1
|
Xu N, Chen SY, Tang AH. Tuning synapse strength by nanocolumn plasticity. Trends Neurosci 2025:S0166-2236(24)00253-4. [PMID: 39848836 DOI: 10.1016/j.tins.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/12/2024] [Accepted: 12/31/2024] [Indexed: 01/25/2025]
Abstract
The precise organization of the complex set of synaptic proteins at the nanometer scale is crucial for synaptic transmission. At the heart of this nanoscale architecture lies the nanocolumn. This aligns presynaptic neurotransmitter release with a high local density of postsynaptic receptor channels, thereby optimizing synaptic strength. Although synapses exhibit diverse protein compositions and nanoscale organizations, the role of structural diversity in the notable differences observed in synaptic physiology remains poorly understood. In this review we examine the current literature on the molecular mechanisms underlying the formation and maintenance of nanocolumns, as well as their role in modulating various aspects of synaptic transmission. We also discuss how the reorganization of nanocolumns contributes to functional dynamics in both synaptic plasticity and pathology.
Collapse
Affiliation(s)
- Na Xu
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China; School of Medicine, Anhui University of Science and Technology, Huainan 232001, China.
| | - Si-Yu Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, Department of Neurology in the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China.
| |
Collapse
|
2
|
Züfle P, Batista LL, Brandão SC, D’Uva G, Daniel C, Martelli C. Impact of developmental temperature on neural growth, connectivity, and function. SCIENCE ADVANCES 2025; 11:eadp9587. [PMID: 39813340 PMCID: PMC11734716 DOI: 10.1126/sciadv.adp9587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/06/2024] [Indexed: 01/18/2025]
Abstract
Environmental temperature dictates the developmental pace of poikilothermic animals. In Drosophila, slower development at lower temperatures results in higher brain connectivity, but the generality of such scaling across temperatures and brain regions and its impact on function are unclear. Here, we show that brain connectivity scales continuously across temperatures, in agreement with a first-principle model that postulates different metabolic constraints for the growth of the brain and the organism. The model predicts brain wiring under temperature cycles and the nonuniform temporal scaling of neural development across temperatures. Developmental temperature has notable effects on odor-driven behavior. Dissecting the circuit architecture and function of neurons in the olfactory pathway, we demonstrate that developmental temperature does not alter odor encoding in first- and second-order neurons, but it shifts the specificity of connections onto third-order neurons that mediate innate behaviors. We conclude that while some circuit computations are robust to the effects of developmental temperature on wiring, others exhibit phenotypic plasticity with possible adaptive advantages.
Collapse
Affiliation(s)
| | | | | | | | | | - Carlotta Martelli
- Johannes Gutenberg University, Mainz, Germany
- Institute for Quantitative and Computational Biosciences, Mainz, Germany
| |
Collapse
|
3
|
Berns MMM, Yildiz M, Winkelmann S, Walter AM. Independently engaging protein tethers of different length enhance synaptic vesicle trafficking to the plasma membrane. J Physiol 2025. [PMID: 39808523 DOI: 10.1113/jp286651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Synaptic vesicle (SV) trafficking toward the plasma membrane (PM) and subsequent SV maturation are essential for neurotransmitter release. These processes, including SV docking and priming, are co-ordinated by various proteins, such as SNAREs, Munc13 and synaptotagmin (Syt), which connect (tether) the SV to the PM. Here, we investigated how tethers of varying lengths mediate SV docking using a simplified mathematical model. The heights of the three tether types, as estimated from the structures of the SNARE complex, Munc13 and Syt, defined the SV-PM distance ranges for tether formation. Geometric considerations linked SV-PM distances to the probability and rate of tether formation. We assumed that SV tethering constrains SV motility and that multiple tethers are associated by independent interactions. The model predicted that forming multiple tethers favours shorter SV-PM distances. Although tethers acted independently in the model, their geometrical properties often caused their sequential assembly, from longer ones (Munc13/Syt), which accelerated SV movement towards the PM, to shorter ones (SNAREs), which stabilized PM-proximal SVs. Modifying tether lengths or numbers affected SV trafficking. The independent implementation of tethering proteins enabled their selective removal to mimic gene knockout (KO) situations. This showed that simulated SV-PM distance distributions qualitatively aligned with published electron microscopy studies upon removal of SNARE and Syt tethers, whereas Munc13 KO data were best approximated when assuming additional disruption of SNARE tethers. Thus, although salient features of SV docking can be accounted for by independent tethering alone, our results suggest that functional tether interactions not yet featured in our model are crucial for biological function. KEY POINTS: A mathematical model describing the role of synaptic protein tethers to localize transmitter-containing vesicles is developed based on geometrical considerations and structural information of synaptotagmin, Munc13 and SNARE proteins. Vesicle movement, along with tether association and dissociation, are modelled as stochastic processes, with tethers functioning independently of each other. Multiple tethers cooperate to recruit vesicles to the plasma membrane and keep them there: Munc13 and Syt as the longer tethers accelerate the movement towards the membrane, whereas short SNARE tethers stabilize them there. Model predictions for situations in which individual tethers are removed agree with the results from experimental studies upon gene knockout. Changing tether length or copy numbers affects vesicle trafficking and steady-state distributions.
Collapse
Affiliation(s)
- Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Alexander M Walter
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Lützkendorf J, Matkovic-Rachid T, Liu S, Götz T, Gao L, Turrel O, Maglione M, Grieger M, Putignano S, Ramesh N, Ghelani T, Neumann A, Gimber N, Schmoranzer J, Stawrakakis A, Brence B, Baum D, Ludwig K, Heine M, Mielke T, Liu F, Walter AM, Wahl MC, Sigrist SJ. Blobby is a synaptic active zone assembly protein required for memory in Drosophila. Nat Commun 2025; 16:271. [PMID: 39747038 PMCID: PMC11696761 DOI: 10.1038/s41467-024-55382-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
At presynaptic active zones (AZs), scaffold proteins are critical for coordinating synaptic vesicle release and forming essential nanoarchitectures. However, regulatory principles steering AZ scaffold assembly, function, and plasticity remain insufficiently understood. We here identify an additional Drosophila AZ protein, "Blobby", essential for proper AZ nano-organization. Blobby biochemically associates with the ELKS family AZ scaffold protein Bruchpilot (BRP) and integrates into newly forming AZs. Loss of Blobby results in fewer AZs forming, ectopic AZ scaffold protein accumulations ("blobs") and disrupts nanoscale architecture of the BRP-AZ scaffold. Functionally, blobby mutants show diminished evoked synaptic currents due to reduced synaptic vesicle release probability and fewer functional release sites. Blobby is also present in adult brain synapses, and post-developmental knockdown of Blobby in the mushroom body impairs olfactory aversive memory consolidation. Thus, our analysis identifies an additional layer of AZ regulation critical for developmental AZ assembly but also for AZ-mediated plasticity controlling behavior.
Collapse
Affiliation(s)
- J Lützkendorf
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Matkovic-Rachid
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Liu
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - T Götz
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - L Gao
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - O Turrel
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - M Maglione
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, SupraFAB, Berlin, Germany
| | - M Grieger
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - S Putignano
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Ramesh
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - T Ghelani
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A Neumann
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - N Gimber
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - J Schmoranzer
- Charité- Universitätsmedizin, Advanced Medical Bioimaging Core Facility, Berlin, Germany
| | - A Stawrakakis
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany
| | - B Brence
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - D Baum
- Zuse Institute Berlin, Department of Visual and Data-Centric Computing, Berlin, Germany
| | - Kai Ludwig
- Freie Universität Berlin, Institut für Chemie and Biochemie, Forschungszentrum für Elektronenmikroskopie, Berlin, Germany
| | - M Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - T Mielke
- Max Planck Institute for Molecular Genetics, Berlin, Microscopy and Cryo-Electron Microscopy Service Group, Berlin, Germany
| | - F Liu
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
| | - A M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin-Buch, Berlin, Germany
- University of Copenhagen, Department of Neuroscience, Copenhagen, Denmark
| | - M C Wahl
- Freie Universität Berlin, Institute of Chemistry and Biochemistry/Structural Biochemistry, Berlin, Germany
| | - S J Sigrist
- Freie Universität Berlin, Institute for Biology and Genetics, Berlin, Germany.
- Charité Universitätsmedizin, NeuroCure Cluster of Excellence, Charitéplatz, Berlin, Germany.
| |
Collapse
|
5
|
Midorikawa M, Sakamoto H, Nakamura Y, Hirose K, Miyata M. Developmental refinement of the active zone nanotopography and axon wiring at the somatosensory thalamus. Cell Rep 2024; 43:114770. [PMID: 39321021 DOI: 10.1016/j.celrep.2024.114770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024] Open
Abstract
Functional refinement of neural circuits is a crucial developmental process in the brain. However, how synaptic maturation and axon wiring proceed cooperatively to establish reliable signal transmission is unclear. Here, we combined nanotopography of release machinery at the active zone (AZ), nanobiophysics of neurotransmitter release, and single-neuron reconstruction of axon arbors of lemniscal fibers (LFs) in the developing mouse somatosensory thalamus. With development, the cluster of Cav2.1 enlarges and translocates closer to vesicle release sites inside the bouton, and LFs drastically shrink their arbors and form larger boutons on the perisomatic region of target neurons. Experimentally constrained simulations show that the nanotopography of mature synapses enables not only rapid vesicular release but also reliable transmission following repetitive firing. Sensory deprivation impairs the developmental shift of molecular nanotopography and axon wiring. Thus, we uncovered the cooperative nanotopographical and morphological mechanisms underlying the developmental establishment of reliable synaptic transmission.
Collapse
Affiliation(s)
- Mitsuharu Midorikawa
- Division of Biofunction, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| | - Hirokazu Sakamoto
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukihiro Nakamura
- Department of Pharmacology, Jikei University School of Medicine, Tokyo, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
6
|
Slater CR. Neuromuscular Transmission in a Biological Context. Compr Physiol 2024; 14:5641-5702. [PMID: 39382166 DOI: 10.1002/cphy.c240001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Neuromuscular transmission is the process by which motor neurons activate muscle contraction and thus plays an essential role in generating the purposeful body movements that aid survival. While many features of this process are common throughout the Animal Kingdom, such as the release of transmitter in multimolecular "quanta," and the response to it by opening ligand-gated postsynaptic ion channels, there is also much diversity between and within species. Much of this diversity is associated with specialization for either slow, sustained movements such as maintain posture or fast but brief movements used during escape or prey capture. In invertebrates, with hydrostatic and exoskeletons, most motor neurons evoke graded depolarizations of the muscle which cause graded muscle contractions. By contrast, vertebrate motor neurons trigger action potentials in the muscle fibers which give rise to all-or-none contractions. The properties of neuromuscular transmission, in particular the intensity and persistence of transmitter release, reflect these differences. Neuromuscular transmission varies both between and within individual animals, which often have distinct tonic and phasic subsystems. Adaptive plasticity of neuromuscular transmission, on a range of time scales, occurs in many species. This article describes the main steps in neuromuscular transmission and how they vary in a number of "model" species, including C. elegans , Drosophila , zebrafish, mice, and humans. © 2024 American Physiological Society. Compr Physiol 14:5641-5702, 2024.
