gene duplications combined with variation in unstructured intramolecular regulatory sequences as well as independent evolution of ankyrin-binding activity by ion transporters involved in action potentials and calcium homeostasis. In addition, giant vertebrate ankyrins with specialized roles in axons acquired new coding sequences by exon shuffling. We speculate that early axon initial segments and epithelial lateral membranes initially were based on spectrin-ankyrin-cell adhesion molecule assemblies and subsequently served as "incubators," where ion transporters independently acquired ankyrin-binding activity through positive selection.
Collapse
|
44
|
Lincoln BL, Alabsi SH, Frendo N, Freund R, Keller LC. Drosophila Neuronal Injury Follows a Temporal Sequence of Cellular Events Leading to Degeneration at the Neuromuscular Junction. J Exp Neurosci 2015; 9:1-9. [PMID: 26512206 PMCID: PMC4612769 DOI: 10.4137/jen.s25516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 11/12/2022] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide, and as the global population ages, there is a critical need to improve our understanding of the molecular and cellular mechanisms that drive neurodegeneration. At the molecular level, neurodegeneration involves the activation of complex signaling pathways that drive the active destruction of neurons and their intracellular components. Here, we use an in vivo motor neuron injury assay to acutely induce neurodegeneration in order to follow the temporal order of events that occur following injury in Drosophila melanogaster. We find that sites of injury can be rapidly identified based on structural defects to the neuronal cytoskeleton that result in disrupted axonal transport. Additionally, the neuromuscular junction accumulates ubiquitinated proteins prior to the neurodegenerative events, occurring at 24 hours post injury. Our data provide insights into the early molecular events that occur during axonal and neuromuscular degeneration in a genetically tractable model organism. Importantly, the mechanisms that mediate neurodegeneration in flies are conserved in humans. Thus, these studies have implications for our understanding of the cellular and molecular events that occur in humans and will facilitate the identification of biomedically relevant targets for future treatments.
Collapse
Affiliation(s)
- Barron L Lincoln
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Sahar H Alabsi
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Nicholas Frendo
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Robert Freund
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| | - Lani C Keller
- Department of Biological Sciences, Quinnipiac University, Hamden, CT, USA
| |
Collapse
|
45
|
Harris KP, Littleton JT. Transmission, Development, and Plasticity of Synapses. Genetics 2015; 201:345-75. [PMID: 26447126 PMCID: PMC4596655 DOI: 10.1534/genetics.115.176529] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/28/2015] [Indexed: 01/03/2023] Open
Abstract
Chemical synapses are sites of contact and information transfer between a neuron and its partner cell. Each synapse is a specialized junction, where the presynaptic cell assembles machinery for the release of neurotransmitter, and the postsynaptic cell assembles components to receive and integrate this signal. Synapses also exhibit plasticity, during which synaptic function and/or structure are modified in response to activity. With a robust panel of genetic, imaging, and electrophysiology approaches, and strong evolutionary conservation of molecular components, Drosophila has emerged as an essential model system for investigating the mechanisms underlying synaptic assembly, function, and plasticity. We will discuss techniques for studying synapses in Drosophila, with a focus on the larval neuromuscular junction (NMJ), a well-established model glutamatergic synapse. Vesicle fusion, which underlies synaptic release of neurotransmitters, has been well characterized at this synapse. In addition, studies of synaptic assembly and organization of active zones and postsynaptic densities have revealed pathways that coordinate those events across the synaptic cleft. We will also review modes of synaptic growth and plasticity at the fly NMJ, and discuss how pre- and postsynaptic cells communicate to regulate plasticity in response to activity.
Collapse
Affiliation(s)
- Kathryn P Harris
- Department of Biology and Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - J Troy Littleton
- Department of Biology and Department of Brain and Cognitive Sciences, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
46
|
Dubey J, Ratnakaran N, Koushika SP. Neurodegeneration and microtubule dynamics: death by a thousand cuts. Front Cell Neurosci 2015; 9:343. [PMID: 26441521 PMCID: PMC4563776 DOI: 10.3389/fncel.2015.00343] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022] Open
Abstract
Microtubules form important cytoskeletal structures that play a role in establishing and maintaining neuronal polarity, regulating neuronal morphology, transporting cargo, and scaffolding signaling molecules to form signaling hubs. Within a neuronal cell, microtubules are found to have variable lengths and can be both stable and dynamic. Microtubule associated proteins, post-translational modifications of tubulin subunits, microtubule severing enzymes, and signaling molecules are all known to influence both stable and dynamic pools of microtubules. Microtubule dynamics, the process of interconversion between stable and dynamic pools, and the proportions of these two pools have the potential to influence a wide variety of cellular processes. Reduced microtubule stability has been observed in several neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic Lateral Sclerosis (ALS), and tauopathies like Progressive Supranuclear Palsy. Hyperstable microtubules, as seen in Hereditary Spastic Paraplegia (HSP), also lead to neurodegeneration. Therefore, the ratio of stable and dynamic microtubules is likely to be important for neuronal function and perturbation in microtubule dynamics might contribute to disease progression.
