101
|
Maxeiner S, Luo F, Tan A, Schmitz F, Südhof TC. How to make a synaptic ribbon: RIBEYE deletion abolishes ribbons in retinal synapses and disrupts neurotransmitter release. EMBO J 2016; 35:1098-114. [PMID: 26929012 DOI: 10.15252/embj.201592701] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 02/01/2016] [Indexed: 12/21/2022] Open
Abstract
Synaptic ribbons are large proteinaceous scaffolds at the active zone of ribbon synapses that are specialized for rapid sustained synaptic vesicles exocytosis. A single ribbon-specific protein is known, RIBEYE, suggesting that ribbons may be constructed from RIBEYE protein. RIBEYE knockdown in zebrafish, however, only reduced but did not eliminate ribbons, indicating a more ancillary role. Here, we show in mice that full deletion of RIBEYE abolishes all presynaptic ribbons in retina synapses. Using paired recordings in acute retina slices, we demonstrate that deletion of RIBEYE severely impaired fast and sustained neurotransmitter release at bipolar neuron/AII amacrine cell synapses and rendered spontaneous miniature release sensitive to the slow Ca(2+)-buffer EGTA, suggesting that synaptic ribbons mediate nano-domain coupling of Ca(2+) channels to synaptic vesicle exocytosis. Our results show that RIBEYE is essential for synaptic ribbons as such, and may organize presynaptic nano-domains that position release-ready synaptic vesicles adjacent to Ca(2+) channels.
Collapse
Affiliation(s)
- Stephan Maxeiner
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute Stanford University School of Medicine, Stanford, CA, USA Department of Neuroanatomy, Institute for Anatomy and Cell Biology Medical School Saarland University, Homburg/Saar, Germany
| | - Fujun Luo
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute Stanford University School of Medicine, Stanford, CA, USA
| | - Alison Tan
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute Stanford University School of Medicine, Stanford, CA, USA
| | - Frank Schmitz
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology Medical School Saarland University, Homburg/Saar, Germany
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
102
|
Binotti B, Jahn R, Chua JJE. Functions of Rab Proteins at Presynaptic Sites. Cells 2016; 5:E7. [PMID: 26861397 PMCID: PMC4810092 DOI: 10.3390/cells5010007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/03/2016] [Indexed: 01/24/2023] Open
Abstract
Presynaptic neurotransmitter release is dominated by the synaptic vesicle (SV) cycle and entails the biogenesis, fusion, recycling, reformation or turnover of synaptic vesicles-a process involving bulk movement of membrane and proteins. As key mediators of membrane trafficking, small GTPases from the Rab family of proteins play critical roles in this process by acting as molecular switches that dynamically interact with and regulate the functions of different sets of macromolecular complexes involved in each stage of the cycle. Importantly, mutations affecting Rabs, and their regulators or effectors have now been identified that are implicated in severe neurological and neurodevelopmental disorders. Here, we summarize the roles and functions of presynaptic Rabs and discuss their involvement in the regulation of presynaptic function.
Collapse
Affiliation(s)
- Beyenech Binotti
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - Reinhard Jahn
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen 37077, Germany
| | - John Jia En Chua
- Interactomics and Intracellular Trafficking laboratory, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore.
- Neurobiology/Ageing Programme, National University of Singapore, Singapore 117456, Singapore.
- Research Group Protein trafficking in synaptic development and function, Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Göttingen 37077, Germany.
| |
Collapse
|
103
|
Acuna C, Liu X, Gonzalez A, Südhof TC. RIM-BPs Mediate Tight Coupling of Action Potentials to Ca(2+)-Triggered Neurotransmitter Release. Neuron 2015; 87:1234-1247. [PMID: 26402606 DOI: 10.1016/j.neuron.2015.08.027] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 07/25/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023]
Abstract
Ultrafast neurotransmitter release requires tight colocalization of voltage-gated Ca(2+) channels with primed, release-ready synaptic vesicles at the presynaptic active zone. RIM-binding proteins (RIM-BPs) are multidomain active zone proteins that bind to RIMs and to Ca(2+) channels. In Drosophila, deletion of RIM-BPs dramatically reduces neurotransmitter release, but little is known about RIM-BP function in mammalian synapses. Here, we generated double conditional knockout mice for RIM-BP1 and RIM-BP2, and analyzed RIM-BP-deficient synapses in cultured hippocampal neurons and the calyx of Held. Surprisingly, we find that in murine synapses, RIM-BPs are not essential for neurotransmitter release as such, but are selectively required for high-fidelity coupling of action potential-induced Ca(2+) influx to Ca(2+)-stimulated synaptic vesicle exocytosis. Deletion of RIM-BPs decelerated action-potential-triggered neurotransmitter release and rendered it unreliable, thereby impairing the fidelity of synaptic transmission. Thus, RIM-BPs ensure optimal organization of the machinery for fast release in mammalian synapses without being a central component of the machinery itself.
Collapse
Affiliation(s)
- Claudio Acuna
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Xinran Liu
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Aneysis Gonzalez
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Cellular and Molecular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
104
|
RIM1/2-Mediated Facilitation of Cav1.4 Channel Opening Is Required for Ca2+-Stimulated Release in Mouse Rod Photoreceptors. J Neurosci 2015; 35:13133-47. [PMID: 26400943 DOI: 10.1523/jneurosci.0658-15.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Night blindness can result from impaired photoreceptor function and a subset of cases have been linked to dysfunction of Cav1.4 calcium channels and in turn compromised synaptic transmission. Here, we show that active zone proteins RIM1/2 are important regulators of Cav1.4 channel function in mouse rod photoreceptors and thus synaptic activity. The conditional double knock-out (cdko) of RIM1 and RIM2 from rods starting a few weeks after birth did not change Cav1.4 protein expression at rod ribbon synapses nor was the morphology of the ribbon altered. Heterologous overexpression of RIM2 with Cav1.4 had no significant influence on current density when examined with BaCl2 as the charge carrier. Nonetheless, whole-cell voltage-clamp recordings from cdko rods revealed a profound reduction in Ca(2+) currents. Concomitantly, we observed a 4-fold reduction in spontaneous miniature release events from the cdko rod terminals and an almost complete absence of evoked responses when monitoring changes in membrane incorporation after strong step depolarizations. Under control conditions, 49 and 83 vesicles were released with 0.2 and 1 s depolarizations, respectively, which is close to the maximal number of vesicles estimated to be docked at the base of the ribbon active zone, but without RIM1/2, only a few vesicles were stimulated for release after a 1 s stimulation. In conclusion, our study shows that RIM1/2 potently enhance the influx of Ca(2+) into rod terminals through Cav1.4 channels, which is vitally important for the release of vesicles from the rod ribbon. Significance statement: Active zone scaffolding proteins are thought to bring multiple components involved in Ca(2+)-dependent exocytosis into functional interactions. We show that removal of scaffolding proteins RIM1/2 from rod photoreceptor ribbon synapses causes a dramatic loss of Ca(2+) influx through Cav1.4 channels and a correlated reduction in evoked release, yet the channels remain localized to synaptic ribbons in a normal fashion. Our findings strongly argue that RIM1/2 facilitate Ca(2+) entry and in turn Ca(2+) evoked release by modulating Cav1.4 channel openings; however, RIM1/2 are not needed for the retention of Cav1.4 at the synapse. In summary, a key function of RIM1/2 at rod ribbons is to enhance Cav1.4 channel activity, possibly through direct or indirect modulation of the channel.
Collapse
|
105
|
Crawford DC, Kavalali ET. Molecular underpinnings of synaptic vesicle pool heterogeneity. Traffic 2015; 16:338-64. [PMID: 25620674 DOI: 10.1111/tra.12262] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/06/2015] [Indexed: 12/31/2022]
Abstract
Neuronal communication relies on chemical synaptic transmission for information transfer and processing. Chemical neurotransmission is initiated by synaptic vesicle fusion with the presynaptic active zone resulting in release of neurotransmitters. Classical models have assumed that all synaptic vesicles within a synapse have the same potential to fuse under different functional contexts. In this model, functional differences among synaptic vesicle populations are ascribed to their spatial distribution in the synapse with respect to the active zone. Emerging evidence suggests, however, that synaptic vesicles are not a homogenous population of organelles, and they possess intrinsic molecular differences and differential interaction partners. Recent studies have reported a diverse array of synaptic molecules that selectively regulate synaptic vesicles' ability to fuse synchronously and asynchronously in response to action potentials or spontaneously irrespective of action potentials. Here we discuss these molecular mediators of vesicle pool heterogeneity that are found on the synaptic vesicle membrane, on the presynaptic plasma membrane, or within the cytosol and consider some of the functional consequences of this diversity. This emerging molecular framework presents novel avenues to probe synaptic function and uncover how synaptic vesicle pools impact neuronal signaling.