Collapse
|
7
|
Medeiros AT, Gratz SJ, Delgado A, Ritt JT, O'Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. eLife 2024; 12:RP88412. [PMID: 39291956 PMCID: PMC11410372 DOI: 10.7554/elife.88412] [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] [Indexed: 09/19/2024] Open
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- Audrey T Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, United States
| | - Scott J Gratz
- Department of Neuroscience, Brown University, Providence, United States
| | - Ambar Delgado
- Department of Neuroscience, Brown University, Providence, United States
| | - Jason T Ritt
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| | - Kate M O'Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, United States
- Department of Neuroscience, Brown University, Providence, United States
- Carney Institute for Brain Science, Brown University, Providence, United States
| |
Collapse
|
8
|
Michael AH, Hana TA, Mousa VG, Ormerod KG. Muscle-fiber specific genetic manipulation of Drosophila sallimus severely impacts neuromuscular development, morphology, and physiology. Front Physiol 2024; 15:1429317. [PMID: 39351283 PMCID: PMC11439786 DOI: 10.3389/fphys.2024.1429317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
The ability of skeletal muscles to contract is derived from the unique genes and proteins expressed within muscles, most notably myofilaments and elastic proteins. Here we investigated the role of the sallimus (sls) gene, which encodes a structural homologue of titin, in regulating development, structure, and function of Drosophila melanogaster. Knockdown of sls using RNA interference (RNAi) in all body-wall muscle fibers resulted in embryonic lethality. A screen for muscle-specific drivers revealed a Gal4 line that expresses in a single larval body wall muscle in each abdominal hemisegment. Disrupting sls expression in single muscle fibers did not impact egg or larval viability nor gross larval morphology but did significantly alter the morphology of individual muscle fibers. Ultrastructural analysis of individual muscles revealed significant changes in organization. Surprisingly, muscle-cell specific disruption of sls also severely impacted neuromuscular junction (NMJ) formation. The extent of motor-neuron (MN) innervation along disrupted muscles was significantly reduced along with the number of glutamatergic boutons, in MN-Is and MN-Ib. Electrophysiological recordings revealed a 40% reduction in excitatory junctional potentials correlating with the extent of motor neuron loss. Analysis of active zone (AZ) composition revealed changes in presynaptic scaffolding protein (brp) abundance, but no changes in postsynaptic glutamate receptors. Ultrastructural changes in muscle and NMJ development at these single muscle fibers were sufficient to lead to observable changes in neuromuscular transduction and ultimately, locomotory behavior. Collectively, the data demonstrate that sls mediates critical aspects of muscle and NMJ development and function, illuminating greater roles for sls/titin.
Collapse
Affiliation(s)
| | | | | | - Kiel G. Ormerod
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| |
Collapse
|
9
|
Chien C, He K, Perry S, Tchitchkan E, Han Y, Li X, Dickman D. Distinct input-specific mechanisms enable presynaptic homeostatic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612361. [PMID: 39314403 PMCID: PMC11419068 DOI: 10.1101/2024.09.10.612361] [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: 09/25/2024]
Abstract
Synapses are endowed with the flexibility to change through experience, but must be sufficiently stable to last a lifetime. This tension is illustrated at the Drosophila neuromuscular junction (NMJ), where two motor inputs that differ in structural and functional properties co-innervate most muscles to coordinate locomotion. To stabilize NMJ activity, motor neurons augment neurotransmitter release following diminished postsynaptic glutamate receptor functionality, termed presynaptic homeostatic potentiation (PHP). How these distinct inputs contribute to PHP plasticity remains enigmatic. We have used a botulinum neurotoxin to selectively silence each input and resolve their roles in PHP, demonstrating that PHP is input-specific: Chronic (genetic) PHP selectively targets the tonic MN-Ib, where active zone remodeling enhances Ca2+ influx to promote increased glutamate release. In contrast, acute (pharmacological) PHP selectively increases vesicle pools to potentiate phasic MN-Is. Thus, distinct homeostatic modulations in active zone nanoarchitecture, vesicle pools, and Ca2+ influx collaborate to enable input-specific PHP expression.
Collapse
Affiliation(s)
- Chun Chien
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Kaikai He
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Sarah Perry
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Elizabeth Tchitchkan
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| | - Yifu Han
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Xiling Li
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
- USC Neuroscience Graduate Program
| | - Dion Dickman
- University of Southern California, Department of Neurobiology, Los Angeles, CA USA
| |
Collapse
|
10
|
Xiong A, Richmond JE, Kim H. Presynaptic neurons self-tune by inversely coupling neurotransmitter release with the abundance of CaV2 voltage-gated Ca 2+ channels. Proc Natl Acad Sci U S A 2024; 121:e2404969121. [PMID: 39172783 PMCID: PMC11363341 DOI: 10.1073/pnas.2404969121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
The abundance of CaV2 voltage-gated calcium channels is linked to presynaptic homeostatic plasticity (PHP), a process that recalibrates synaptic strength to maintain the stability of neural circuits. However, the molecular and cellular mechanisms governing PHP and CaV2 channels are not completely understood. Here, we uncover a previously not described form of PHP in Caenorhabditis elegans, revealing an inverse regulatory relationship between the efficiency of neurotransmitter release and the abundance of UNC-2/CaV2 channels. Gain-of-function unc-2SL(S240L) mutants, which carry a mutation analogous to the one causing familial hemiplegic migraine type 1 in humans, showed markedly reduced channel abundance despite increased channel functionality. Reducing synaptic release in these unc-2SL(S240L) mutants restored channel levels to those observed in wild-type animals. Conversely, loss-of-function unc-2DA(D726A) mutants, which harbor the D726A mutation in the channel pore, exhibited a marked increase in channel abundance. Enhancing synaptic release in unc-2DA mutants reversed this increase in channel levels. Importantly, this homeostatic regulation of UNC-2 channel levels is accompanied by the structural remodeling of the active zone (AZ); specifically, unc-2DA mutants, which exhibit increased channel abundance, showed parallel increases in select AZ proteins. Finally, our forward genetic screen revealed that WWP-1, a HECT family E3 ubiquitin ligase, is a key homeostatic mediator that removes UNC-2 from synapses. These findings highlight a self-tuning PHP regulating UNC-2/CaV2 channel abundance along with AZ reorganization, ensuring synaptic strength and stability.
Collapse
Affiliation(s)
- Ame Xiong
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois, Chicago, IL60607
| | - Hongkyun Kim
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
11
|
Fukata Y, Fukata M, MacGillavry HD, Nair D, Hosy E. Celebrating the Birthday of AMPA Receptor Nanodomains: Illuminating the Nanoscale Organization of Excitatory Synapses with 10 Nanocandles. J Neurosci 2024; 44:e2104232024. [PMID: 38839340 PMCID: PMC11154862 DOI: 10.1523/jneurosci.2104-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 06/07/2024] Open
Abstract
A decade ago, in 2013, and over the course of 4 summer months, three separate observations were reported that each shed light independently on a new molecular organization that fundamentally reshaped our perception of excitatory synaptic transmission (Fukata et al., 2013; MacGillavry et al., 2013; Nair et al., 2013). This discovery unveiled an intricate arrangement of AMPA-type glutamate receptors and their principal scaffolding protein PSD-95, at synapses. This breakthrough was made possible, thanks to advanced super-resolution imaging techniques. It fundamentally changed our understanding of excitatory synaptic architecture and paved the way for a brand-new area of research. In this Progressions article, the primary investigators of the nanoscale organization of synapses have come together to chronicle the tale of their discovery. We recount the initial inquiry that prompted our research, the preceding studies that inspired our work, the technical obstacles that were encountered, and the breakthroughs that were made in the subsequent decade in the realm of nanoscale synaptic transmission. We review the new discoveries made possible by the democratization of super-resolution imaging techniques in the field of excitatory synaptic physiology and architecture, first by the extension to other glutamate receptors and to presynaptic proteins and then by the notion of trans-synaptic organization. After describing the organizational modifications occurring in various pathologies, we discuss briefly the latest technical developments made possible by super-resolution imaging and emerging concepts in synaptic physiology.
Collapse
Affiliation(s)
- Yuko Fukata
- Division of Molecular and Cellular Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - Harold D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Eric Hosy
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, CNRS UMR5297, Bordeaux F-33000, France
| |
Collapse
|
12
|
Barti B, Dudok B, Kenesei K, Zöldi M, Miczán V, Balla GY, Zala D, Tasso M, Sagheddu C, Kisfali M, Tóth B, Ledri M, Vizi ES, Melis M, Barna L, Lenkei Z, Soltész I, Katona I. Presynaptic nanoscale components of retrograde synaptic signaling. SCIENCE ADVANCES 2024; 10:eado0077. [PMID: 38809980 PMCID: PMC11135421 DOI: 10.1126/sciadv.ado0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.