Collapse
Affiliation(s)
- Jyoti Dubey
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India ; InStem Bangalore, India
| | - Neena Ratnakaran
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research Mumbai, India
| |
Collapse
|
47
|
Peuralinna T, Myllykangas L, Oinas M, Nalls MA, Keage HAD, Isoviita VM, Valori M, Polvikoski T, Paetau A, Sulkava R, Ince PG, Zaccai J, Brayne C, Traynor BJ, Hardy J, Singleton AB, Tienari PJ. Genome-wide association study of neocortical Lewy-related pathology. Ann Clin Transl Neurol 2015; 2:920-31. [PMID: 26401513 PMCID: PMC4574809 DOI: 10.1002/acn3.231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/22/2015] [Indexed: 02/04/2023] Open
Abstract
Objective Dementia with Lewy bodies is an α-synucleinopathy characterized by neocortical Lewy-related pathology (LRP). We carried out a genome-wide association study (GWAS) on neocortical LRP in a population-based sample of subjects aged 85 or over. Methods LRP was analyzed in 304 subjects in the Vantaa 85+ sample from Southern Finland. The GWAS included 41 cases with midbrain, hippocampal, and neocortical LRP and 177 controls without midbrain and hippocampal LRP. The Medical Research Council Cognitive Function and Ageing Study (CFAS) material was used for replication (51 cases and 131 controls). Results By analyzing 327,010 markers the top signal was obtained at the HLA-DPA1/DPB1 locus (P = 1.29 × 10−7); five other loci on chromosomes 15q14, 2p21, 2q31, 18p11, and 5q23 were associated with neocortical LRP at P < 10−5. Two loci were marked by multiple markers, 2p21 (P = 3.9 × 10−6, upstream of the SPTBN1 gene), and HLA-DPA1/DPB1; these were tested in the CFAS material. Single marker (P = 0.0035) and haplotype (P = 0.04) associations on 2p21 were replicated in CFAS, whereas HLA-DPA1/DPB1 association was not. Bioinformatic analyses suggest functional effects for the HLA-DPA1/DPB1 markers as well as the 15q14 marker rs8037309. Interpretation We identified suggestive novel risk factors for neocortical LRP. SPTBN1 is the candidate on 2p21, it encodes beta-spectrin, an α-synuclein binding protein and a component of Lewy bodies. The HLA-DPA1/DPB1 association suggests a role for antigen presentation or alternatively, cis-regulatory effects, one of the regulated neighboring genes identified here (vacuolar protein sorting 52) plays a role in vesicular trafficking and has been shown to interact with α-synuclein in a yeast model.