Collapse
Affiliation(s)
- Devon C Crawford
- Department of Neuroscience, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9111, USA
| | | |
Collapse
|
106
|
Abstract
The first synapses transmitting visual information contain an unusual organelle, the ribbon, which is involved in the transport and priming of vesicles to be released at the active zone. The ribbon is one of many design features that allow efficient refilling of the active zone, which in turn enables graded changes in membrane potential to be transmitted using a continuous mode of neurotransmitter release. The ribbon also plays a key role in supplying vesicles for rapid and transient bursts of release that signal fast changes, such as the onset of light. We increasingly understand how the physiological properties of ribbon synapses determine basic transformations of the visual signal and, in particular, how the process of refilling the active zone regulates the gain and adaptive properties of the retinal circuit. The molecular basis of ribbon function is, however, far from clear.
Collapse
Affiliation(s)
- Leon Lagnado
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom;
| | - Frank Schmitz
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Medical School Saarland University, Homburg/Saar, Germany;
| |
Collapse
|
107
|
Mutational Analysis of Rab3 Function for Controlling Active Zone Protein Composition at the Drosophila Neuromuscular Junction. PLoS One 2015; 10:e0136938. [PMID: 26317909 PMCID: PMC4552854 DOI: 10.1371/journal.pone.0136938] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/11/2015] [Indexed: 11/26/2022] Open
Abstract
At synapses, the release of neurotransmitter is regulated by molecular machinery that aggregates at specialized presynaptic release sites termed active zones. The complement of active zone proteins at each site is a determinant of release efficacy and can be remodeled to alter synapse function. The small GTPase Rab3 was previously identified as playing a novel role that controls the distribution of active zone proteins to individual release sites at the Drosophila neuromuscular junction. Rab3 has been extensively studied for its role in the synaptic vesicle cycle; however, the mechanism by which Rab3 controls active zone development remains unknown. To explore this mechanism, we conducted a mutational analysis to determine the molecular and structural requirements of Rab3 function at Drosophila synapses. We find that GTP-binding is required for Rab3 to traffick to synapses and distribute active zone components across release sites. Conversely, the hydrolytic activity of Rab3 is unnecessary for this function. Through a structure-function analysis we identify specific residues within the effector-binding switch regions that are required for Rab3 function and determine that membrane attachment is essential. Our findings suggest that Rab3 controls the distribution of active zone components via a vesicle docking mechanism that is consistent with standard Rab protein function.
Collapse
|
108
|
Wichmann C, Moser T. Relating structure and function of inner hair cell ribbon synapses. Cell Tissue Res 2015; 361:95-114. [PMID: 25874597 PMCID: PMC4487357 DOI: 10.1007/s00441-014-2102-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/18/2014] [Indexed: 01/28/2023]
Abstract
In the mammalian cochlea, sound is encoded at synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs). Each SGN receives input from a single IHC ribbon-type active zone (AZ) and yet SGNs indefatigably spike up to hundreds of Hz to encode acoustic stimuli with submillisecond precision. Accumulating evidence indicates a highly specialized molecular composition and structure of the presynapse, adapted to suit these high functional demands. However, we are only beginning to understand key features such as stimulus-secretion coupling, exocytosis mechanisms, exo-endocytosis coupling, modes of endocytosis and vesicle reformation, as well as replenishment of the readily releasable pool. Relating structure and function has become an important avenue in addressing these points and has been applied to normal and genetically manipulated hair cell synapses. Here, we review some of the exciting new insights gained from recent studies of the molecular anatomy and physiology of IHC ribbon synapses.
Collapse
Affiliation(s)
- C. Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University Medical Center Göttingen, Göttingen, Germany
| | - T. Moser
- Collaborative Research Center 889, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University of Göttingen, Göttingen, Germany
- Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany
| |
Collapse
|
109
|
|
110
|
Di Giovanni J, Sheng ZH. Regulation of synaptic activity by snapin-mediated endolysosomal transport and sorting. EMBO J 2015; 34:2059-77. [PMID: 26108535 DOI: 10.15252/embj.201591125] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/29/2015] [Indexed: 11/09/2022] Open
Abstract
Recycling synaptic vesicles (SVs) transit through early endosomal sorting stations, which raises a fundamental question: are SVs sorted toward endolysosomal pathways? Here, we used snapin mutants as tools to assess how endolysosomal sorting and trafficking impact presynaptic activity in wild-type and snapin(-/-) neurons. Snapin acts as a dynein adaptor that mediates the retrograde transport of late endosomes (LEs) and interacts with dysbindin, a subunit of the endosomal sorting complex BLOC-1. Expressing dynein-binding defective snapin mutants induced SV accumulation at presynaptic terminals, mimicking the snapin(-/-) phenotype. Conversely, over-expressing snapin reduced SV pool size by enhancing SV trafficking to the endolysosomal pathway. Using a SV-targeted Ca(2+) sensor, we demonstrate that snapin-dysbindin interaction regulates SV positional priming through BLOC-1/AP-3-dependent sorting. Our study reveals a bipartite regulation of presynaptic activity by endolysosomal trafficking and sorting: LE transport regulates SV pool size, and BLOC-1/AP-3-dependent sorting fine-tunes the Ca(2+) sensitivity of SV release. Therefore, our study provides new mechanistic insights into the maintenance and regulation of SV pool size and synchronized SV fusion through snapin-mediated LE trafficking and endosomal sorting.
Collapse
Affiliation(s)
- Jerome Di Giovanni
- Synaptic Functions Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Zu-Hang Sheng
- Synaptic Functions Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
111
|
Singh CO, Xin HH, Chen RT, Wang MX, Liang S, Lu Y, Cai ZZ, Zhang DP, Miao YG. RNAi KNOCKDOWN OF BmRab3 LED TO LARVA AND PUPA LETHALITY IN SILKWORM Bombyx mori L. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2015; 89:98-110. [PMID: 25735242 DOI: 10.1002/arch.21228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Rab3 GTPases are known to play key a role in vesicular trafficking, and express highest in brain and endocrine tissues. In mammals, Rab3 GTPases are paralogs unlike in insect. In this study, we cloned Rab3 from the silk gland tissue of silkworm Bombyx mori, and identified it as BmRab3. Our in silico analysis indicated that BmRab3 is an isoform with a theoretical isoelectric point and molecular weight of 5.52 and 24.3 kDa, respectively. Further, BmRab3 showed the C-terminal hypervariability for GGT2 site but having two other putative guanine nucleotide exchange factor/GDP dissociation inhibitor interaction sites. Multiple alignment sequence indicated high similarities of BmRab3 with Rab3 isoforms of other species. The phylogeny tree showed BmRab3 clustered between the species of Tribolium castaneum and Aedes aegypti. Meanwhile, the expression analysis of BmRab3 showed the highest expression in middle silk glands (MSGs) than all other tissues in the third day of fifth-instar larva. Simultaneously, we showed the differential expression of BmRab3 in the early instar larva development, followed by higher expression in male than female pupae. In vivo dsRNA interference of BmRab3 reduced the expression of BmRab3 by 75% compared to the control in the MSGs in the first day. But as the worm grew to the third day, the difference of BmRab3 between knockdown and control was only about 10%. The knockdown later witnessed underdevelopment of the larvae and pharate pupae lethality in the overall development of silkworm B. mori L.
Collapse
Affiliation(s)
- Chabungbam Orville Singh
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Hu-hu Xin
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Rui-ting Chen
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Mei-xian Wang
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Shuang Liang
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Yan Lu
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Zi-zheng Cai
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Deng-pan Zhang
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Yun-gen Miao
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou, P. R. China
| |
Collapse
|
112
|
Rab3-interacting molecules 2α and 2β promote the abundance of voltage-gated CaV1.3 Ca2+ channels at hair cell active zones. Proc Natl Acad Sci U S A 2015; 112:E3141-9. [PMID: 26034270 DOI: 10.1073/pnas.1417207112] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ca(2+) influx triggers the fusion of synaptic vesicles at the presynaptic active zone (AZ). Here we demonstrate a role of Ras-related in brain 3 (Rab3)-interacting molecules 2α and β (RIM2α and RIM2β) in clustering voltage-gated CaV1.3 Ca(2+) channels at the AZs of sensory inner hair cells (IHCs). We show that IHCs of hearing mice express mainly RIM2α, but also RIM2β and RIM3γ, which all localize to the AZs, as shown by immunofluorescence microscopy. Immunohistochemistry, patch-clamp, fluctuation analysis, and confocal Ca(2+) imaging demonstrate that AZs of RIM2α-deficient IHCs cluster fewer synaptic CaV1.3 Ca(2+) channels, resulting in reduced synaptic Ca(2+) influx. Using superresolution microscopy, we found that Ca(2+) channels remained clustered in stripes underneath anchored ribbons. Electron tomography of high-pressure frozen synapses revealed a reduced fraction of membrane-tethered vesicles, whereas the total number of membrane-proximal vesicles was unaltered. Membrane capacitance measurements revealed a reduction of exocytosis largely in proportion with the Ca(2+) current, whereas the apparent Ca(2+) dependence of exocytosis was unchanged. Hair cell-specific deletion of all RIM2 isoforms caused a stronger reduction of Ca(2+) influx and exocytosis and significantly impaired the encoding of sound onset in the postsynaptic spiral ganglion neurons. Auditory brainstem responses indicated a mild hearing impairment on hair cell-specific deletion of all RIM2 isoforms or global inactivation of RIM2α. We conclude that RIM2α and RIM2β promote a large complement of synaptic Ca(2+) channels at IHC AZs and are required for normal hearing.