Collapse
Affiliation(s)
- Benjámin Barti
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Barna Dudok
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Departments of Neurology and Neuroscience, Baylor College of Medicine, 1 Baylor Plz, Houston, TX 77030, USA
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - Kata Kenesei
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miklós Zöldi
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Vivien Miczán
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Center, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Gyula Y. Balla
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
- Translational Behavioral Neuroscience Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Diana Zala
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Mariana Tasso
- Institute of Nanosystems, School of Bio and Nanotechnologies, National University of San Martín - CONICET, 25 de Mayo Ave., 1021 San Martín, Argentina
| | - Claudia Sagheddu
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - Máté Kisfali
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- BiTrial Ltd., Tállya st 23, H-1121 Budapest, Hungary
| | - Blanka Tóth
- Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Szt. Gellért square 4, H-1111 Budapest, Hungary
- Department of Molecular Biology, Semmelweis University, Üllői st 26, H-1085 Budapest, Hungary
| | - Marco Ledri
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
- Epilepsy Center, Department of Clinical Sciences, Faculty of Medicine, Lund University, Sölvegatan 17, BMC A11, 221 84 Lund, Sweden
| | - E. Sylvester Vizi
- Molecular Pharmacology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
| | - László Barna
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
| | - Zsolt Lenkei
- Université Paris Cité, INSERM, Institute of Psychiatry and Neurosciences of Paris, F-75014 Paris, France
| | - Iván Soltész
- Department of Neurosurgery, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA
| | - István Katona
- Department of Psychological and Brain Sciences, Indiana University Bloomington, 702 N Walnut Grove Ave, Bloomington, IN 47405-2204, USA
- Molecular Neurobiology Research Group, HUN-REN Institute of Experimental Medicine, Szigony st 43, H-1083 Budapest, Hungary
| |
Collapse
|
13
|
Medeiros AT, Gratz S, Delgado A, Ritt J, O’Connor-Giles KM. Ca 2+ channel and active zone protein abundance intersects with input-specific synapse organization to shape functional synaptic diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535290. [PMID: 37034654 PMCID: PMC10081318 DOI: 10.1101/2023.04.02.535290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Synaptic heterogeneity is a hallmark of nervous systems that enables complex and adaptable communication in neural circuits. To understand circuit function, it is thus critical to determine the factors that contribute to the functional diversity of synapses. We investigated the contributions of voltage-gated calcium channel (VGCC) abundance, spatial organization, and subunit composition to synapse diversity among and between synapses formed by two closely related Drosophila glutamatergic motor neurons with distinct neurotransmitter release probabilities (Pr). Surprisingly, VGCC levels are highly predictive of heterogeneous Pr among individual synapses of either low- or high-Pr inputs, but not between inputs. We find that the same number of VGCCs are more densely organized at high-Pr synapses, consistent with tighter VGCC-synaptic vesicle coupling. We generated endogenously tagged lines to investigate VGCC subunits in vivo and found that the α2δ-3 subunit Straightjacket along with the CAST/ELKS active zone (AZ) protein Bruchpilot, both key regulators of VGCCs, are less abundant at high-Pr inputs, yet positively correlate with Pr among synapses formed by either input. Consistently, both Straightjacket and Bruchpilot levels are dynamically increased across AZs of both inputs when neurotransmitter release is potentiated to maintain stable communication following glutamate receptor inhibition. Together, these findings suggest a model in which VGCC and AZ protein abundance intersects with input-specific spatial and molecular organization to shape the functional diversity of synapses.
Collapse
Affiliation(s)
- A. T. Medeiros
- Neuroscience Graduate Training Program, Brown University, Providence, RI
| | - S.J. Gratz
- Department of Neuroscience, Brown University, Providence, RI
| | - A. Delgado
- Department of Neuroscience, Brown University, Providence, RI
| | - J.T. Ritt
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| | - Kate M. O’Connor-Giles
- Neuroscience Graduate Training Program, Brown University, Providence, RI
- Department of Neuroscience, Brown University, Providence, RI
- Carney Institute for Brain Science, Brown University, Providence, RI
| |
Collapse
|
14
|
Turrel O, Gao L, Sigrist SJ. Presynaptic regulators in memory formation. Learn Mem 2024; 31:a054013. [PMID: 38862173 PMCID: PMC11199941 DOI: 10.1101/lm.054013.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 06/13/2024]
Abstract
The intricate molecular and structural sequences guiding the formation and consolidation of memories within neuronal circuits remain largely elusive. In this study, we investigate the roles of two pivotal presynaptic regulators, the small GTPase Rab3, enriched at synaptic vesicles, and the cell adhesion protein Neurexin-1, in the formation of distinct memory phases within the Drosophila mushroom body Kenyon cells. Our findings suggest that both proteins play crucial roles in memory-supporting processes within the presynaptic terminal, operating within distinct plasticity modules. These modules likely encompass remodeling and maturation of existing active zones (AZs), as well as the formation of new AZs.
Collapse
Affiliation(s)
- Oriane Turrel
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Lili Gao
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology, Genetics, Freie Universität Berlin, 14195 Berlin, Germany
- Cluster of Excellence NeuroCure, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
15
|
Pribbenow C, Owald D. Skewing information flow through pre- and postsynaptic plasticity in the mushroom bodies of Drosophila. Learn Mem 2024; 31:a053919. [PMID: 38876487 PMCID: PMC11199954 DOI: 10.1101/lm.053919.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/26/2024] [Indexed: 06/16/2024]
Abstract
Animal brains need to store information to construct a representation of their environment. Knowledge of what happened in the past allows both vertebrates and invertebrates to predict future outcomes by recalling previous experience. Although invertebrate and vertebrate brains share common principles at the molecular, cellular, and circuit-architectural levels, there are also obvious differences as exemplified by the use of acetylcholine versus glutamate as the considered main excitatory neurotransmitters in the respective central nervous systems. Nonetheless, across central nervous systems, synaptic plasticity is thought to be a main substrate for memory storage. Therefore, how brain circuits and synaptic contacts change following learning is of fundamental interest for understanding brain computations tied to behavior in any animal. Recent progress has been made in understanding such plastic changes following olfactory associative learning in the mushroom bodies (MBs) of Drosophila A current framework of memory-guided behavioral selection is based on the MB skew model, in which antagonistic synaptic pathways are selectively changed in strength. Here, we review insights into plasticity at dedicated Drosophila MB output pathways and update what is known about the plasticity of both pre- and postsynaptic compartments of Drosophila MB neurons.
Collapse
Affiliation(s)
- Carlotta Pribbenow
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - David Owald
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Einstein Center for Neurosciences Berlin, 10117 Berlin, Germany
- NeuroCure, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
16
|
Wang T, Frank CA. Using Electrophysiology to Study Homeostatic Plasticity at the Drosophila Neuromuscular Junction. Cold Spring Harb Protoc 2024:pdb.top108393. [PMID: 38688539 PMCID: PMC11522024 DOI: 10.1101/pdb.top108393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The Drosophila melanogaster neuromuscular junction (NMJ) is a superb system for studying synapse function. Beyond that, the NMJ is also great for studying forms of synaptic plasticity. Over the last 25 years, Drosophila NMJ neuroscientists have pioneered understanding of a form of plasticity called homeostatic synaptic plasticity, which imparts functional stability on synaptic connections. The reason is straightforward: The NMJ has a robust capacity for stability. Moreover, many strategies that the NMJ uses to maintain appropriate levels of function are mirrored at other metazoan synapses. Here, we introduce core approaches that neurophysiologists use to study homeostatic synaptic plasticity at the peripheral Drosophila NMJ. We focus on methods to study a specific form of homeostatic plasticity termed presynaptic homeostatic potentiation (PHP), which is the most well-characterized one. Other forms such as presynaptic homeostatic depression and developmental forms of homeostasis are briefly discussed. Finally, we share lists of several dozen factors and conditions known to influence the execution of PHP.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, D.C. 20007, USA
| | - C Andrew Frank
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| |
Collapse
|
17
|
Beckers CJ, Mrestani A, Komma F, Dannhäuser S. Versatile Endogenous Editing of GluRIIA in Drosophila melanogaster. Cells 2024; 13:323. [PMID: 38391936 PMCID: PMC10887371 DOI: 10.3390/cells13040323] [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: 01/15/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Glutamate receptors at the postsynaptic side translate neurotransmitter release from presynapses into postsynaptic excitation. They play a role in many forms of synaptic plasticity, e.g., homeostatic scaling of the receptor field, activity-dependent synaptic plasticity and the induction of presynaptic homeostatic potentiation (PHP). The latter process has been extensively studied at Drosophila melanogaster neuromuscular junctions (NMJs). The genetic removal of the glutamate receptor subunit IIA (GluRIIA) leads to an induction of PHP at the synapse. So far, mostly imprecise knockouts of the GluRIIA gene have been utilized. Furthermore, mutated and tagged versions of GluRIIA have been examined in the past, but most of these constructs were not expressed under endogenous regulatory control or involved the mentioned imprecise GluRIIA knockouts. We performed CRISPR/Cas9-assisted gene editing at the endogenous locus of GluRIIA. This enabled the investigation of the endogenous expression pattern of GluRIIA using tagged constructs with an EGFP and an ALFA tag for super-resolution immunofluorescence imaging, including structured illumination microscopy (SIM) and direct stochastic optical reconstruction microscopy (dSTORM). All GluRIIA constructs exhibited full functionality and PHP could be induced by philanthotoxin at control levels. By applying hierarchical clustering algorithms to analyze the dSTORM data, we detected postsynaptic receptor cluster areas of ~0.15 µm2. Consequently, our constructs are suitable for ultrastructural analyses of GluRIIA.
Collapse
Affiliation(s)
- Constantin J. Beckers
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| | - Achmed Mrestani
- Department of Neurology, University of Leipzig Medical Center, D-04103 Leipzig, Germany;
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, D-04103 Leipzig, Germany
| | - Fabian Komma
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| |
Collapse
|
18
|
Mrestani A, Dannhäuser S, Pauli M, Kollmannsberger P, Hübsch M, Morris L, Langenhan T, Heckmann M, Paul MM. Nanoscaled RIM clustering at presynaptic active zones revealed by endogenous tagging. Life Sci Alliance 2023; 6:e202302021. [PMID: 37696575 PMCID: PMC10494931 DOI: 10.26508/lsa.202302021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/13/2023] Open
Abstract
Chemical synaptic transmission involves neurotransmitter release from presynaptic active zones (AZs). The AZ protein Rab-3-interacting molecule (RIM) is important for normal Ca2+-triggered release. However, its precise localization within AZs of the glutamatergic neuromuscular junctions of Drosophila melanogaster remains elusive. We used CRISPR/Cas9-assisted genome engineering of the rim locus to incorporate small epitope tags for targeted super-resolution imaging. A V5-tag, derived from simian virus 5, and an HA-tag, derived from human influenza virus, were N-terminally fused to the RIM Zinc finger. Whereas both variants are expressed in co-localization with the core AZ scaffold Bruchpilot, electrophysiological characterization reveals that AP-evoked synaptic release is disturbed in rimV5-Znf but not in rimHA-Znf In addition, rimHA-Znf synapses show intact presynaptic homeostatic potentiation. Combining super-resolution localization microscopy and hierarchical clustering, we detect ∼10 RIMHA-Znf subclusters with ∼13 nm diameter per AZ that are compacted and increased in numbers in presynaptic homeostatic potentiation.
