101
|
Voleti R, Jaczynska K, Rizo J. Ca 2+-dependent release of synaptotagmin-1 from the SNARE complex on phosphatidylinositol 4,5-bisphosphate-containing membranes. eLife 2020; 9:57154. [PMID: 32808925 PMCID: PMC7498268 DOI: 10.7554/elife.57154] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/12/2020] [Indexed: 11/13/2022] Open
Abstract
The Ca2+ sensor synaptotagmin-1 and the SNARE complex cooperate to trigger neurotransmitter release. Structural studies elucidated three distinct synaptotagmin-1-SNARE complex binding modes involving 'polybasic', 'primary' and 'tripartite' interfaces of synaptotagmin-1. We investigated these interactions using NMR and fluorescence spectroscopy. Synaptotagmin-1 binds to the SNARE complex through the polybasic and primary interfaces in solution. Ca2+-free synaptotagmin-1 binds to SNARE complexes anchored on PIP2-containing nanodiscs. R398Q/R399Q and E295A/Y338W mutations at the primary interface, which strongly impair neurotransmitter release, disrupt and enhance synaptotagmin-1-SNARE complex binding, respectively. Ca2+ induces tight binding of synaptotagmin-1 to PIP2-containing nanodiscs, disrupting synaptotagmin-1-SNARE interactions. Specific effects of mutations in the polybasic region on Ca2+-dependent synaptotagmin-1-PIP2-membrane interactions correlate with their effects on release. Our data suggest that synaptotagmin-1 binds to the SNARE complex through the primary interface and that Ca2+ releases this interaction, inducing PIP2/membrane binding and allowing cooperation between synaptotagmin-1 and the SNAREs in membrane fusion to trigger release.
Collapse
Affiliation(s)
- Rashmi Voleti
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Klaudia Jaczynska
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
102
|
Prasad R, Zhou HX. Membrane Association and Functional Mechanism of Synaptotagmin-1 in Triggering Vesicle Fusion. Biophys J 2020; 119:1255-1265. [PMID: 32882186 DOI: 10.1016/j.bpj.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/23/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Upon Ca2+ influx, synaptic vesicles fuse with the presynaptic plasma membrane (PM) to release neurotransmitters. Membrane fusion is triggered by synaptotagmin-1, a transmembrane protein in the vesicle membrane (VM), but the mechanism is under debate. Synaptotagmin-1 contains a single transmembrane helix (TM) and two tandem C2 domains (C2A and C2B). This study aimed to use molecular dynamics simulations to elucidate how Ca2+-bound synaptotagmin-1, by simultaneously associating with VM and PM, brings them together for fusion. Although C2A stably associates with VM via two Ca2+-binding loops, C2B has a propensity to partially dissociate. Importantly, an acidic motif in the TM-C2A linker competes with VM for interacting with C2B, thereby flipping its orientation to face PM. Subsequently, C2B readily associates with PM via a polybasic cluster and a Ca2+-binding loop. The resulting mechanistic model for the triggering of membrane fusion by synaptotagmin-1 reconciles many experimental observations.