Collapse
|
113
|
Cheng Y, Wang J, Wang Y, Ding M. Synaptotagmin 1 directs repetitive release by coupling vesicle exocytosis to the Rab3 cycle. eLife 2015; 4. [PMID: 25710274 PMCID: PMC4374511 DOI: 10.7554/elife.05118] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/23/2015] [Indexed: 01/09/2023] Open
Abstract
In response to Ca2+ influx, a synapse needs to release neurotransmitters quickly while immediately preparing for repeat firing. How this harmonization is achieved is not known. In this study, we found that the Ca2+ sensor synaptotagmin 1 orchestrates the membrane association/disassociation cycle of Rab3, which functions in activity-dependent recruitment of synaptic vesicles. In the absence of Ca2+, synaptotagmin 1 binds to Rab3 GTPase activating protein (GAP) and inhibits the GTP hydrolysis of Rab3 protein. Rab3 GAP resides on synaptic vesicles, and synaptotagmin 1 is essential for the synaptic localization of Rab3 GAP. In the presence of Ca2+, synaptotagmin 1 releases Rab3 GAP and promotes membrane disassociation of Rab3. Without synaptotagmin 1, the tight coupling between vesicle exocytosis and Rab3 membrane disassociation is disrupted. We uncovered the long-sought molecular apparatus linking vesicle exocytosis to Rab3 cycling and we also revealed the important function of synaptotagmin 1 in repetitive synaptic vesicle release. DOI:http://dx.doi.org/10.7554/eLife.05118.001 Neurons communicate with one another at junctions called synapses. The arrival of an electrical signal called an action potential causes calcium ions to enter the first cell, which in turn triggers the release of molecules called neurotransmitters into the gap between the neurons. The binding of these molecules to receptors on the second cell then enables the action potential to be regenerated. For cells to respond rapidly and reliably to incoming electrical signals, they must maintain a supply of vesicles—the packages that contain neurotransmitters—close to the site where they are released from the first cell. The vesicles are held in contact with the cell membrane by a structure called the docking complex. A number of the proteins in this docking complex have been identified, including two that have been referred to as the ‘yin and yang’ of vesicle fusion: synaptotagmin, which promotes fusion, and Rab3, which limits it. However, little is known about how these and other proteins interact to keep vesicles docked at the membrane. Cheng, Wang et al. have now clarified the docking process with the aid of experiments in nematode worms. In resting neurons that are not releasing neurotransmitters, synaptotagmin (‘yin’) binds to an enzyme called GAP and prevents it from converting GTP—an energy-storage molecule—into GDP. Given that Rab3 (‘yang’) requires a molecule of GTP to power its own activity, the actions of synaptotagmin ensure that Rab3 has enough energy to remain bound to other proteins within the docking complex. However, when an action potential arrives, calcium ions enter the neuron, and some of them bind to synaptotagmin. This disrupts its interaction with the GAP enzyme, which thus becomes free to convert the GTP molecule bound to Rab3 into GDP. The loss of its energy source causes Rab3 to separate from its binding partners, and docking complex collapses. As a result, vesicles fuse with the membrane and release neurotransmitter molecules into the synapse. Given that Rab3 and synaptotagmin have changed little over the course of evolution, it is highly likely that the same indirect interaction between these two proteins also regulates the release of transmitter at synapses in the mammalian brain. DOI:http://dx.doi.org/10.7554/eLife.05118.002
Collapse
Affiliation(s)
- Yunsheng Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jiaming Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
114
|
Protein mutated in paroxysmal dyskinesia interacts with the active zone protein RIM and suppresses synaptic vesicle exocytosis. Proc Natl Acad Sci U S A 2015; 112:2935-41. [PMID: 25730884 DOI: 10.1073/pnas.1501364112] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Paroxysmal nonkinesigenic dyskinesia (PNKD) is an autosomal dominant episodic movement disorder precipitated by coffee, alcohol, and stress. We previously identified the causative gene but the function of the encoded protein remains unknown. We also generated a PNKD mouse model that revealed dysregulated dopamine signaling in vivo. Here, we show that PNKD interacts with synaptic active zone proteins Rab3-interacting molecule (RIM)1 and RIM2, localizes to synapses, and modulates neurotransmitter release. Overexpressed PNKD protein suppresses release, and mutant PNKD protein is less effective than wild-type at inhibiting exocytosis. In PNKD KO mice, RIM1/2 protein levels are reduced and synaptic strength is impaired. Thus, PNKD is a novel synaptic protein with a regulatory role in neurotransmitter release.
Collapse
|
115
|
Lardong JA, Driller JH, Depner H, Weise C, Petzoldt A, Wahl MC, Sigrist SJ, Loll B. Structures of Drosophila melanogaster Rab2 and Rab3 bound to GMPPNP. Acta Crystallogr F Struct Biol Commun 2015; 71:34-40. [PMID: 25615965 PMCID: PMC4304744 DOI: 10.1107/s2053230x1402617x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/28/2014] [Indexed: 01/11/2023] Open
Abstract
Rab GTPases belong to the large family of Ras proteins. They act as key regulators of membrane organization and intracellular trafficking. Functionally, they act as switches. In the active GTP-bound form they can bind to effector proteins to facilitate the delivery of transport vesicles. Upon stimulation, the GTP is hydrolyzed and the Rab proteins undergo conformational changes in their switch regions. This study focuses on Rab2 and Rab3 from Drosophila melanogaster. Whereas Rab2 is involved in vesicle transport between the Golgi and the endoplasmatic reticulum, Rab3 is a key player in exocytosis, and in the synapse it is involved in the assembly of the presynaptic active zone. Here, high-resolution crystal structures of Rab2 and Rab3 in complex with GMPPNP and Mg2+ are presented. In the structure of Rab3 a modified cysteine residue is observed with an enigmatic electron density attached to its thiol function.
Collapse
Affiliation(s)
- Jennifer A. Lardong
- Institut für Chemie und Biochemie Abteilung Strukturbiochemie, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
| | - Jan H. Driller
- Institut für Chemie und Biochemie Abteilung Strukturbiochemie, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
| | - Harald Depner
- Biologie Abteilung Genetik, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Christoph Weise
- Institut für Chemie und Biochemie, BioSupraMol Core Facility, Freie Universität Berlin, Thielallee 63, 15195 Berlin, Germany
| | - Astrid Petzoldt
- Biologie Abteilung Genetik, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Markus C. Wahl
- Institut für Chemie und Biochemie Abteilung Strukturbiochemie, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
| | - Stephan J. Sigrist
- Biologie Abteilung Genetik, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
- NeuroCure Cluster of Excellence, Charité Universitätsmedizin Berlin, Virchowweg 6, 10117 Berlin, Germany
| | - Bernhard Loll
- Institut für Chemie und Biochemie Abteilung Strukturbiochemie, Freie Universität Berlin, Takustrasse 6, 15195 Berlin, Germany
| |
Collapse
|
116
|
Abstract
Synaptic vesicles release their vesicular contents to the extracellular space by Ca(2+)-triggered exocytosis. The Ca(2+)-triggered exocytotic process is regulated by synaptotagmin (Syt), a vesicular Ca(2+)-binding C2 domain protein. Synaptotagmin 1 (Syt1), the most studied major isoform among 16 Syt isoforms, mediates Ca(2+)-triggered synaptic vesicle exocytosis by interacting with the target membranes and SNARE/complexin complex. In synapses of the central nervous system, synaptobrevin 2, a major vesicular SNARE protein, forms a ternary SNARE complex with the plasma membrane SNARE proteins, syntaxin 1 and SNAP25. The affinities of Ca(2+)-dependent interactions between Syt1 and its targets (i.e., SNARE complexes and membranes) are well correlated with the efficacies of the corresponding exocytotic processes. Therefore, different SNARE protein isoforms and membrane lipids, which interact with Syt1 with various affinities, are capable of regulating the efficacy of Syt1-mediated exocytosis. Otoferlin, another type of vesicular C2 domain protein that binds to the membrane in a Ca(2+)-dependent manner, is also involved in the Ca(2+)-triggered synaptic vesicle exocytosis in auditory hair cells. However, the functions of otoferlin in the exocytotic process are not well understood. In addition, at least five different types of synaptic vesicle proteins such as synaptic vesicle protein 2, cysteine string protein α, rab3, synapsin, and a group of proteins containing four transmembrane regions, which includes synaptophysin, synaptogyrin, and secretory carrier membrane protein, are involved in modulating the exocytotic process by regulating the formation and trafficking of synaptic vesicles.