Collapse
Affiliation(s)
- Achmed Mrestani
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Neurology, Leipzig University Medical Center, Leipzig, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Martha Hübsch
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lydia Morris
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Mila M Paul
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Zhang Y, Wang T, Cai Y, Cui T, Kuah M, Vicini S, Wang T. Role of α2δ-3 in regulating calcium channel localization at presynaptic active zones during homeostatic plasticity. Front Mol Neurosci 2023; 16:1253669. [PMID: 38025261 PMCID: PMC10662314 DOI: 10.3389/fnmol.2023.1253669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
The homeostatic modulation of synaptic transmission is an evolutionarily conserved mechanism that is critical for stabilizing the nervous system. At the Drosophila neuromuscular junction (NMJ), presynaptic homeostatic potentiation (PHP) compensates for impairments in postsynaptic glutamate receptors due to pharmacological blockade or genetic deletion. During PHP, there is an increase in presynaptic neurotransmitter release, counteracting postsynaptic changes and restoring excitation to baseline levels. Previous studies have shown that α2δ-3, an auxiliary subunit of voltage-gated calcium channels (VGCCs), is essential for both the rapid induction and sustained expression of PHP at the Drosophila NMJ. However, the molecular mechanisms by which α2δ-3 regulates neurotransmitter release during PHP remain to be elucidated. In this study, we utilized electrophysiological, confocal imaging, and super-resolution imaging approaches to explore how α2δ-3 regulates synaptic transmission during PHP. Our findings suggest that α2δ-3 governs PHP by controlling the localization of the calcium channel pore-forming α1 subunit at presynaptic release sites, or active zones. Moreover, we examined the role of two structural domains within α2δ-3 in regulating neurotransmitter release and calcium channel localization. Our results highlight that these domains in α2δ-3 serve distinct functions in controlling synaptic transmission and presynaptic calcium channel abundance, at baseline in the absence of perturbations and during PHP. In summary, our research offers compelling evidence that α2δ-3 is an indispensable signaling component for controlling calcium channel trafficking and stabilization in homeostatic plasticity.
Collapse
Affiliation(s)
- Yanfeng Zhang
- Department of Pediatric Neurology, First Hospital of Jilin University, Changchun, Jilin, China
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Ting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Yimei Cai
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Tao Cui
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Michelle Kuah
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Tingting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
20
|
Ramesh N, Escher M, Turrel O, Lützkendorf J, Matkovic T, Liu F, Sigrist SJ. An antagonism between Spinophilin and Syd-1 operates upstream of memory-promoting presynaptic long-term plasticity. eLife 2023; 12:e86084. [PMID: 37767892 PMCID: PMC10588984 DOI: 10.7554/elife.86084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
We still face fundamental gaps in understanding how molecular plastic changes of synapses intersect with circuit operation to define behavioral states. Here, we show that an antagonism between two conserved regulatory proteins, Spinophilin (Spn) and Syd-1, controls presynaptic long-term plasticity and the maintenance of olfactory memories in Drosophila. While Spn mutants could not trigger nanoscopic active zone remodeling under homeostatic challenge and failed to stably potentiate neurotransmitter release, concomitant reduction of Syd-1 rescued all these deficits. The Spn/Syd-1 antagonism converged on active zone close F-actin, and genetic or acute pharmacological depolymerization of F-actin rescued the Spn deficits by allowing access to synaptic vesicle release sites. Within the intrinsic mushroom body neurons, the Spn/Syd-1 antagonism specifically controlled olfactory memory stabilization but not initial learning. Thus, this evolutionarily conserved protein complex controls behaviorally relevant presynaptic long-term plasticity, also observed in the mammalian brain but still enigmatic concerning its molecular mechanisms and behavioral relevance.
Collapse
Affiliation(s)
- Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Marc Escher
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Oriane Turrel
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | | | - Tanja Matkovic
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| | - Fan Liu
- Leibniz-Forschungsinstitut für Molekulare PharmakologieBerlinGermany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität BerlinBerlinGermany
| |
Collapse
|
21
|
Fukaya R, Miyano R, Hirai H, Sakaba T. Mechanistic insights into cAMP-mediated presynaptic potentiation at hippocampal mossy fiber synapses. Front Cell Neurosci 2023; 17:1237589. [PMID: 37519634 PMCID: PMC10372368 DOI: 10.3389/fncel.2023.1237589] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
Presynaptic plasticity is an activity-dependent change in the neurotransmitter release and plays a key role in dynamic modulation of synaptic strength. Particularly, presynaptic potentiation mediated by cyclic adenosine monophosphate (cAMP) is widely seen across the animals and thought to contribute to learning and memory. Hippocampal mossy fiber-CA3 pyramidal cell synapses have been used as a model because of robust presynaptic potentiation in short- and long-term forms. Moreover, direct presynaptic recordings from large mossy fiber terminals allow one to dissect the potentiation mechanisms. Recently, super-resolution microscopy and flash-and-freeze electron microscopy have revealed the localizations of release site molecules and synaptic vesicles during the potentiation at a nanoscale, identifying the molecular mechanisms of the potentiation. Incorporating these growing knowledges, we try to present plausible mechanisms underlying the cAMP-mediated presynaptic potentiation.
Collapse
Affiliation(s)
- Ryota Fukaya
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Rinako Miyano
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
22
|
Jusyte M, Blaum N, Böhme MA, Berns MMM, Bonard AE, Vámosi ÁB, Pushpalatha KV, Kobbersmed JRL, Walter AM. Unc13A dynamically stabilizes vesicle priming at synaptic release sites for short-term facilitation and homeostatic potentiation. Cell Rep 2023; 42:112541. [PMID: 37243591 DOI: 10.1016/j.celrep.2023.112541] [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: 11/08/2022] [Revised: 03/10/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023] Open
Abstract
Presynaptic plasticity adjusts neurotransmitter (NT) liberation. Short-term facilitation (STF) tunes synapses to millisecond repetitive activation, while presynaptic homeostatic potentiation (PHP) of NT release stabilizes transmission over minutes. Despite different timescales of STF and PHP, our analysis of Drosophila neuromuscular junctions reveals functional overlap and shared molecular dependence on the release-site protein Unc13A. Mutating Unc13A's calmodulin binding domain (CaM-domain) increases baseline transmission while blocking STF and PHP. Mathematical modeling suggests that Ca2+/calmodulin/Unc13A interaction plastically stabilizes vesicle priming at release sites and that CaM-domain mutation causes constitutive stabilization, thereby blocking plasticity. Labeling the functionally essential Unc13A MUN domain reveals higher STED microscopy signals closer to release sites following CaM-domain mutation. Acute phorbol ester treatment similarly enhances NT release and blocks STF/PHP in synapses expressing wild-type Unc13A, while CaM-domain mutation occludes this, indicating common downstream effects. Thus, Unc13A regulatory domains integrate signals across timescales to switch release-site participation for synaptic plasticity.
Collapse
Affiliation(s)
- Meida Jusyte
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Natalie Blaum
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mathias A Böhme
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Manon M M Berns
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Alix E Bonard
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Ábel B Vámosi
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | | | - Janus R L Kobbersmed
- Department of Mathematical Sciences, University of Copenhagen, Copenhagen, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alexander M Walter
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany; Einstein Center for Neurosciences Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
He K, Han Y, Li X, Hernandez RX, Riboul DV, Feghhi T, Justs KA, Mahneva O, Perry S, Macleod GT, Dickman D. Physiologic and Nanoscale Distinctions Define Glutamatergic Synapses in Tonic vs Phasic Neurons. J Neurosci 2023; 43:4598-4611. [PMID: 37221096 PMCID: PMC10286941 DOI: 10.1523/jneurosci.0046-23.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Neurons exhibit a striking degree of functional diversity, each one tuned to the needs of the circuitry in which it is embedded. A fundamental functional dichotomy occurs in activity patterns, with some neurons firing at a relatively constant "tonic" rate, while others fire in bursts, a "phasic" pattern. Synapses formed by tonic versus phasic neurons are also functionally differentiated, yet the bases of their distinctive properties remain enigmatic. A major challenge toward illuminating the synaptic differences between tonic and phasic neurons is the difficulty in isolating their physiological properties. At the Drosophila neuromuscular junction, most muscle fibers are coinnervated by two motor neurons: the tonic "MN-Ib" and phasic "MN-Is." Here, we used selective expression of a newly developed botulinum neurotoxin transgene to silence tonic or phasic motor neurons in Drosophila larvae of either sex. This approach highlighted major differences in their neurotransmitter release properties, including probability, short-term plasticity, and vesicle pools. Furthermore, Ca2+ imaging demonstrated ∼2-fold greater Ca2+ influx at phasic neuron release sites relative to tonic, along with an enhanced synaptic vesicle coupling. Finally, confocal and super-resolution imaging revealed that phasic neuron release sites are organized in a more compact arrangement, with enhanced stoichiometry of voltage-gated Ca2+ channels relative to other active zone scaffolds. These data suggest that distinctions in active zone nano-architecture and Ca2+ influx collaborate to differentially tune glutamate release at tonic versus phasic synaptic subtypes.SIGNIFICANCE STATEMENT "Tonic" and "phasic" neuronal subtypes, based on differential firing properties, are common across many nervous systems. Using a recently developed approach to selectively silence transmission from one of these two neurons, we reveal specialized synaptic functional and structural properties that distinguish these specialized neurons. This study provides important insights into how input-specific synaptic diversity is achieved, which could have implications for neurologic disorders that involve changes in synaptic function.
Collapse
Affiliation(s)
- Kaikai He
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
- USC Neuroscience Graduate Program, Los Angeles, California 90089
| | - Roberto X Hernandez
- Integrative Biology and Neuroscience Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
- International Max Planck Research School for Brain and Behavior, Jupiter, Florida 33458
| | - Danielle V Riboul
- Integrative Biology Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
| | - Touhid Feghhi
- Department of Physics, Florida Atlantic University, Boca Raton, Florida 33431
| | - Karlis A Justs
- Integrative Biology and Neuroscience Graduate Program, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida 33431
| | - Olena Mahneva
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida 33458
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
| | - Gregory T Macleod
- Wilkes Honors College, Florida Atlantic University, Jupiter, Florida 33458
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, Florida 33458
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
24
|
Rozenfeld E, Ehmann N, Manoim JE, Kittel RJ, Parnas M. Homeostatic synaptic plasticity rescues neural coding reliability. Nat Commun 2023; 14:2993. [PMID: 37225688 DOI: 10.1038/s41467-023-38575-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
To survive, animals must recognize reoccurring stimuli. This necessitates a reliable stimulus representation by the neural code. While synaptic transmission underlies the propagation of neural codes, it is unclear how synaptic plasticity can maintain coding reliability. By studying the olfactory system of Drosophila melanogaster, we aimed to obtain a deeper mechanistic understanding of how synaptic function shapes neural coding in the live, behaving animal. We show that the properties of the active zone (AZ), the presynaptic site of neurotransmitter release, are critical for generating a reliable neural code. Reducing neurotransmitter release probability of olfactory sensory neurons disrupts both neural coding and behavioral reliability. Strikingly, a target-specific homeostatic increase of AZ numbers rescues these defects within a day. These findings demonstrate an important role for synaptic plasticity in maintaining neural coding reliability and are of pathophysiological interest by uncovering an elegant mechanism through which the neural circuitry can counterbalance perturbations.