Collapse
Affiliation(s)
- Ramesh Prasad
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois; Department of Physics, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
103
|
Neurotrophic factors and target-specific retrograde signaling interactions define the specificity of classical and neuropeptide cotransmitter release at identified Lymnaea synapses. Sci Rep 2020; 10:13526. [PMID: 32782285 PMCID: PMC7419297 DOI: 10.1038/s41598-020-70322-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/22/2020] [Indexed: 12/26/2022] Open
Abstract
Many neurons concurrently and/or differentially release multiple neurotransmitter substances to selectively modulate the activity of distinct postsynaptic targets within a network. However, the molecular mechanisms that produce synaptic heterogeneity by regulating the cotransmitter release characteristics of individual presynaptic terminals remain poorly defined. In particular, we know little about the regulation of neuropeptide corelease, despite the fact that they mediate synaptic transmission, plasticity and neuromodulation. Here, we report that an identified Lymnaea neuron selectively releases its classical small molecule and peptide neurotransmitters, acetylcholine and FMRFamide-derived neuropeptides, to differentially influence the activity of distinct postsynaptic targets that coordinate cardiorespiratory behaviour. Using a combination of electrophysiological, molecular, and pharmacological approaches, we found that neuropeptide cotransmitter release was regulated by cross-talk between extrinsic neurotrophic factor signaling and target-specific retrograde arachidonic acid signaling, which converged on modulation of glycogen synthase kinase 3. In this context, we identified a novel role for the Lymnaea synaptophysin homologue as a specific and synapse-delimited inhibitory regulator of peptide neurotransmitter release. This study is among the first to define the cellular and molecular mechanisms underlying the differential release of cotransmitter substances from individual presynaptic terminals, which allow for context-dependent tuning and plasticity of the synaptic networks underlying patterned motor behaviour.
Collapse
|
104
|
Molecular Evolution and Characterization of Fish Stathmin Genes. Animals (Basel) 2020; 10:ani10081328. [PMID: 32752168 PMCID: PMC7460142 DOI: 10.3390/ani10081328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Stathmin is a highly conserved microtubule remodeling protein. Here, 175 putative stathmin genes were identified in 27 species of fish. Gene organization, motif distribution, divergence of duplicated genes, functional divergence, synteny relationship, and protein-protein interaction were performed to investigate their evolutionary history. In addition, expression profiles of some stathmins were examined under dimethoate treatment. The results will provide useful references for further functional analyses. Abstract Stathmin is a highly conserved microtubule remodeling protein, involved in many biological processes such as signal transduction, cell proliferation, neurogenesis and so on. However, little evolutional information has been reported about this gene family in fish. In this study, 175 stathmin genes were identified in 27 species of fish. Conserved exon-intron structure and motif distributions were found in each group. Divergence of duplicated genes implied the species’ adaptation to the environment. Functional divergence suggested that the evolution of stathmin is mainly influenced by purifying selection, and some residues may undergo positive selection. Moreover, synteny relationship near the stathmin locus was relatively conserved in some fish. Network analyses also exhibited 74 interactions, implying functional diversity. The expression pattern of some stathmin genes was also investigated under pesticide stress. These will provide useful references for their functional research in the future.
Collapse
|
105
|
Niclass T, Le Guyader G, Beneteau C, Joubert M, Pizzuti A, Giuffrida MG, Bernardini L, Gilbert-Dussardier B, Bilan F, Egloff M. 12q21 deletion syndrome: Narrowing the critical region down to 1.6 Mb including SYT1 and PPP1R12A. Am J Med Genet A 2020; 182:2133-2138. [PMID: 32633079 DOI: 10.1002/ajmg.a.61734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/04/2020] [Accepted: 05/21/2020] [Indexed: 11/12/2022]
Abstract
Deletions in the 12q21 region are rare and non-recurrent CNVs. To date, only 11 patients with deletions in this region have been reported in the literature. These patients most often presented with syndromic intellectual deficiency, ventriculomegaly or hydrocephalus, ectodermal abnormalities, growth retardation and renal and cardiac malformations, suggesting a recognizable microdeletion syndrome. We report three new patients with overlapping deletions of the 12q21 region, including the smallest deletion reported to date and the first case characterized by array CGH during pregnancy. We describe specific clinical findings and shared facial features as developmental delay, ectodermal abnormalities, ventriculomegaly or hydrocephalus, axial hypotonia or spastic diplegia, growth retardation, heart defect, hydronephrosis, ureteral reflux or horseshoe kidney, large thorax or pectus excavatum, syndactyly of 2-3 toes, pterygium coli or excess nuchal skin, large anterior fontanel, low set ears, prominent forehead, short-upturned nose with nostril hypoplasia, microretrognathia and hypertelorism. These new patients and a comprehensive review of the literature allow us to define a minimum critical region spanning 1.6 Mb in 12q21. By screening the critical region using prediction tools, we identified two candidate genes: SYT1and PPP1R12A.