Collapse
Affiliation(s)
- Ok-Ho Shin
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
117
|
Südhof TC. Der molekulare Mechanismus der Neurotransmitterfreisetzung und Nervenzell-Synapsen (Nobel-Aufsatz). Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201406359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
118
|
Südhof TC. The molecular machinery of neurotransmitter release (Nobel lecture). Angew Chem Int Ed Engl 2014; 53:12696-717. [PMID: 25339369 DOI: 10.1002/anie.201406359] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Indexed: 12/18/2022]
Abstract
The most important property of synaptic transmission is its speed, which is crucial for the overall workings of the brain. In his Nobel Lecture, T. C. Südhof explains how the synaptic vesicle and the plasma membrane undergo rapid fusion during neurotransmitter release and how this process is spatially organized, such that opening of Ca(2+) -channels allows rapid translation of the entering Ca(2+) signal into a fusion event.
Collapse
Affiliation(s)
- Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Lorry Lokey SIM1 Building 07-535 Room G1021, 265 Campus Drive, Stanford University School of Medicine, CA 94305 (USA)
| |
Collapse
|
119
|
Kaeser PS. Pushing synaptic vesicles over the RIM. CELLULAR LOGISTICS 2014; 1:106-110. [PMID: 21922075 DOI: 10.4161/cl.1.3.16429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/01/2011] [Accepted: 06/01/2011] [Indexed: 01/19/2023]
Abstract
In a presynaptic nerve terminal, neurotransmitter release is largely restricted to specialized sites called active zones. Active zones consist of a complex protein network, and they organize fusion of synaptic vesicles with the presynaptic plasma membrane in response to action potentials. Rab3-interacting molecules (RIMs) are central components of active zones. In a recent series of experiments, we have systematically dissected the molecular mechanisms by which RIMs operate in synaptic vesicle release. We found that RIMs execute two critical functions of active zones by virtue of independent protein domains. They tether presyanptic Ca(2+) channels to the active zone, and they activate priming of synaptic vesicles by monomerizing homodimeric, constitutively inactive Munc13. These data indicate that RIMs orchestrate synaptic vesicle release into a coherent process. In conjunction with previous studies, they suggest that RIMs form a molecular platform on which plasticity of synaptic vesicle release can operate.
Collapse
Affiliation(s)
- Pascal S Kaeser
- Stanford Institute for Neuro-Innovation & Translational Neurosciences; Department of Molecular and Cellular Physiology; Stanford University; Stanford, CA USA
| |
Collapse
|
120
|
Han Y, Babai N, Kaeser P, Südhof TC, Schneggenburger R. RIM1 and RIM2 redundantly determine Ca2+ channel density and readily releasable pool size at a large hindbrain synapse. J Neurophysiol 2014; 113:255-63. [PMID: 25343783 DOI: 10.1152/jn.00488.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The localization and density of voltage-gated Ca(2+) channels at active zones are essential for the amount and kinetics of transmitter release at synapses. RIM proteins are scaffolding proteins at the active zone that bind to several other presynaptic proteins, including voltage-gated Ca(2+) channel α-subunits. The long isoforms of RIM proteins, which contain NH2-terminal Rab3- and Munc13-interacting domains, as well as a central PDZ domain and two COOH-terminal C2 domains, are encoded by two genes, Rim1 and Rim2. Here, we used the ideal accessibility of the large calyx of Held synapse for direct presynaptic electrophysiology to investigate whether the two Rim genes have redundant, or separate, functions in determining the presynaptic Ca(2+) channel density, and the size of a readily releasable vesicle pool (RRP). Quantitative PCR showed that cochlear nucleus neurons, which include calyx of Held generating neurons, express both RIM1 and RIM2. Conditional genetic inactivation of RIM2 at the calyx of Held led to a subtle reduction in presynaptic Ca(2+) current density, whereas deletion of RIM1 was ineffective. The release efficiency of brief presynaptic Ca(2+) "tail" currents and the RRP were unaffected in conditional single RIM1 and RIM2 knockout (KO) mice, whereas both parameters were strongly reduced in RIM1/2 double KO mice. Thus, despite a somewhat more decisive role for RIM2 in determining presynaptic Ca(2+) channel density, RIM1 and RIM2 can overall replace each other's presynaptic functions at a large relay synapse in the hindbrain, the calyx of Held.
Collapse
Affiliation(s)
- Yunyun Han
- Laboratory of Synaptic Mechanisms, Brain-Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; and
| | - Norbert Babai
- Laboratory of Synaptic Mechanisms, Brain-Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; and
| | - Pascal Kaeser
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California
| | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain-Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; and
| |
Collapse
|
121
|
Chua JJE. Macromolecular complexes at active zones: integrated nano-machineries for neurotransmitter release. Cell Mol Life Sci 2014; 71:3903-16. [PMID: 24912984 PMCID: PMC11113288 DOI: 10.1007/s00018-014-1657-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 02/06/2023]
Abstract
The release of neurotransmitters from synaptic vesicles exocytosing at presynaptic nerve terminals is a critical event in the initiation of synaptic transmission. This event occurs at specialized sites known as active zones. The task of faithfully executing various steps in the process is undertaken by careful orchestration of overlapping sets of molecular nano-machineries upon a core macromolecular scaffold situated at active zones. However, their composition remains incompletely elucidated. This review provides an overview of the role of the active zone in mediating neurotransmitter release and summarizes the recent progress using neuroproteomic approaches to decipher their composition. Key proteins of these nano-machineries are highlighted.
Collapse
Affiliation(s)
- John Jia En Chua
- Department of Neurobiology, Max-Planck-Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany,
| |
Collapse
|
122
|
Rajagopalan A, Schweizer N, Nordenankar K, Nilufar Jahan S, Emilsson L, Wallén-Mackenzie Å. Reduced gene expression levels of Munc13-1 and additional components of the presynaptic exocytosis machinery upon conditional targeting of Vglut2 in the adolescent mouse. Synapse 2014; 68:624-633. [PMID: 25139798 DOI: 10.1002/syn.21776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/04/2014] [Accepted: 07/11/2014] [Indexed: 11/06/2022]
Abstract
Presynaptic proteins orchestrate an intricate interplay of dynamic interactions in order to regulate quantal exocytosis of transmitter-filled vesicles, and their dysregulation might cause neurological and neuropsychiatric dysfunction. Mice carrying a spatiotemporal restriction in the expression of the Vesicular glutamate transporter 2 (Vglut2; aka Slc17a6) in the cortex, amygdala and hippocampal subiculum from the third postnatal week show a strong anxiolytic phenotype and certain behavioral correlates of schizophrenia. To further understand the molecular consequences of this targeted deletion of Vglut2, we performed an unbiased microarray analysis comparing gene expression levels in the subiculum of these conditional Vglut2 knockout mice (Vglut2f/f;CamKII cKO) to those in control littermates. Expression of Unc13C (Munc13-3), a member of the Unc/Munc family, previously shown to be important for glutamatergic transmission, was identified to be significantly down-regulated. Subsequent analysis by quantitative RT-PCR revealed a 50% down-regulation of Munc 13-1, the gene encoding the Unc/Munc subtype described as an essential component in the majority of glutamtergic synapses in the hippocampus. Genes encoding additional components of the presynaptic machinery were also found regulated, including Rab3A, RIM1α, as well as Syntaxin1 and Synaptobrevin. Altered expression levels of these genes were further found in the amygdala and in the retrosplenial group of the cortex, additional regions in which Vglut2 was conditionally targeted. These findings suggest that expression levels of Vglut2 might be important for the maintenance of gene expression in the presynaptic machinery in the adult mouse brain. Synapse 68:624-633, 2014. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aparna Rajagopalan
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden
| | - Nadine Schweizer
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden
| | - Karin Nordenankar
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden
| | - Sultana Nilufar Jahan
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden
| | - Lina Emilsson
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden.,Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, S-752 36, Uppsala, Sweden
| | - Åsa Wallén-Mackenzie
- Department of Neuroscience, Units of Functional Neurobiology and Developmental Genetics, Uppsala University, S-752 37, Uppsala, Sweden
| |
Collapse
|
123
|
Anjum R, Ayoubian H, Schmitz F. Differential synaptic distribution of the scaffold proteins Cask and Caskin1 in the bovine retina. Mol Cell Neurosci 2014; 62:19-29. [DOI: 10.1016/j.mcn.2014.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/24/2014] [Accepted: 08/10/2014] [Indexed: 11/26/2022] Open
|
124
|
Atwood BK, Lovinger DM, Mathur BN. Presynaptic long-term depression mediated by Gi/o-coupled receptors. Trends Neurosci 2014; 37:663-73. [PMID: 25160683 DOI: 10.1016/j.tins.2014.07.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/09/2014] [Accepted: 07/25/2014] [Indexed: 01/20/2023]
Abstract
Long-term depression (LTD) of the efficacy of synaptic transmission is now recognized as an important mechanism for the regulation of information storage and the control of actions, as well as for synapse, neuron, and circuit development. Studies of LTD mechanisms have focused mainly on postsynaptic AMPA-type glutamate receptor trafficking. However, the focus has now expanded to include presynaptically expressed plasticity, the predominant form being initiated by presynaptically expressed Gi/o-coupled metabotropic receptor (Gi/o-GPCR) activation. Several forms of LTD involving activation of different presynaptic Gi/o-GPCRs as a 'common pathway' are described. We review here the literature on presynaptic Gi/o-GPCR-mediated LTD, discuss known mechanisms, gaps in our knowledge, and evaluate whether all Gi/o-GPCRs are capable of inducing presynaptic LTD.