Collapse
Affiliation(s)
- Eyal Rozenfeld
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nadine Ehmann
- Department of Animal Physiology, Institute of Biology, Leipzig University, 04103, Leipzig, Germany
| | - Julia E Manoim
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Robert J Kittel
- Department of Animal Physiology, Institute of Biology, Leipzig University, 04103, Leipzig, Germany.
| | - Moshe Parnas
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
25
|
McMullen E, Hertenstein H, Strassburger K, Deharde L, Brankatschk M, Schirmeier S. Glycolytically impaired Drosophila glial cells fuel neural metabolism via β-oxidation. Nat Commun 2023; 14:2996. [PMID: 37225684 DOI: 10.1038/s41467-023-38813-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/17/2023] [Indexed: 05/26/2023] Open
Abstract
Neuronal function is highly energy demanding and thus requires efficient and constant metabolite delivery by glia. Drosophila glia are highly glycolytic and provide lactate to fuel neuronal metabolism. Flies are able to survive for several weeks in the absence of glial glycolysis. Here, we study how Drosophila glial cells maintain sufficient nutrient supply to neurons under conditions of impaired glycolysis. We show that glycolytically impaired glia rely on mitochondrial fatty acid breakdown and ketone body production to nourish neurons, suggesting that ketone bodies serve as an alternate neuronal fuel to prevent neurodegeneration. We show that in times of long-term starvation, glial degradation of absorbed fatty acids is essential to ensure survival of the fly. Further, we show that Drosophila glial cells act as a metabolic sensor and can induce mobilization of peripheral lipid stores to preserve brain metabolic homeostasis. Our study gives evidence of the importance of glial fatty acid degradation for brain function, and survival, under adverse conditions in Drosophila.
Collapse
Affiliation(s)
- Ellen McMullen
- Department of Molecular Biology and Genetics, University of South Bohemia, České Budějovice, Czech Republic
| | - Helen Hertenstein
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Katrin Strassburger
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Leon Deharde
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Marko Brankatschk
- Biotechnologisches Zentrum, Technische Universität Dresden, Dresden, Germany.
| | - Stefanie Schirmeier
- Zoology and Animal Physiology, Faculty of Biology, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
26
|
Kudryashova I. Presynaptic Plasticity Is Associated with Actin Polymerization. BIOCHEMISTRY (MOSCOW) 2023; 88:392-403. [PMID: 37076285 DOI: 10.1134/s0006297923030082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Modulation of presynaptic short-term plasticity induced by actin polymerization was studied in rat hippocampal slices using the paired-pulse paradigm. Schaffer collaterals were stimulated with paired pulses with a 70-ms interstimulus interval every 30 s before and during perfusion with jasplakinolide, an activator of actin polymerization. Jasplakinolide application resulted in the increase in the amplitudes of CA3-CA1 responses (potentiation) accompanied by a decrease in the paired-pulse facilitation, suggesting induction of presynaptic modifications. Jasplakinolide-induced potentiation depended on the initial paired-pulse rate. These data indicate that the jasplakinolide-mediated changes in actin polymerization increased the probability of neurotransmitter release. Less typical for CA3-CA1 synapses responses, such as a very low paired-pulse ratio (close to 1 or even lower) or even paired-pulse depression, were affected differently. Thus, jasplakinolide caused potentiation of the second, but not the first response to the paired stimulus, which increased the paired-pulse ratio from 0.8 to 1.0 on average, suggesting a negative impact of jasplakinolide on the mechanisms promoting paired-pulse depression. In general, actin polymerization facilitated potentiation, although the patterns of potentiation differed depending on the initial synapse characteristics. We conclude that in addition to the increase in the neurotransmitter release probability, jasplakinolide induced other actin polymerization-dependent mechanisms, including those involved in the paired-pulse depression.
Collapse
Affiliation(s)
- Irina Kudryashova
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
27
|
Fukaya R, Hirai H, Sakamoto H, Hashimotodani Y, Hirose K, Sakaba T. Increased vesicle fusion competence underlies long-term potentiation at hippocampal mossy fiber synapses. SCIENCE ADVANCES 2023; 9:eadd3616. [PMID: 36812326 PMCID: PMC9946361 DOI: 10.1126/sciadv.add3616] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Presynaptic long-term potentiation (LTP) is thought to play an important role in learning and memory. However, the underlying mechanism remains elusive because of the difficulty of direct recording during LTP. Hippocampal mossy fiber synapses exhibit pronounced LTP of transmitter release after tetanic stimulation and have been used as a model of presynaptic LTP. Here, we induced LTP by optogenetic tools and applied direct presynaptic patch-clamp recordings. The action potential waveform and evoked presynaptic Ca2+ currents remained unchanged after LTP induction. Membrane capacitance measurements suggested higher release probability of synaptic vesicles without changing the number of release-ready vesicles after LTP induction. Synaptic vesicle replenishment was also enhanced. Furthermore, stimulated emission depletion microscopy suggested an increase in the numbers of Munc13-1 and RIM1 molecules within active zones. We propose that dynamic changes in the active zone components may be relevant for the increased fusion competence and synaptic vesicle replenishment during LTP.
Collapse
Affiliation(s)
- Ryota Fukaya
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
- Institute of Biology/Genetics, Freie Universität Berlin, 14195 Berlin, Germany
| | - Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Hirokazu Sakamoto
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuki Hashimotodani
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Kenzo Hirose
- Department of Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|
28
|
Ghelani T, Escher M, Thomas U, Esch K, Lützkendorf J, Depner H, Maglione M, Parutto P, Gratz S, Matkovic-Rachid T, Ryglewski S, Walter AM, Holcman D, O‘Connor Giles K, Heine M, Sigrist SJ. Interactive nanocluster compaction of the ELKS scaffold and Cacophony Ca 2+ channels drives sustained active zone potentiation. SCIENCE ADVANCES 2023; 9:eade7804. [PMID: 36800417 PMCID: PMC9937578 DOI: 10.1126/sciadv.ade7804] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 01/17/2023] [Indexed: 06/01/2023]
Abstract
At presynaptic active zones (AZs), conserved scaffold protein architectures control synaptic vesicle (SV) release by defining the nanoscale distribution and density of voltage-gated Ca2+ channels (VGCCs). While AZs can potentiate SV release in the minutes range, we lack an understanding of how AZ scaffold components and VGCCs engage into potentiation. We here establish dynamic, intravital single-molecule imaging of endogenously tagged proteins at Drosophila AZs undergoing presynaptic homeostatic potentiation. During potentiation, the numbers of α1 VGCC subunit Cacophony (Cac) increased per AZ, while their mobility decreased and nanoscale distribution compacted. These dynamic Cac changes depended on the interaction between Cac channel's intracellular carboxyl terminus and the membrane-close amino-terminal region of the ELKS-family protein Bruchpilot, whose distribution compacted drastically. The Cac-ELKS/Bruchpilot interaction was also needed for sustained AZ potentiation. Our single-molecule analysis illustrates how the AZ scaffold couples to VGCC nanoscale distribution and dynamics to establish a state of sustained potentiation.
Collapse
Affiliation(s)
- Tina Ghelani
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| | - Marc Escher
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Ulrich Thomas
- Department of Cellular Neurobiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Klara Esch
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Janine Lützkendorf
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Harald Depner
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Marta Maglione
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
- Institute for Chemistry and Biochemistry, SupraFAB, Freie Universität Berlin, Altensteinstr. 23a, 14195 Berlin, Germany
| | - Pierre Parutto
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Scott Gratz
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Tanja Matkovic-Rachid
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
| | - Stefanie Ryglewski
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Alexander M. Walter
- Molecular and Theoretical Neuroscience Leibniz-Forschungs Institut für Molekulare Pharmakologie (FMP) im CharitéCrossOver (CCO) Charité–University Medicine Berlin Charité Campus Mitte, Charité Platz, 110117 Berlin, Germany
- Department of Neuroscience, University of Copenhagen, Copenhagen 2200, Denmark
| | - David Holcman
- Group of Applied Mathematics and Computational Biology, IBENS, Ecole Normale Superieure, Paris, France
- Churchill College, University of Cambridge, Cambridge CB3 0DS, UK
| | - Kate O‘Connor Giles
- Department of Neuroscience, Brown University, Providence, RI 02912, USA
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118 Magdeburg, Germany
| | - Stephan J. Sigrist
- Institute for Biology and Genetics, Freie Universität Berlin, Takustraße 6, 14195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
29
|
Mrestani A, Lichter K, Sirén AL, Heckmann M, Paul MM, Pauli M. Single-Molecule Localization Microscopy of Presynaptic Active Zones in Drosophila melanogaster after Rapid Cryofixation. Int J Mol Sci 2023; 24:ijms24032128. [PMID: 36768451 PMCID: PMC9917252 DOI: 10.3390/ijms24032128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Single-molecule localization microscopy (SMLM) greatly advances structural studies of diverse biological tissues. For example, presynaptic active zone (AZ) nanotopology is resolved in increasing detail. Immunofluorescence imaging of AZ proteins usually relies on epitope preservation using aldehyde-based immunocompetent fixation. Cryofixation techniques, such as high-pressure freezing (HPF) and freeze substitution (FS), are widely used for ultrastructural studies of presynaptic architecture in electron microscopy (EM). HPF/FS demonstrated nearer-to-native preservation of AZ ultrastructure, e.g., by facilitating single filamentous structures. Here, we present a protocol combining the advantages of HPF/FS and direct stochastic optical reconstruction microscopy (dSTORM) to quantify nanotopology of the AZ scaffold protein Bruchpilot (Brp) at neuromuscular junctions (NMJs) of Drosophila melanogaster. Using this standardized model, we tested for preservation of Brp clusters in different FS protocols compared to classical aldehyde fixation. In HPF/FS samples, presynaptic boutons were structurally well preserved with ~22% smaller Brp clusters that allowed quantification of subcluster topology. In summary, we established a standardized near-to-native preparation and immunohistochemistry protocol for SMLM analyses of AZ protein clusters in a defined model synapse. Our protocol could be adapted to study protein arrangements at single-molecule resolution in other intact tissue preparations.