Collapse
Affiliation(s)
- Terhi Peuralinna
- Molecular Neurology, Research Program Unit, Biomedicum, University of Helsinki Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB Helsinki, Finland ; Folkhalsan Institute of Genetics Helsinki, Finland
| | - Minna Oinas
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB Helsinki, Finland ; Department of Neurosurgery, Helsinki University Central Hospital Helsinki, Finland
| | - Mike A Nalls
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, NIH Bethesda, Maryland
| | - Hannah A D Keage
- School of Psychology, Social Work and Social Policy, University of South Australia Adelaide, Australia ; Department of Public Health and Primary Care, University of Cambridge Cambridge, United Kingdom
| | - Veli-Matti Isoviita
- Molecular Neurology, Research Program Unit, Biomedicum, University of Helsinki Helsinki, Finland
| | - Miko Valori
- Molecular Neurology, Research Program Unit, Biomedicum, University of Helsinki Helsinki, Finland
| | - Tuomo Polvikoski
- Institute for Ageing and Health, Newcastle University Newcastle, United Kingdom
| | - Anders Paetau
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB Helsinki, Finland
| | - Raimo Sulkava
- School of Public Health and Clinical Nutrition, University of Eastern Finland Kuopio, Finland
| | - Paul G Ince
- Department of Neuroscience, University of Sheffield Sheffield, United Kingdom
| | - Julia Zaccai
- Department of Public Health and Primary Care, University of Cambridge Cambridge, United Kingdom
| | - Carol Brayne
- Department of Public Health and Primary Care, University of Cambridge Cambridge, United Kingdom
| | - Bryan J Traynor
- Neuromuscular Diseases Research Group, Laboratory of Neurogenetics, National Institute on Aging, NIH Bethesda, Maryland
| | - John Hardy
- Reta Lila Weston Research Laboratories, Departments of Molecular Neuroscience and of Clinical Neuroscience, UCL Institute of Neurology Queen Square, London, United Kingdom
| | - Andrew B Singleton
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, NIH Bethesda, Maryland
| | - Pentti J Tienari
- Molecular Neurology, Research Program Unit, Biomedicum, University of Helsinki Helsinki, Finland ; Department of Neurology, Helsinki University Central Hospital Helsinki, Finland
| |
Collapse
|
48
|
Tian X, Wu C. Active zone stability: insights from fly neuromuscular junction. Neural Regen Res 2015; 10:677-8. [PMID: 26109929 PMCID: PMC4468746 DOI: 10.4103/1673-5374.156942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2015] [Indexed: 11/23/2022] Open
Abstract
The presynaptic active zone is a dynamic structure that orchestrates regulated release of neurotransmitters. Developmental and aging processes, and changes in neuronal network activity can all modulate the number, size and composition of active zone and thereby synaptic efficacy. However, very little is known about the mechanism that controls the structural stability of active zone. By studying a model synapse, the Drosophila neuromuscular junction, our recent work shed light on how two scaffolding proteins at the active zone regulate active zone stability by promoting a localized dephosphorylation event at the nerve terminal. Here we discuss the major insights from our findings and their implications for future research.
Collapse
Affiliation(s)
- Xiaolin Tian
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Chunlai Wu
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
49
|
Mosca TJ. On the Teneurin track: a new synaptic organization molecule emerges. Front Cell Neurosci 2015; 9:204. [PMID: 26074772 PMCID: PMC4444827 DOI: 10.3389/fncel.2015.00204] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/11/2015] [Indexed: 11/16/2022] Open
Abstract
To achieve proper synaptic development and function, coordinated signals must pass between the pre- and postsynaptic membranes. Such transsynaptic signals can be comprised of receptors and secreted ligands, membrane associated receptors, and also pairs of synaptic cell adhesion molecules. A critical open question bridging neuroscience, developmental biology, and cell biology involves identifying those signals and elucidating how they function. Recent work in Drosophila and vertebrate systems has implicated a family of proteins, the Teneurins, as a new transsynaptic signal in both the peripheral and central nervous systems. The Teneurins have established roles in neuronal wiring, but studies now show their involvement in regulating synaptic connections between neurons and bridging the synaptic membrane and the cytoskeleton. This review will examine the Teneurins as synaptic cell adhesion molecules, explore how they regulate synaptic organization, and consider how some consequences of human Teneurin mutations may have synaptopathic origins.
Collapse
Affiliation(s)
- Timothy J Mosca
- Department of Biology, Stanford University Stanford, CA, USA
| |
Collapse
|
50
|
Glial cells in neuronal development: recent advances and insights from Drosophila melanogaster. Neurosci Bull 2015; 30:584-94. [PMID: 25015062 DOI: 10.1007/s12264-014-1448-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/22/2014] [Indexed: 12/30/2022] Open
Abstract
Glia outnumber neurons and are the most abundant cell type in the nervous system. Whereas neurons are the major carriers, transducers, and processors of information, glial cells, once considered mainly to play a passive supporting role, are now recognized for their active contributions to almost every aspect of nervous system development. Recently, insights from the invertebrate organism Drosophila melanogaster have advanced our knowledge of glial cell biology. In particular, findings on neuron-glia interactions via intrinsic and extrinsic mechanisms have shed light on the importance of glia during different stages of neuronal development. Here, we summarize recent advances in understanding the functions of Drosophila glia, which resemble their mammalian counterparts in morphology and function, neural stem-cell conversion, synapse formation, and developmental axon pruning. These discoveries reinforce the idea that glia are substantial players in the developing nervous system and further advance the understanding of mechanisms leading to neurodegeneration.
Collapse
|