Collapse
Affiliation(s)
- Brady K Atwood
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, 5625 Fishers Lane, MSC 9411, Bethesda, MD 20852-9411, USA
| | - David M Lovinger
- Section on Synaptic Pharmacology, Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, US National Institutes of Health, 5625 Fishers Lane, MSC 9411, Bethesda, MD 20852-9411, USA
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
125
|
Lacoste B, Comin CH, Ben-Zvi A, Kaeser PS, Xu X, Costa LDF, Gu C. Sensory-related neural activity regulates the structure of vascular networks in the cerebral cortex. Neuron 2014; 83:1117-30. [PMID: 25155955 DOI: 10.1016/j.neuron.2014.07.034] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2014] [Indexed: 11/15/2022]
Abstract
Neurovascular interactions are essential for proper brain function. While the effect of neural activity on cerebral blood flow has been extensively studied, whether or not neural activity influences vascular patterning remains elusive. Here, we demonstrate that neural activity promotes the formation of vascular networks in the early postnatal mouse barrel cortex. Using a combination of genetics, imaging, and computational tools to allow simultaneous analysis of neuronal and vascular components, we found that vascular density and branching were decreased in the barrel cortex when sensory input was reduced by either a complete deafferentation, a genetic impairment of neurotransmitter release at thalamocortical synapses, or a selective reduction of sensory-related neural activity by whisker plucking. In contrast, enhancement of neural activity by whisker stimulation led to an increase in vascular density and branching. The finding that neural activity is necessary and sufficient to trigger alterations of vascular networks reveals an important feature of neurovascular interactions.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Cesar H Comin
- IFSC, University of Sao Paulo, Sao Carlos, SP, Brazil
| | - Ayal Ben-Zvi
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Xiaoyin Xu
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
126
|
Wasik U, Filipek A. Non-nuclear function of sumoylated proteins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:2878-2885. [PMID: 25110347 DOI: 10.1016/j.bbamcr.2014.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/11/2014] [Accepted: 07/30/2014] [Indexed: 12/16/2022]
Abstract
Post-translational modification by the SUMO moiety is now regarded as one of the key regulatory modifications in eukaryotic cells. Up to now, plenty of sumoylated proteins have been found to be involved in nuclear processes such as chromatin organization, transcription and DNA repair as well as in other cellular functions. Since the number of data concerning sumoylated proteins and their function outside the nucleus has grown rapidly, in this review we summarized the results describing the non-nuclear role of SUMO substrates. In particular, we focused on the role of sumoylation in the regulation of channel activity, receptor function, G-protein signaling, activity of enzymes, cytoskeletal organization, exocytosis, autophagy and mitochondrial dynamics.
Collapse
Affiliation(s)
- Urszula Wasik
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Warsaw, Poland.
| |
Collapse
|
127
|
3D structure generation, virtual screening and docking of human Ras-associated binding (Rab3A) protein involved in tumourigenesis. Mol Biol Rep 2014; 41:3951-9. [DOI: 10.1007/s11033-014-3263-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
|
128
|
Ramírez-Franco J, Bartolomé-Martín D, Alonso B, Torres M, Sánchez-Prieto J. Cannabinoid type 1 receptors transiently silence glutamatergic nerve terminals of cultured cerebellar granule cells. PLoS One 2014; 9:e88594. [PMID: 24533119 PMCID: PMC3922925 DOI: 10.1371/journal.pone.0088594] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 01/13/2014] [Indexed: 12/25/2022] Open
Abstract
Cannabinoid receptors are the most abundant G protein-coupled receptors in the brain and they mediate retrograde short-term inhibition of neurotransmitter release, as well as long-term depression of synaptic transmission at many excitatory synapses. The induction of presynaptically silent synapses is a means of modulating synaptic strength, which is important for synaptic plasticity. Persistent activation of cannabinoid type 1 receptors (CB1Rs) mutes GABAergic terminals, although it is unclear if CB1Rs can also induce silencing at glutamatergic synapses. Cerebellar granule cells were transfected with VGLUT1-pHluorin to visualise the exo-endocytotic cycle. We found that prolonged stimulation (10 min) of cannabinoid receptors with the agonist HU-210 induces the silencing of previously active synapses. However, the presynaptic silencing induced by HU-210 is transient as it reverses after 20 min. cAMP with forskolin prevented CB1R-induced synaptic silencing, via activation of the Exchange Protein directly Activated by cAMP (Epac). Furthermore, Epac activation accelerated awakening of already silent boutons. Electron microscopy revealed that silencing was associated with synaptic vesicle (SV) redistribution within the nerve terminal, which diminished the number of vesicles close to the active zone of the plasma membrane. Finally, by combining functional and immunocytochemical approaches, we observed a strong correlation between the release capacity of the nerve terminals and RIM1α protein content, but not that of Munc13-1 protein. These results suggest that prolonged stimulation of cannabinoid receptors can transiently silence glutamatergic nerve terminals.
Collapse
Affiliation(s)
- Jorge Ramírez-Franco
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - David Bartolomé-Martín
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Beatris Alonso
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Magdalena Torres
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- * E-mail: (JSP); (MT)
| | - José Sánchez-Prieto
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
- * E-mail: (JSP); (MT)
| |
Collapse
|
129
|
Schmitz F. Presynaptic [Ca(2+)] and GCAPs: aspects on the structure and function of photoreceptor ribbon synapses. Front Mol Neurosci 2014; 7:3. [PMID: 24567702 PMCID: PMC3915146 DOI: 10.3389/fnmol.2014.00003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/15/2014] [Indexed: 12/21/2022] Open
Abstract
Changes in intracellular calcium ions [Ca2+] play important roles in photoreceptor signaling. Consequently, intracellular [Ca2+] levels need to be tightly controlled. In the light-sensitive outer segments (OS) of photoreceptors, Ca2+ regulates the activity of retinal guanylate cyclases thus playing a central role in phototransduction and light-adaptation by restoring light-induced decreases in cGMP. In the synaptic terminals, changes of intracellular Ca2+ trigger various aspects of neurotransmission. Photoreceptors employ tonically active ribbon synapses that encode light-induced, graded changes of membrane potential into modulation of continuous synaptic vesicle exocytosis. The active zones of ribbon synapses contain large electron-dense structures, synaptic ribbons, that are associated with large numbers of synaptic vesicles. Synaptic coding at ribbon synapses differs from synaptic coding at conventional (phasic) synapses. Recent studies revealed new insights how synaptic ribbons are involved in this process. This review focuses on the regulation of [Ca2+] in presynaptic photoreceptor terminals and on the function of a particular Ca2+-regulated protein, the neuronal calcium sensor protein GCAP2 (guanylate cyclase-activating protein-2) in the photoreceptor ribbon synapse. GCAP2, an EF-hand-containing protein plays multiple roles in the OS and in the photoreceptor synapse. In the OS, GCAP2 works as a Ca2+-sensor within a Ca2+-regulated feedback loop that adjusts cGMP levels. In the photoreceptor synapse, GCAP2 binds to RIBEYE, a component of synaptic ribbons, and mediates Ca2+-dependent plasticity at that site. Possible mechanisms are discussed.
Collapse
Affiliation(s)
- Frank Schmitz
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Medical School Homburg/Saar, Saarland University Saarland, Germany
| |
Collapse
|
130
|
Abstract
During an action potential, Ca(2+) entering a presynaptic terminal triggers synaptic vesicle exocytosis and neurotransmitter release in less than a millisecond. How does Ca(2+) stimulate release so rapidly and precisely? Work over the last decades revealed that Ca(2+) binding to synaptotagmin triggers release by stimulating synaptotagmin binding to a core fusion machinery composed of SNARE and SM proteins that mediates membrane fusion during exocytosis. Complexin adaptor proteins assist synaptotagmin by activating and clamping this core fusion machinery. Synaptic vesicles containing synaptotagmin are positioned at the active zone, the site of vesicle fusion, by a protein complex containing RIM proteins. RIM proteins activate docking and priming of synaptic vesicles and simultaneously recruit Ca(2+) channels to active zones, thereby connecting in a single complex primed synaptic vesicles to Ca(2+) channels. This architecture allows direct flow of Ca(2+) ions from Ca(2+) channels to synaptotagmin, which then triggers fusion, thus mediating tight millisecond coupling of an action potential to neurotransmitter release.