Collapse
Affiliation(s)
- Achmed Mrestani
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
- Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Katharina Lichter
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Anna-Leena Sirén
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
- Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
- Correspondence:
| | - Manfred Heckmann
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Mila M. Paul
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
- Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Wurzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute for Physiology, University of Würzburg, 97070 Würzburg, Germany
| |
Collapse
|
30
|
Armstrong NS, Frank CA. The calcineurin regulator Sarah enables distinct forms of homeostatic plasticity at the Drosophila neuromuscular junction. Front Synaptic Neurosci 2023; 14:1033743. [PMID: 36685082 PMCID: PMC9846150 DOI: 10.3389/fnsyn.2022.1033743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction: The ability of synapses to maintain physiological levels of evoked neurotransmission is essential for neuronal stability. A variety of perturbations can disrupt neurotransmission, but synapses often compensate for disruptions and work to stabilize activity levels, using forms of homeostatic synaptic plasticity. Presynaptic homeostatic potentiation (PHP) is one such mechanism. PHP is expressed at the Drosophila melanogaster larval neuromuscular junction (NMJ) synapse, as well as other NMJs. In PHP, presynaptic neurotransmitter release increases to offset the effects of impairing muscle transmitter receptors. Prior Drosophila work has studied PHP using different ways to perturb muscle receptor function-either acutely (using pharmacology) or chronically (using genetics). Some of our prior data suggested that cytoplasmic calcium signaling was important for expression of PHP after genetic impairment of glutamate receptors. Here we followed up on that observation. Methods: We used a combination of transgenic Drosophila RNA interference and overexpression lines, along with NMJ electrophysiology, synapse imaging, and pharmacology to test if regulators of the calcium/calmodulin-dependent protein phosphatase calcineurin are necessary for the normal expression of PHP. Results: We found that either pre- or postsynaptic dysregulation of a Drosophila gene regulating calcineurin, sarah (sra), blocks PHP. Tissue-specific manipulations showed that either increases or decreases in sra expression are detrimental to PHP. Additionally, pharmacologically and genetically induced forms of expression of PHP are functionally separable depending entirely upon which sra genetic manipulation is used. Surprisingly, dual-tissue pre- and postsynaptic sra knockdown or overexpression can ameliorate PHP blocks revealed in single-tissue experiments. Pharmacological and genetic inhibition of calcineurin corroborated this latter finding. Discussion: Our results suggest tight calcineurin regulation is needed across multiple tissue types to stabilize peripheral synaptic outputs.
Collapse
Affiliation(s)
- Noah S. Armstrong
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, United States,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, United States,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States,*Correspondence: C. Andrew Frank
| |
Collapse
|
31
|
Stone A, Cujic O, Rowlett A, Aderhold S, Savage E, Graham B, Steinert JR. Triose-phosphate isomerase deficiency is associated with a dysregulation of synaptic vesicle recycling in Drosophila melanogaster. Front Synaptic Neurosci 2023; 15:1124061. [PMID: 36926383 PMCID: PMC10011161 DOI: 10.3389/fnsyn.2023.1124061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
Introduction Numerous neurodegenerative diseases are associated with neuronal dysfunction caused by increased redox stress, often linked to aberrant production of redox-active molecules such as nitric oxide (NO) or oxygen free radicals. One such protein affected by redox-mediated changes is the glycolytic enzyme triose-phosphate isomerase (TPI), which has been shown to undergo 3-nitrotyrosination (a NO-mediated post-translational modification) rendering it inactive. The resulting neuronal changes caused by this modification are not well understood. However, associated glycation-induced cytotoxicity has been reported, thus potentially causing neuronal and synaptic dysfunction via compromising synaptic vesicle recycling. Methods This work uses Drosophila melanogaster to identify the impacts of altered TPI activity on neuronal physiology, linking aberrant TPI function and redox stress to neuronal defects. We used Drosophila mutants expressing a missense allele of the TPI protein, M81T, identified in a previous screen and resulting in an inactive mutant of the TPI protein (TPIM81T , wstd1). We assessed synaptic physiology at the glutamatergic Drosophila neuromuscular junction (NMJ), synapse morphology and behavioural phenotypes, as well as impacts on longevity. Results Electrophysiological recordings of evoked and spontaneous excitatory junctional currents, alongside high frequency train stimulations and recovery protocols, were applied to investigate synaptic depletion and subsequent recovery. Single synaptic currents were unaltered in the presence of the wstd1 mutation, but frequencies of spontaneous events were reduced. Wstd1 larvae also showed enhanced vesicle depletion rates at higher frequency stimulation, and subsequent recovery times for evoked synaptic responses were prolonged. A computational model showed that TPI mutant larvae exhibited a significant decline in activity-dependent vesicle recycling, which manifests itself as increased recovery times for the readily-releasable vesicle pool. Confocal images of NMJs showed no morphological or developmental differences between wild-type and wstd1 but TPI mutants exhibited learning impairments as assessed by olfactory associative learning assays. Discussion Our data suggests that the wstd1 phenotype is partially due to altered vesicle dynamics, involving a reduced vesicle pool replenishment, and altered endo/exocytosis processes. This may result in learning and memory impairments and neuronal dysfunction potentially also presenting a contributing factor to other reported neuronal phenotypes.
Collapse
Affiliation(s)
- Aelfwin Stone
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Oliver Cujic
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Angel Rowlett
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Sophia Aderhold
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Emma Savage
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Bruce Graham
- Division of Computing Science and Mathematics, Faculty of Natural Sciences, University of Stirling, Stirling, United Kingdom
| | - Joern R Steinert
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
32
|
Dannhäuser S, Mrestani A, Gundelach F, Pauli M, Komma F, Kollmannsberger P, Sauer M, Heckmann M, Paul MM. Endogenous tagging of Unc-13 reveals nanoscale reorganization at active zones during presynaptic homeostatic potentiation. Front Cell Neurosci 2022; 16:1074304. [PMID: 36589286 PMCID: PMC9797049 DOI: 10.3389/fncel.2022.1074304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Neurotransmitter release at presynaptic active zones (AZs) requires concerted protein interactions within a dense 3D nano-hemisphere. Among the complex protein meshwork the (M)unc-13 family member Unc-13 of Drosophila melanogaster is essential for docking of synaptic vesicles and transmitter release. Methods We employ minos-mediated integration cassette (MiMIC)-based gene editing using GFSTF (EGFP-FlAsH-StrepII-TEV-3xFlag) to endogenously tag all annotated Drosophila Unc-13 isoforms enabling visualization of endogenous Unc-13 expression within the central and peripheral nervous system. Results and discussion Electrophysiological characterization using two-electrode voltage clamp (TEVC) reveals that evoked and spontaneous synaptic transmission remain unaffected in unc-13 GFSTF 3rd instar larvae and acute presynaptic homeostatic potentiation (PHP) can be induced at control levels. Furthermore, multi-color structured-illumination shows precise co-localization of Unc-13GFSTF, Bruchpilot, and GluRIIA-receptor subunits within the synaptic mesoscale. Localization microscopy in combination with HDBSCAN algorithms detect Unc-13GFSTF subclusters that move toward the AZ center during PHP with unaltered Unc-13GFSTF protein levels.
Collapse
Affiliation(s)
- Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Achmed Mrestani
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Neurology, Leipzig University Medical Center, Leipzig, Germany
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Florian Gundelach
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Center of Mental Health, Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Würzburg, Würzburg, Germany
| | - Martin Pauli
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Fabian Komma
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Mila M Paul
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
- Department of Orthopedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
33
|
Perry S, Han Y, Qiu C, Chien C, Goel P, Nishimura S, Sajnani M, Schmid A, Sigrist SJ, Dickman D. A glutamate receptor C-tail recruits CaMKII to suppress retrograde homeostatic signaling. Nat Commun 2022; 13:7656. [PMID: 36496500 PMCID: PMC9741633 DOI: 10.1038/s41467-022-35417-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Presynaptic homeostatic plasticity (PHP) adaptively enhances neurotransmitter release following diminished postsynaptic glutamate receptor (GluR) functionality to maintain synaptic strength. While much is known about PHP expression mechanisms, postsynaptic induction remains enigmatic. For over 20 years, diminished postsynaptic Ca2+ influx was hypothesized to reduce CaMKII activity and enable retrograde PHP signaling at the Drosophila neuromuscular junction. Here, we have interrogated inductive signaling and find that active CaMKII colocalizes with and requires the GluRIIA receptor subunit. Next, we generated Ca2+-impermeable GluRs to reveal that both CaMKII activity and PHP induction are Ca2+-insensitive. Rather, a GluRIIA C-tail domain is necessary and sufficient to recruit active CaMKII. Finally, chimeric receptors demonstrate that the GluRIIA tail constitutively occludes retrograde homeostatic signaling by stabilizing active CaMKII. Thus, the physical loss of the GluRIIA tail is sensed, rather than reduced Ca2+, to enable retrograde PHP signaling, highlighting a unique, Ca2+-independent control mechanism for CaMKII in gating homeostatic plasticity.
Collapse
Affiliation(s)
- Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Yifu Han
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chengjie Qiu
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Chun Chien
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Samantha Nishimura
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Manisha Sajnani
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA
| | - Andreas Schmid
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- Faculty of Life Sciences, Albstadt-Sigmaringen University, Sigmaringen, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustraße 6, 14195, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
34
|
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
|
35
|
Turrel O, Ramesh N, Escher MJF, Pooryasin A, Sigrist SJ. Transient active zone remodeling in the Drosophila mushroom body supports memory. Curr Biol 2022; 32:4900-4913.e4. [PMID: 36327980 DOI: 10.1016/j.cub.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 08/15/2022] [Accepted: 10/06/2022] [Indexed: 11/22/2022]
Abstract
Elucidating how the distinct components of synaptic plasticity dynamically orchestrate the distinct stages of memory acquisition and maintenance within neuronal networks remains a major challenge. Specifically, plasticity processes tuning the functional and also structural state of presynaptic active zone (AZ) release sites are widely observed in vertebrates and invertebrates, but their behavioral relevance remains mostly unclear. We here provide evidence that a transient upregulation of presynaptic AZ release site proteins supports aversive olfactory mid-term memory in the Drosophila mushroom body (MB). Upon paired aversive olfactory conditioning, AZ protein levels (ELKS-family BRP/(m)unc13-family release factor Unc13A) increased for a few hours with MB-lobe-specific dynamics. Kenyon cell (KC, intrinsic MB neurons)-specific knockdown (KD) of BRP did not affect aversive olfactory short-term memory (STM) but strongly suppressed aversive mid-term memory (MTM). Different proteins crucial for the transport of AZ biosynthetic precursors (transport adaptor Aplip1/Jip-1; kinesin motor IMAC/Unc104; small GTPase Arl8) were also specifically required for the formation of aversive olfactory MTM. Consistent with the merely transitory increase of AZ proteins, BRP KD did not interfere with the formation of aversive olfactory long-term memory (LTM; i.e., 1 day). Our data suggest that the remodeling of presynaptic AZ refines the MB circuitry after paired aversive conditioning, over a time window of a few hours, to display aversive olfactory memories.
Collapse
Affiliation(s)
- Oriane Turrel
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Niraja Ramesh
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Marc J F Escher
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Atefeh Pooryasin
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany
| | - Stephan J Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Takustrasse 6, 14195 Berlin, Germany; NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
36
|
Toonen RF, Verhage M. Homing in on homeostatic plasticity. Neuron 2022; 110:3645-3647. [PMID: 36395749 DOI: 10.1016/j.neuron.2022.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this issue of Neuron, Orr et al.1 demonstrate a detailed molecular cascade that drives presynaptic homeostatic plasticity and enhances presynaptic vesicle fusion in response to reduced postsynaptic activity. Two large presynaptic signaling complexes are central hubs.
Collapse
Affiliation(s)
- Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands; Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam Neuroscience, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Muttathukunnel P, Frei P, Perry S, Dickman D, Müller M. Rapid homeostatic modulation of transsynaptic nanocolumn rings. Proc Natl Acad Sci U S A 2022; 119:e2119044119. [PMID: 36322725 PMCID: PMC9659372 DOI: 10.1073/pnas.2119044119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 09/10/2022] [Indexed: 11/07/2022] Open
Abstract
Robust neural information transfer relies on a delicate molecular nano-architecture of chemical synapses. Neurotransmitter release is controlled by a specific arrangement of proteins within presynaptic active zones. How the specific presynaptic molecular architecture relates to postsynaptic organization and how synaptic nano-architecture is transsynaptically regulated to enable stable synaptic transmission remain enigmatic. Using time-gated stimulated emission-depletion microscopy at the Drosophila neuromuscular junction, we found that presynaptic nanorings formed by the active-zone scaffold Bruchpilot (Brp) align with postsynaptic glutamate receptor (GluR) rings. Individual rings harbor approximately four transsynaptically aligned Brp-GluR nanocolumns. Similar nanocolumn rings are formed by the presynaptic protein Unc13A and GluRs. Intriguingly, acute GluR impairment triggers transsynaptic nanocolumn formation on the minute timescale during homeostatic plasticity. We reveal distinct phases of structural transsynaptic homeostatic plasticity, with postsynaptic GluR reorganization preceding presynaptic Brp modulation. Finally, homeostatic control of transsynaptic nano-architecture and neurotransmitter release requires the auxiliary GluR subunit Neto. Thus, transsynaptic nanocolumn rings provide a substrate for rapid homeostatic stabilization of synaptic efficacy.
Collapse
Affiliation(s)
- Paola Muttathukunnel
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich/Swiss Federal Institute of Technology (ETH) Zurich, Zurich, 8057 Switzerland
| | - Patrick Frei
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - Sarah Perry
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089
| | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA 90089
| | - Martin Müller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich/Swiss Federal Institute of Technology (ETH) Zurich, Zurich, 8057 Switzerland
| |
Collapse
|
38
|
Sardoo AM, Zhang S, Ferraro TN, Keck TM, Chen Y. Decoding brain memory formation by single-cell RNA sequencing. Brief Bioinform 2022; 23:6713514. [PMID: 36156112 PMCID: PMC9677489 DOI: 10.1093/bib/bbac412] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/10/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022] Open
Abstract
To understand how distinct memories are formed and stored in the brain is an important and fundamental question in neuroscience and computational biology. A population of neurons, termed engram cells, represents the physiological manifestation of a specific memory trace and is characterized by dynamic changes in gene expression, which in turn alters the synaptic connectivity and excitability of these cells. Recent applications of single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq) are promising approaches for delineating the dynamic expression profiles in these subsets of neurons, and thus understanding memory-specific genes, their combinatorial patterns and regulatory networks. The aim of this article is to review and discuss the experimental and computational procedures of sc/snRNA-seq, new studies of molecular mechanisms of memory aided by sc/snRNA-seq in human brain diseases and related mouse models, and computational challenges in understanding the regulatory mechanisms underlying long-term memory formation.
Collapse
Affiliation(s)
- Atlas M Sardoo
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Thomas M Keck
- Department of Biological & Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA,Department of Chemistry & Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Yong Chen
- Corresponding author. Yong Chen, Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA. Tel.: +1 856 256 4500; E-mail:
| |
Collapse
|
39
|
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
|
40
|
Cunningham KL, Sauvola CW, Tavana S, Littleton JT. Regulation of presynaptic Ca 2+ channel abundance at active zones through a balance of delivery and turnover. eLife 2022; 11:78648. [PMID: 35833625 PMCID: PMC9352347 DOI: 10.7554/elife.78648] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/13/2022] [Indexed: 12/03/2022] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr), a key presynaptic determinant of synaptic strength. Although biosynthesis, delivery, and recycling cooperate to establish AZ VGCC abundance, experimentally isolating these distinct regulatory processes has been difficult. Here, we describe how the AZ levels of cacophony (Cac), the sole VGCC-mediating synaptic transmission in Drosophila, are determined. We also analyzed the relationship between Cac, the conserved VGCC regulatory subunit α2δ, and the core AZ scaffold protein Bruchpilot (BRP) in establishing a functional AZ. We find that Cac and BRP are independently regulated at growing AZs, as Cac is dispensable for AZ formation and structural maturation, and BRP abundance is not limiting for Cac accumulation. Additionally, AZs stop accumulating Cac after an initial growth phase, whereas BRP levels continue to increase given extended developmental time. AZ Cac is also buffered against moderate increases or decreases in biosynthesis, whereas BRP lacks this buffering. To probe mechanisms that determine AZ Cac abundance, intravital FRAP and Cac photoconversion were used to separately measure delivery and turnover at individual AZs over a multi-day period. Cac delivery occurs broadly across the AZ population, correlates with AZ size, and is rate-limited by α2δ. Although Cac does not undergo significant lateral transfer between neighboring AZs over the course of development, Cac removal from AZs does occur and is promoted by new Cac delivery, generating a cap on Cac accumulation at mature AZs. Together, these findings reveal how Cac biosynthesis, synaptic delivery, and recycling set the abundance of VGCCs at individual AZs throughout synapse development and maintenance.
Collapse
Affiliation(s)
- Karen L Cunningham
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| | - Chad W Sauvola
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| | - Sara Tavana
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
41
|
Mochida S. Mechanisms of Synaptic Vesicle Exo- and Endocytosis. Biomedicines 2022; 10:1593. [PMID: 35884898 PMCID: PMC9313035 DOI: 10.3390/biomedicines10071593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 01/05/2023] Open
Abstract
Within 1 millisecond of action potential arrival at presynaptic terminals voltage-gated Ca2+ channels open. The Ca2+ channels are linked to synaptic vesicles which are tethered by active zone proteins. Ca2+ entrance into the active zone triggers: (1) the fusion of the vesicle and exocytosis, (2) the replenishment of the active zone with vesicles for incoming exocytosis, and (3) various types of endocytosis for vesicle reuse, dependent on the pattern of firing. These time-dependent vesicle dynamics are controlled by presynaptic Ca2+ sensor proteins, regulating active zone scaffold proteins, fusion machinery proteins, motor proteins, endocytic proteins, several enzymes, and even Ca2+ channels, following the decay of Ca2+ concentration after the action potential. Here, I summarize the Ca2+-dependent protein controls of synchronous and asynchronous vesicle release, rapid replenishment of the active zone, endocytosis, and short-term plasticity within 100 msec after the action potential. Furthermore, I discuss the contribution of active zone proteins to presynaptic plasticity and to homeostatic readjustment during and after intense activity, in addition to activity-dependent endocytosis.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
42
|
Kusick GF, Ogunmowo TH, Watanabe S. Transient docking of synaptic vesicles: Implications and mechanisms. Curr Opin Neurobiol 2022; 74:102535. [PMID: 35398664 PMCID: PMC9167714 DOI: 10.1016/j.conb.2022.102535] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/19/2022] [Accepted: 03/06/2022] [Indexed: 02/03/2023]
Abstract
As synaptic vesicles fuse, they must continually be replaced with new docked, fusion-competent vesicles to sustain neurotransmission. It has long been appreciated that vesicles are recruited to docking sites in an activity-dependent manner. However, once entering the sites, vesicles were thought to be stably docked, awaiting calcium signals. Based on recent data from electrophysiology, electron microscopy, biochemistry, and computer simulations, a picture emerges in which vesicles can rapidly and reversibly transit between docking and undocking during activity. This "transient docking" can account for many aspects of synaptic physiology. In this review, we cover recent evidence for transient docking, physiological processes at the synapse that it may support, and progress on the underlying mechanisms. We also discuss an open question: what determines for how long and whether vesicles stay docked, or eventually undock?
Collapse
Affiliation(s)
- Grant F Kusick
- Department of Cell Biology, Johns Hopkins University, School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA; Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, School of Medicine, 1830 E. Monument St., Baltimore, MD 21287, USA. https://twitter.com/@ultrafastgrant
| | - Tyler H Ogunmowo
- Department of Cell Biology, Johns Hopkins University, School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA; Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University, School of Medicine, 1830 E. Monument St., Baltimore, MD 21287, USA. https://twitter.com/@unculturedTy
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, School of Medicine, 725 N Wolfe St., Baltimore, MD 21287, USA.
| |
Collapse
|
43
|
Midorikawa M. Pathway-specific maturation of presynaptic functions of the somatosensory thalamus. Neurosci Res 2022; 181:1-8. [DOI: 10.1016/j.neures.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023]
|
44
|
Piao C, Sigrist SJ. (M)Unc13s in Active Zone Diversity: A Drosophila Perspective. Front Synaptic Neurosci 2022; 13:798204. [PMID: 35046788 PMCID: PMC8762327 DOI: 10.3389/fnsyn.2021.798204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/29/2021] [Indexed: 12/03/2022] Open
Abstract
The so-called active zones at pre-synaptic terminals are the ultimate filtering devices, which couple between action potential frequency and shape, and the information transferred to the post-synaptic neurons, finally tuning behaviors. Within active zones, the release of the synaptic vesicle operates from specialized “release sites.” The (M)Unc13 class of proteins is meant to define release sites topologically and biochemically, and diversity between Unc13-type release factor isoforms is suspected to steer diversity at active zones. The two major Unc13-type isoforms, namely, Unc13A and Unc13B, have recently been described from the molecular to the behavioral level, exploiting Drosophila being uniquely suited to causally link between these levels. The exact nanoscale distribution of voltage-gated Ca2+ channels relative to release sites (“coupling”) at pre-synaptic active zones fundamentally steers the release of the synaptic vesicle. Unc13A and B were found to be either tightly or loosely coupled across Drosophila synapses. In this review, we reported recent findings on diverse aspects of Drosophila Unc13A and B, importantly, their nano-topological distribution at active zones and their roles in release site generation, active zone assembly, and pre-synaptic homeostatic plasticity. We compared their stoichiometric composition at different synapse types, reviewing the correlation between nanoscale distribution of these two isoforms and release physiology and, finally, discuss how isoform-specific release components might drive the functional heterogeneity of synapses and encode discrete behavior.