Collapse
Affiliation(s)
- Thomas C Südhof
- Department of Molecular and Cellular Physiology, and Howard Hughes Medical Institute, Lorry Lokey SIM1 Building, 265 Campus Drive, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
131
|
Joshi CS, Khan SA, Khole VV. Regulation of acrosome reaction by Liprin α3, LAR and its ligands in mouse spermatozoa. Andrology 2013; 2:165-74. [PMID: 24327330 DOI: 10.1111/j.2047-2927.2013.00167] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 11/01/2013] [Accepted: 11/04/2013] [Indexed: 11/30/2022]
Abstract
Zona pellucida-based induction of acrosome reaction (AR) is a popular and well-accepted hypothesis. However, this hypothesis is being challenged in recent years and it has been proposed that the cumulus cells might be the site of AR. In our previous study, we reported the presence of a synaptic protein Liprin α3 on sperm acrosome, and proposed its role in AR. This study was designed to understand the role of Liprin α3 and its interacting proteins in regulation of AR. It is observed that the presence of anti-Liprin α3 antibody inhibits the process of AR. Colocalization experiments demonstrate the coexistence of leucocyte antigen related (LAR) protein, Rab-interacting molecule (RIM) and Liprin α3 on sperm acrosome thereby completing the identification of all the members of RIM/MUNC/Rab3A/liprinα complex required for membrane fusion. This study demonstrates the effect of LAR ligands such as Syndecans, Nidogens and LAR wedge domain peptide on AR. We could see an increase in AR in presence of these ligands. On the basis of these data, we speculate that in presence of ligands or wedge peptide, LAR undergoes dimerization leading to inhibition of phosphatase activity and increase in AR. The presence of one of the ligands Syndecan-1 on cumulus cells led us to hypothesize that it is Syndecan which induces AR in vivo and thus another site of AR could lie in cumulus.
Collapse
Affiliation(s)
- C S Joshi
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health, Mumbai, India
| | | | | |
Collapse
|
132
|
Kittelmann M, Hegermann J, Goncharov A, Taru H, Ellisman MH, Richmond JE, Jin Y, Eimer S. Liprin-α/SYD-2 determines the size of dense projections in presynaptic active zones in C. elegans. J Cell Biol 2013; 203:849-63. [PMID: 24322429 PMCID: PMC3857474 DOI: 10.1083/jcb.201302022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 11/06/2013] [Indexed: 01/23/2023] Open
Abstract
Synaptic vesicle (SV) release is spatially and temporally regulated by a network of proteins that form the presynaptic active zone (AZ). The hallmark of most AZs is an electron-dense projection (DP) surrounded by SVs. Despite their importance for our understanding of triggered SV release, high-resolution analyses of DP structures are limited. Using electron microscopy, we show that DPs at Caenorhabditis elegans neuromuscular junctions (NMJs) were highly structured, composed of building units forming bays in which SVs are docked to the AZ membrane. Furthermore, larger ribbonlike DPs that were multimers of the NMJ building unit are found at synapses between inter- and motoneurons. We also demonstrate that DP size is determined by the activity of the AZ protein SYD-2/Liprin-α. Whereas loss of syd-2 function led to smaller DPs, syd-2 gain-of-function mutants displayed larger ribbonlike DPs through increased recruitment of ELKS-1/ELKS. Therefore, our data suggest that a main role of SYD-2/Liprin-α in synaptogenesis is to regulate the polymerization of DPs.
Collapse
Affiliation(s)
- Maike Kittelmann
- European Neuroscience Institute, 37077 Göttingen, Germany
- Cellular Neurobiology, Schwann-Schleiden-Centre for Molecular Cell Biology, 37077 Göttingen, Germany
| | - Jan Hegermann
- European Neuroscience Institute, 37077 Göttingen, Germany
- Center for Molecular Physiology of the Brain, 37073 Göttingen, Germany
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Alexandr Goncharov
- Howard Hughes Medical Institute, Division of Biological Sciences; and Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neurosciences; University of California, San Diego, La Jolla, CA 92093
| | - Hidenori Taru
- Howard Hughes Medical Institute, Division of Biological Sciences; and Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neurosciences; University of California, San Diego, La Jolla, CA 92093
- Laboratory of Neuronal Cell Biology, Graduate School of Pharmaceutical Sciences and Creative Research Institute, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Mark H. Ellisman
- Howard Hughes Medical Institute, Division of Biological Sciences; and Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neurosciences; University of California, San Diego, La Jolla, CA 92093
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607
| | - Yishi Jin
- Howard Hughes Medical Institute, Division of Biological Sciences; and Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neurosciences; University of California, San Diego, La Jolla, CA 92093
| | - Stefan Eimer
- European Neuroscience Institute, 37077 Göttingen, Germany
- Center for Molecular Physiology of the Brain, 37073 Göttingen, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79108 Freiburg, Germany
| |
Collapse
|
133
|
Girach F, Craig TJ, Rocca DL, Henley JM. RIM1α SUMOylation is required for fast synaptic vesicle exocytosis. Cell Rep 2013; 5:1294-301. [PMID: 24290762 PMCID: PMC3898736 DOI: 10.1016/j.celrep.2013.10.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/01/2013] [Accepted: 10/24/2013] [Indexed: 01/14/2023] Open
Abstract
The rapid, activity-dependent quantal presynaptic release of neurotransmitter is vital for brain function. The complex process of vesicle priming, fusion, and retrieval is very precisely controlled and requires the spatiotemporal coordination of multiple protein-protein interactions. Here, we show that posttranslational modification of the active zone protein Rab3-interacting molecule 1α (RIM1α) by the small ubiquitin-like modifier 1 (SUMO-1) functions as a molecular switch to direct these interactions and is essential for fast synaptic vesicle exocytosis. RIM1α SUMOylation at lysine residue K502 facilitates the clustering of CaV2.1 calcium channels and enhances the Ca2+ influx necessary for vesicular release, whereas non-SUMOylated RIM1α participates in the docking/priming of synaptic vesicles and maintenance of active zone structure. These results demonstrate that SUMOylation of RIM1α is a key determinant of rapid, synchronous neurotransmitter release, and the SUMO-mediated “switching” of RIM1α between binding proteins provides insight into the mechanisms underpinning synaptic function and dysfunction. RIM1α is SUMOylated at K502 SUMOylation of RIM1α is required for its PDZ-dependent binding to CaV2.1 Preventing RIM1α SUMOylation reduces CaV2.1 clustering and Ca2+ entry RIM1α SUMOylation is necessary for fast synaptic vesicle exocytosis
Collapse
Affiliation(s)
- Fatima Girach
- Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Tim J Craig
- Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Daniel L Rocca
- Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Jeremy M Henley
- Department of Biochemistry, School of Medical Sciences, University Walk, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
134
|
Kaeser PS, Regehr WG. Molecular mechanisms for synchronous, asynchronous, and spontaneous neurotransmitter release. Annu Rev Physiol 2013; 76:333-63. [PMID: 24274737 DOI: 10.1146/annurev-physiol-021113-170338] [Citation(s) in RCA: 311] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Most neuronal communication relies upon the synchronous release of neurotransmitters, which occurs through synaptic vesicle exocytosis triggered by action potential invasion of a presynaptic bouton. However, neurotransmitters are also released asynchronously with a longer, variable delay following an action potential or spontaneously in the absence of action potentials. A compelling body of research has identified roles and mechanisms for synchronous release, but asynchronous release and spontaneous release are less well understood. In this review, we analyze how the mechanisms of the three release modes overlap and what molecular pathways underlie asynchronous and spontaneous release. We conclude that the modes of release have key fusion processes in common but may differ in the source of and necessity for Ca(2+) to trigger release and in the identity of the Ca(2+) sensor for release.
Collapse
Affiliation(s)
- Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115; ,
| | | |
Collapse
|
135
|
Ramírez-Franco J, Alonso B, Bartolomé-Martín D, Sánchez-Prieto J, Torres M. Studying synaptic efficiency by post-hoc immunolabelling. BMC Neurosci 2013; 14:127. [PMID: 24138605 PMCID: PMC3854067 DOI: 10.1186/1471-2202-14-127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 10/10/2013] [Indexed: 11/10/2022] Open
Abstract
Background In terms of vesicular recycling, synaptic efficiency is a key determinant of the fidelity of synaptic transmission. The ability of a presynaptic terminal to reuse its vesicular content is thought to be a signature of synaptic maturity and this process depends on the activity of several proteins that govern exo/endocytosis. Upon stimulation, individual terminals in networks of cultured cerebellar granule neurons exhibit heterogeneous exocytic responses, which reflect the distinct states of maturity and plasticity intrinsic to individual synaptic terminals. This dynamic scenario serves as the substrate for processes such as scaling, plasticity and synaptic weight redistribution. Presynaptic strength has been associated with the activity of several types of proteins, including the scaffolding proteins that form the active zone cytomatrix and the proteins involved in presynaptic exocytosis. Methods We have combined fluorescence imaging techniques using the styryl dye FM1-43 in primary cultures of cerebellar granule cells with subsequent post-hoc immunocytochemistry in order to study synaptic efficiency in terms of vesicular release. We describe a protocol to easily quantify these results with minimal user intervention. Results In this study we describe a technique that specifically correlates presynaptic activity with the levels of presynaptic markers. This method involves the use of the styryl dye FM1-43 to estimate the release capacity of a synaptic terminal, and the subsequent post-hoc immunolabelling of thousands of individual nerve terminals. We observed a strong correlation between the release capacity of the nerve terminal and the levels of the RIM1α but not the Munc13-1 protein in the active zone. Conclusions Our findings support those of previous studies and point out to RIM1α as a crucial factor in determining synaptic efficiency. These results also demonstrate that this technique is a useful tool to analyse the molecular differences underlying the heterogeneous responses exhibited by neuronal networks.