Collapse
Affiliation(s)
- Chengji Piao
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
- *Correspondence: Stephan J. Sigrist
| |
Collapse
|
45
|
Pribbenow C, Chen YC, Heim MM, Laber D, Reubold S, Reynolds E, Balles I, Fernández-d V Alquicira T, Suárez-Grimalt R, Scheunemann L, Rauch C, Matkovic T, Rösner J, Lichtner G, Jagannathan SR, Owald D. Postsynaptic plasticity of cholinergic synapses underlies the induction and expression of appetitive and familiarity memories in Drosophila. eLife 2022; 11:80445. [PMID: 36250621 PMCID: PMC9733945 DOI: 10.7554/elife.80445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022] Open
Abstract
In vertebrates, several forms of memory-relevant synaptic plasticity involve postsynaptic rearrangements of glutamate receptors. In contrast, previous work indicates that Drosophila and other invertebrates store memories using presynaptic plasticity of cholinergic synapses. Here, we provide evidence for postsynaptic plasticity at cholinergic output synapses from the Drosophila mushroom bodies (MBs). We find that the nicotinic acetylcholine receptor (nAChR) subunit α5 is required within specific MB output neurons for appetitive memory induction but is dispensable for aversive memories. In addition, nAChR α2 subunits mediate memory expression and likely function downstream of α5 and the postsynaptic scaffold protein discs large (Dlg). We show that postsynaptic plasticity traces can be induced independently of the presynapse, and that in vivo dynamics of α2 nAChR subunits are changed both in the context of associative and non-associative (familiarity) memory formation, underlying different plasticity rules. Therefore, regardless of neurotransmitter identity, key principles of postsynaptic plasticity support memory storage across phyla.
Collapse
Affiliation(s)
- Carlotta Pribbenow
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Yi-chun Chen
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - M-Marcel Heim
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Desiree Laber
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Silas Reubold
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Eric Reynolds
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Isabella Balles
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Tania Fernández-d V Alquicira
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Raquel Suárez-Grimalt
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany,Einstein Center for Neurosciences BerlinBerlinGermany
| | - Lisa Scheunemann
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany,NeuroCure, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany,Institut für Biologie, Freie Universität BerlinBerlinGermany
| | - Carolin Rauch
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - Tanja Matkovic
- Institut für Biologie, Freie Universität BerlinBerlinGermany
| | - Jörg Rösner
- NWFZ, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthGreifswaldGermany
| | - Gregor Lichtner
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany,Universitätsmedizin Greifswald, Department of Anesthesia, Critical Care, Emergency and Pain MedicineGreifswaldGermany
| | - Sridhar R Jagannathan
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| | - David Owald
- Institute of Neurophysiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany,Einstein Center for Neurosciences BerlinBerlinGermany,NeuroCure, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of HealthBerlinGermany
| |
Collapse
|
46
|
Nair AG, Muttathukunnel P, Müller M. Distinct molecular pathways govern presynaptic homeostatic plasticity. Cell Rep 2021; 37:110105. [PMID: 34910905 PMCID: PMC8692748 DOI: 10.1016/j.celrep.2021.110105] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 10/05/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
Presynaptic homeostatic plasticity (PHP) stabilizes synaptic transmission by counteracting impaired neurotransmitter receptor function through neurotransmitter release potentiation. PHP is thought to be triggered by impaired receptor function and to involve a stereotypic signaling pathway. However, here we demonstrate that different receptor perturbations that similarly reduce synaptic transmission result in different responses at the Drosophila neuromuscular junction. While receptor inhibition by the glutamate receptor (GluR) antagonist γ-D-glutamylglycine (γDGG) is not compensated by PHP, the GluR inhibitors Philanthotoxin-433 (PhTx) and Gyki-53655 (Gyki) induce compensatory PHP. Intriguingly, PHP triggered by PhTx and Gyki involve separable signaling pathways, including inhibition of distinct GluR subtypes, differential modulation of the active-zone scaffold Bruchpilot, and short-term plasticity. Moreover, while PHP upon Gyki treatment does not require genes promoting PhTx-induced PHP, it involves presynaptic protein kinase D. Thus, synapses not only respond differentially to similar activity impairments, but achieve homeostatic compensation via distinct mechanisms, highlighting the diversity of homeostatic signaling.
Collapse
Affiliation(s)
- Anu G Nair
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Paola Muttathukunnel
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich/ETH Zurich, 8057 Zurich, Switzerland
| | - Martin Müller
- Department of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich/ETH Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
47
|
Mochida S. Stable and Flexible Synaptic Transmission Controlled by the Active Zone Protein Interactions. Int J Mol Sci 2021; 22:ijms222111775. [PMID: 34769208 PMCID: PMC8583982 DOI: 10.3390/ijms222111775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022] Open
Abstract
An action potential triggers neurotransmitter release from synaptic vesicles docking to a specialized release site of the presynaptic plasma membrane, the active zone. The active zone is a highly organized structure with proteins that serves as a platform for synaptic vesicle exocytosis, mediated by SNAREs complex and Ca2+ sensor proteins, within a sub-millisecond opening of nearby Ca2+ channels with the membrane depolarization. In response to incoming neuronal signals, each active zone protein plays a role in the release-ready site replenishment with synaptic vesicles for sustainable synaptic transmission. The active zone release apparatus provides a possible link between neuronal activity and plasticity. This review summarizes the mostly physiological role of active zone protein interactions that control synaptic strength, presynaptic short-term plasticity, and homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Sumiko Mochida
- Department of Physiology, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
48
|
Sauvola CW, Akbergenova Y, Cunningham KL, Aponte-Santiago NA, Littleton JT. The decoy SNARE Tomosyn sets tonic versus phasic release properties and is required for homeostatic synaptic plasticity. eLife 2021; 10:e72841. [PMID: 34713802 PMCID: PMC8612732 DOI: 10.7554/elife.72841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022] Open
Abstract
Synaptic vesicle (SV) release probability (Pr) is a key presynaptic determinant of synaptic strength established by cell-intrinsic properties and further refined by plasticity. To characterize mechanisms that generate Pr heterogeneity between distinct neuronal populations, we examined glutamatergic tonic (Ib) and phasic (Is) motoneurons in Drosophila with stereotyped differences in Pr and synaptic plasticity. We found the decoy soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) Tomosyn is differentially expressed between these motoneuron subclasses and contributes to intrinsic differences in their synaptic output. Tomosyn expression enables tonic release in Ib motoneurons by reducing SNARE complex formation and suppressing Pr to generate decreased levels of SV fusion and enhanced resistance to synaptic fatigue. In contrast, phasic release dominates when Tomosyn expression is low, enabling high intrinsic Pr at Is terminals at the expense of sustained release and robust presynaptic potentiation. In addition, loss of Tomosyn disrupts the ability of tonic synapses to undergo presynaptic homeostatic potentiation.
Collapse
Affiliation(s)
- Chad W Sauvola
- Department of Brain and Cognitive Sciences, The Picower Institute of Learning and Memory, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Yulia Akbergenova
- Department of Brain and Cognitive Sciences, The Picower Institute of Learning and Memory, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Karen L Cunningham
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| | | | - J Troy Littleton
- Department of Brain and Cognitive Sciences, The Picower Institute of Learning and Memory, Massachusetts Institute of TechnologyCambridgeUnited States
- Department of Biology, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
49
|
Mrestani A, Pauli M, Kollmannsberger P, Repp F, Kittel RJ, Eilers J, Doose S, Sauer M, Sirén AL, Heckmann M, Paul MM. Active zone compaction correlates with presynaptic homeostatic potentiation. Cell Rep 2021; 37:109770. [PMID: 34610300 DOI: 10.1016/j.celrep.2021.109770] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/14/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022] Open
Abstract
Neurotransmitter release is stabilized by homeostatic plasticity. Presynaptic homeostatic potentiation (PHP) operates on timescales ranging from minute- to life-long adaptations and likely involves reorganization of presynaptic active zones (AZs). At Drosophila melanogaster neuromuscular junctions, earlier work ascribed AZ enlargement by incorporating more Bruchpilot (Brp) scaffold protein a role in PHP. We use localization microscopy (direct stochastic optical reconstruction microscopy [dSTORM]) and hierarchical density-based spatial clustering of applications with noise (HDBSCAN) to study AZ plasticity during PHP at the synaptic mesoscale. We find compaction of individual AZs in acute philanthotoxin-induced and chronic genetically induced PHP but unchanged copy numbers of AZ proteins. Compaction even occurs at the level of Brp subclusters, which move toward AZ centers, and in Rab3 interacting molecule (RIM)-binding protein (RBP) subclusters. Furthermore, correlative confocal and dSTORM imaging reveals how AZ compaction in PHP translates into apparent increases in AZ area and Brp protein content, as implied earlier.
Collapse
Affiliation(s)
- Achmed Mrestani
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurology, Leipzig University Medical Center, 04103 Leipzig, Germany
| | - Martin Pauli
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Philip Kollmannsberger
- Center for Computational and Theoretical Biology, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Felix Repp
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Center for Computational and Theoretical Biology, Julius Maximilians University Würzburg, 97074 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Robert J Kittel
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Institute of Biology, Department of Animal Physiology, Leipzig University, 04103 Leipzig, Germany; Carl-Ludwig-Institute for Physiology, Leipzig University, 04103 Leipzig, Germany
| | - Jens Eilers
- Carl-Ludwig-Institute for Physiology, Leipzig University, 04103 Leipzig, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Julius Maximilians University Würzburg, 97074 Würzburg, Germany
| | - Anna-Leena Sirén
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Neurosurgery, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Manfred Heckmann
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany.
| | - Mila M Paul
- Institute for Physiology, Department of Neurophysiology, Julius Maximilians University Würzburg, 97070 Würzburg, Germany; Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
50
|
Kudryashova IV. The Reorganization of the Actin Matrix as a Factor of Presynaptic Plasticity. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421030089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|