Collapse
Affiliation(s)
| | | | | | - José Sánchez-Prieto
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid 28040, Spain.
| | | |
Collapse
|
136
|
Yang Y, Calakos N. Presynaptic long-term plasticity. Front Synaptic Neurosci 2013; 5:8. [PMID: 24146648 PMCID: PMC3797957 DOI: 10.3389/fnsyn.2013.00008] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 09/09/2013] [Indexed: 01/01/2023] Open
Abstract
Long-term synaptic plasticity is a major cellular substrate for learning, memory, and behavioral adaptation. Although early examples of long-term synaptic plasticity described a mechanism by which postsynaptic signal transduction was potentiated, it is now apparent that there is a vast array of mechanisms for long-term synaptic plasticity that involve modifications to either or both the presynaptic terminal and postsynaptic site. In this article, we discuss current and evolving approaches to identify presynaptic mechanisms as well as discuss their limitations. We next provide examples of the diverse circuits in which presynaptic forms of long-term synaptic plasticity have been described and discuss the potential contribution this form of plasticity might add to circuit function. Finally, we examine the present evidence for the molecular pathways and cellular events underlying presynaptic long-term synaptic plasticity.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pediatrics, Stanford University School of Medicine Stanford, CA, USA
| | | |
Collapse
|
137
|
Südhof TC. A molecular machine for neurotransmitter release: synaptotagmin and beyond. Nat Med 2013; 19:1227-31. [DOI: 10.1038/nm.3338] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
138
|
Ferrero JJ, Alvarez AM, Ramírez-Franco J, Godino MC, Bartolomé-Martín D, Aguado C, Torres M, Luján R, Ciruela F, Sánchez-Prieto J. β-Adrenergic receptors activate exchange protein directly activated by cAMP (Epac), translocate Munc13-1, and enhance the Rab3A-RIM1α interaction to potentiate glutamate release at cerebrocortical nerve terminals. J Biol Chem 2013; 288:31370-85. [PMID: 24036110 DOI: 10.1074/jbc.m113.463877] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The adenylyl cyclase activator forskolin facilitates synaptic transmission presynaptically via cAMP-dependent protein kinase (PKA). In addition, cAMP also increases glutamate release via PKA-independent mechanisms, although the downstream presynaptic targets remain largely unknown. Here, we describe the isolation of a PKA-independent component of glutamate release in cerebrocortical nerve terminals after blocking Na(+) channels with tetrodotoxin. We found that 8-pCPT-2'-O-Me-cAMP, a specific activator of the exchange protein directly activated by cAMP (Epac), mimicked and occluded forskolin-induced potentiation of glutamate release. This Epac-mediated increase in glutamate release was dependent on phospholipase C, and it increased the hydrolysis of phosphatidylinositol 4,5-bisphosphate. Moreover, the potentiation of glutamate release by Epac was independent of protein kinase C, although it was attenuated by the diacylglycerol-binding site antagonist calphostin C. Epac activation translocated the active zone protein Munc13-1 from soluble to particulate fractions; it increased the association between Rab3A and RIM1α and redistributed synaptic vesicles closer to the presynaptic membrane. Furthermore, these responses were mimicked by the β-adrenergic receptor (βAR) agonist isoproterenol, consistent with the immunoelectron microscopy and immunocytochemical data demonstrating presynaptic expression of βARs in a subset of glutamatergic synapses in the cerebral cortex. Based on these findings, we conclude that βARs couple to a cAMP/Epac/PLC/Munc13/Rab3/RIM-dependent pathway to enhance glutamate release at cerebrocortical nerve terminals.
Collapse
Affiliation(s)
- Jose J Ferrero
- From the Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Atlas D. The Voltage-Gated Calcium Channel Functions as the Molecular Switch of Synaptic Transmission. Annu Rev Biochem 2013; 82:607-35. [DOI: 10.1146/annurev-biochem-080411-121438] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Daphne Atlas
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904 Jerusalem, Israel;
| |
Collapse
|
140
|
Zabouri N, Haverkamp S. Calcium channel-dependent molecular maturation of photoreceptor synapses. PLoS One 2013; 8:e63853. [PMID: 23675510 PMCID: PMC3652833 DOI: 10.1371/journal.pone.0063853] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/08/2013] [Indexed: 01/08/2023] Open
Abstract
Several studies have shown the importance of calcium channels in the development and/or maturation of synapses. The CaV1.4(α1F) knockout mouse is a unique model to study the role of calcium channels in photoreceptor synapse formation. It features abnormal ribbon synapses and aberrant cone morphology. We investigated the expression and targeting of several key elements of ribbon synapses and analyzed the cone morphology in the CaV1.4(α1F) knockout retina. Our data demonstrate that most abnormalities occur after eye opening. Indeed, scaffolding proteins such as Bassoon and RIM2 are properly targeted at first, but their expression and localization are not maintained in adulthood. This indicates that either calcium or the CaV1.4 channel, or both are necessary for the maintenance of their normal expression and distribution in photoreceptors. Other proteins, such as Veli3 and PSD-95, also display abnormal expression in rods prior to eye opening. Conversely, vesicle related proteins appear normal. Our data demonstrate that the CaV1.4 channel is important for maintaining scaffolding proteins in the ribbon synapse but less vital for proteins related to vesicular release. This study also confirms that in adult retinae, cones show developmental features such as sprouting and synaptogenesis. Overall we present evidence that in the absence of the CaV1.4 channel, photoreceptor synapses remain immature and are unable to stabilize.
Collapse
Affiliation(s)
- Nawal Zabouri
- Neuroanatomy, Max-Planck-Institute for Brain Research, Frankfurt am Main, Germany.
| | | |
Collapse
|
141
|
Ohtsuka T. CAST: Functional scaffold for the integrity of the presynaptic active zone. Neurosci Res 2013; 76:10-5. [DOI: 10.1016/j.neures.2013.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/11/2013] [Accepted: 03/11/2013] [Indexed: 12/24/2022]
|
142
|
Abstract
The large isoforms of the Rab3 interacting molecule (RIM) family, RIM1α/β and RIM2α/β, have been shown to be centrally involved in mediating presynaptic active zone function. The RIM protein family contains two additional small isoforms, RIM3γ and RIM4γ, which are composed only of the RIM-specific C-terminal C2B domain and varying N-terminal sequences and whose function remains to be elucidated. Here, we report that both, RIM3γ and RIM4γ, play an essential role for the development of neuronal arborization and of dendritic spines independent of synaptic function. γ-RIM knock-down in rat primary neuronal cultures and in vivo resulted in a drastic reduction in the complexity of neuronal arborization, affecting both axonal and dendritic outgrowth, independent of the time point of γ-RIM downregulation during dendrite development. Rescue experiments revealed that the phenotype is caused by a function common to both γ-RIMs. These findings indicate that γ-RIMs are involved in cell biological functions distinct from the regulation of synaptic vesicle exocytosis and play a role in the molecular mechanisms controlling the establishment of dendritic complexity and axonal outgrowth.
Collapse
|
143
|
Fife, a Drosophila Piccolo-RIM homolog, promotes active zone organization and neurotransmitter release. J Neurosci 2013. [PMID: 23197698 DOI: 10.1523/jneurosci.3267-12.2012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal communication depends on the precisely orchestrated release of neurotransmitter at specialized sites called active zones (AZs). A small number of scaffolding and cytoskeletal proteins comprising the cytomatrix of the active zone (CAZ) are thought to organize the architecture and functional properties of AZs. The majority of CAZ proteins are evolutionarily conserved, underscoring the fundamental similarities in neurotransmission at all synapses. However, core CAZ proteins Piccolo and Bassoon have long been believed exclusive to vertebrates, raising intriguing questions about the conservation of the molecular mechanisms that regulate presynaptic properties. Here, we present the identification of a piccolo-rim-related gene in invertebrates, together with molecular phylogenetic analyses that indicate the encoded proteins may represent Piccolo orthologs. In accordance, we find that the Drosophila homolog, Fife, is neuronal and localizes to presynaptic AZs. To investigate the in vivo function of Fife, we generated a deletion of the fife locus. We find that evoked neurotransmitter release is substantially decreased in fife mutants and loss of fife results in motor deficits. Through morphological analysis of fife synapses, we identify underlying AZ abnormalities including pervasive presynaptic membrane detachments and reduced synaptic vesicle clustering. Our data demonstrate the conservation of a Piccolo-related protein in invertebrates and identify critical roles for Fife in regulating AZ structure and function. These findings suggest the CAZ is more conserved than previously thought, and open the door to a more complete understanding of how CAZ proteins regulate presynaptic structure and function through genetic studies in simpler model systems.
Collapse
|
144
|
RIM controls homeostatic plasticity through modulation of the readily-releasable vesicle pool. J Neurosci 2013; 32:16574-85. [PMID: 23175813 DOI: 10.1523/jneurosci.0981-12.2012] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rab3 interacting molecules (RIMs) are evolutionarily conserved scaffolding proteins that are located at presynaptic active zones. In the mammalian nervous system, RIMs have two major activities that contribute to the fidelity of baseline synaptic transmission: they concentrate calcium channels at the active zone and facilitate synaptic vesicle docking/priming. Here we confirm that RIM has an evolutionarily conserved function at the Drosophila neuromuscular junction and then define a novel role for RIM during homeostatic synaptic plasticity. We show that loss of RIM disrupts baseline vesicle release, diminishes presynaptic calcium influx, and diminishes the size of the readily-releasable pool (RRP) of synaptic vesicles, consistent with known activities of RIM. However, loss of RIM also completely blocks the homeostatic enhancement of presynaptic neurotransmitter release that normally occurs after inhibition of postsynaptic glutamate receptors, a process termed synaptic homeostasis. It is established that synaptic homeostasis requires enhanced presynaptic calcium influx as a mechanism to potentiate vesicle release. However, despite a defect in baseline calcium influx in rim mutants, the homeostatic modulation of calcium influx proceeds normally. Synaptic homeostasis is also correlated with an increase in the size of the RRP of synaptic vesicles, although the mechanism remains unknown. Here we demonstrate that the homeostatic modulation of the RRP is blocked in the rim mutant background. Therefore, RIM-dependent modulation of the RRP is a required step during homeostatic plasticity. By extension, homeostatic plasticity appears to require two genetically separable processes, the enhancement of presynaptic calcium influx and a RIM-dependent modulation of the RRP.
Collapse
|
145
|
RIM promotes calcium channel accumulation at active zones of the Drosophila neuromuscular junction. J Neurosci 2013; 32:16586-96. [PMID: 23175814 DOI: 10.1523/jneurosci.0965-12.2012] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Synaptic communication requires the controlled release of synaptic vesicles from presynaptic axon terminals. Release efficacy is regulated by the many proteins that comprise the presynaptic release apparatus, including Ca(2+) channels and proteins that influence Ca(2+) channel accumulation at release sites. Here we identify Drosophila RIM (Rab3 interacting molecule) and demonstrate that it localizes to active zones at the larval neuromuscular junction. In Drosophila RIM mutants, there is a large decrease in evoked synaptic transmission because of a significant reduction in both the clustering of Ca(2+) channels and the size of the readily releasable pool of synaptic vesicles at active zones. Hence, RIM plays an evolutionarily conserved role in regulating synaptic calcium channel localization and readily releasable pool size. Because RIM has traditionally been studied as an effector of Rab3 function, we investigate whether RIM is involved in the newly identified function of Rab3 in the distribution of presynaptic release machinery components across release sites. Bruchpilot (Brp), an essential component of the active zone cytomatrix T bar, is unaffected by RIM disruption, indicating that Brp localization and distribution across active zones does not require wild-type RIM. In addition, larvae containing mutations in both RIM and rab3 have reduced Ca(2+) channel levels and a Brp distribution that is very similar to that of the rab3 single mutant, indicating that RIM functions to regulate Ca(2+) channel accumulation but is not a Rab3 effector for release machinery distribution across release sites.
Collapse
|
146
|
Rizo J, Südhof TC. The Membrane Fusion Enigma: SNAREs, Sec1/Munc18 Proteins, and Their Accomplices—Guilty as Charged? Annu Rev Cell Dev Biol 2012; 28:279-308. [DOI: 10.1146/annurev-cellbio-101011-155818] [Citation(s) in RCA: 318] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Josep Rizo
- Departments of Biophysics, Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390;
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford University Medical School, Stanford, California 94305;
| |
Collapse
|
147
|
Joshi CS, Suryawanshi AR, Khan SA, Balasinor NH, Khole VV. Liprin α3: a putative estrogen regulated acrosomal protein. Histochem Cell Biol 2012; 139:535-48. [PMID: 23124857 DOI: 10.1007/s00418-012-1044-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2012] [Indexed: 11/28/2022]
Abstract
Liprin α3 was reported for the first time using sperm proteomics. Present study reports its localization on sperm and immunochemical characterization. Liprin α3 is identified as a 133 kDa protein in testis and epididymal protein extracts. In testis, immunohistochemical localization was seen in pachytenes, diplotenes, round spermatids whereas it was localized in the epithelial cells and luminal sperm in all the three regions of epididymis. Protein was localized in acrosome of rat sperm, which was further confirmed by sequential treatment of sperm with hypertonic solution. In the spermatogenic cells the protein was found to be located in developing acrosome as evident by its co-localization with Golgi marker. Protein was found to be developmentally regulated. In silico analysis of Liprin α3 revealed presence of the estrogen responsive elements upstream to initiation site and its regulation by estrogen was experimentally validated using a tamoxifen treated rat model. Western blot analysis of epididymosomes showed the presence of Liprin α3, indicating its involvement in trafficking of vesicle. The protein expression was seen in both mouse and human sperm indicating conserved nature and a probable role in acrosome reaction.
Collapse
Affiliation(s)
- Chetanchandra S Joshi
- Department of Gamete Immunobiology, National Institute for Research in Reproductive Health (ICMR), J. M. Street, Parel, Mumbai, 400012, India
| | | | | | | | | |
Collapse
|
148
|
A window into domain amplification through Piccolo in teleost fish. G3-GENES GENOMES GENETICS 2012; 2:1325-39. [PMID: 23173084 PMCID: PMC3484663 DOI: 10.1534/g3.112.003624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/31/2012] [Indexed: 12/20/2022]
Abstract
I describe and characterize the extensive amplification of the zinc finger domain of Piccolo selectively in teleost fish. Piccolo and Bassoon are partially functionally redundant and play roles in regulating the pool of neurotransmitter-filled synaptic vesicles present at synapses. In mice, each protein contains two N-terminal zinc finger domains that have been implicated in interacting with synaptic vesicles. In all teleosts examined, both the Bassoon and Piccolo genes are duplicated. Both teleost bassoon genes and one piccolo gene show very similar domain structure and intron-exon organization to their mouse homologs. In contrast, in piccolo b a single exon that encodes a zinc finger domain is amplified 8 to 16 times in different teleost species. Analysis of the amplified exons suggests they were added and/or deleted from the gene as individual exons in rare events that are likely the result of unequal crossovers between homologous sequences. Surprisingly, the structure of the repeats from cod and zebrafish suggest that amplification of this exon has occurred independently multiple times in the teleost lineage. Based on the structure of the exons, I propose a model in which selection for high sequence similarity at the 5′ and 3′ ends of the exon drives amplification of the repeats and diversity in repeat length likely promotes the stability of the repeated exons by minimizing the likelihood of mispairing of adjacent repeat sequences. Further analysis of piccolo b in teleosts should provide a window through which to examine the process of domain amplification.
Collapse
|
149
|
Otoferlin: a multi-C2 domain protein essential for hearing. Trends Neurosci 2012; 35:671-80. [DOI: 10.1016/j.tins.2012.08.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/25/2012] [Accepted: 08/13/2012] [Indexed: 12/21/2022]
|
150
|
Kasai H, Takahashi N, Tokumaru H. Distinct Initial SNARE Configurations Underlying the Diversity of Exocytosis. Physiol Rev 2012; 92:1915-64. [DOI: 10.1152/physrev.00007.2012] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The dynamics of exocytosis are diverse and have been optimized for the functions of synapses and a wide variety of cell types. For example, the kinetics of exocytosis varies by more than five orders of magnitude between ultrafast exocytosis in synaptic vesicles and slow exocytosis in large dense-core vesicles. However, in all cases, exocytosis is mediated by the same fundamental mechanism, i.e., the assembly of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. It is often assumed that vesicles need to be docked at the plasma membrane and SNARE proteins must be preassembled before exocytosis is triggered. However, this model cannot account for the dynamics of exocytosis recently reported in synapses and other cells. For example, vesicles undergo exocytosis without prestimulus docking during tonic exocytosis of synaptic vesicles in the active zone. In addition, epithelial and hematopoietic cells utilize cAMP and kinases to trigger slow exocytosis of nondocked vesicles. In this review, we summarize the manner in which the diversity of exocytosis reflects the initial configurations of SNARE assembly, including trans-SNARE, binary-SNARE, unitary-SNARE, and cis-SNARE configurations. The initial SNARE configurations depend on the particular SNARE subtype (syntaxin, SNAP25, or VAMP), priming proteins (Munc18, Munc13, CAPS, complexin, or snapin), triggering proteins (synaptotagmins, Doc2, and various protein kinases), and the submembraneous cytomatrix, and they are the key to determining the kinetics of subsequent exocytosis. These distinct initial configurations will help us clarify the common SNARE assembly processes underlying exocytosis and membrane trafficking in eukaryotic cells.
Collapse
Affiliation(s)
- Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; and Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, Kagawa, Japan
| | - Noriko Takahashi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; and Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, Kagawa, Japan
| | - Hiroshi Tokumaru
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; and Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, Kagawa, Japan
| |
Collapse
|