Collapse
Affiliation(s)
- Tanguy Niclass
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Gwenael Le Guyader
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,EA 3808 NEUVACOD, Université de Poitiers, Poitiers, France
| | - Claire Beneteau
- Department of Medical Genetics, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Madeleine Joubert
- Department of Anatomic and Fetal Pathology, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Antonio Pizzuti
- Department of Medical Genetics, Policlinico di Roma, Rome, Italy
| | - Maria Grazia Giuffrida
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, FG, Italy
| | - Laura Bernardini
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cytogenetics Unit, San Giovanni Rotondo, FG, Italy
| | - Brigitte Gilbert-Dussardier
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,EA 3808 NEUVACOD, Université de Poitiers, Poitiers, France
| | - Frederic Bilan
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,EA 3808 NEUVACOD, Université de Poitiers, Poitiers, France
| | - Matthieu Egloff
- Department of Medical Genetics, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Laboratoire de Neurosciences Experimentales et Cliniques, INSERM, Poitiers, France
| |
Collapse
|
106
|
Bonnycastle K, Davenport EC, Cousin MA. Presynaptic dysfunction in neurodevelopmental disorders: Insights from the synaptic vesicle life cycle. J Neurochem 2020; 157:179-207. [PMID: 32378740 DOI: 10.1111/jnc.15035] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/14/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The activity-dependent fusion, retrieval and recycling of synaptic vesicles is essential for the maintenance of neurotransmission. Until relatively recently it was believed that most mutations in genes that were essential for this process would be incompatible with life, because of this fundamental role. However, an ever-expanding number of mutations in this very cohort of genes are being identified in individuals with neurodevelopmental disorders, including autism, intellectual disability and epilepsy. This article will summarize the current state of knowledge linking mutations in presynaptic genes to neurodevelopmental disorders by sequentially covering the various stages of the synaptic vesicle life cycle. It will also discuss how perturbations of specific stages within this recycling process could translate into human disease. Finally, it will also provide perspectives on the potential for future therapy that are targeted to presynaptic function.
Collapse
Affiliation(s)
- Katherine Bonnycastle
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Elizabeth C Davenport
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK.,Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
107
|
Banerjee A, Lee J, Nemcova P, Liu C, Kaeser PS. Synaptotagmin-1 is the Ca 2+ sensor for fast striatal dopamine release. eLife 2020; 9:58359. [PMID: 32490813 PMCID: PMC7319770 DOI: 10.7554/elife.58359] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/02/2020] [Indexed: 11/13/2022] Open
Abstract
Dopamine powerfully controls neural circuits through neuromodulation. In the vertebrate striatum, dopamine adjusts cellular functions to regulate behaviors across broad time scales, but how the dopamine secretory system is built to support fast and slow neuromodulation is not known. Here, we set out to identify Ca2+-triggering mechanisms for dopamine release. We find that synchronous dopamine secretion is abolished in acute brain slices of conditional knockout mice in which Synaptotagmin-1 is removed from dopamine neurons. This indicates that Synaptotagmin-1 is the Ca2+ sensor for fast dopamine release. Remarkably, dopamine release induced by strong depolarization and asynchronous release during stimulus trains are unaffected by Synaptotagmin-1 knockout. Microdialysis further reveals that these modes and action potential-independent release provide significant amounts of extracellular dopamine in vivo. We propose that the molecular machinery for dopamine secretion has evolved to support fast and slow signaling modes, with fast release requiring the Ca2+ sensor Synaptotagmin-1.
Collapse
Affiliation(s)
- Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Jinoh Lee
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Paulina Nemcova
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
108
|
Bekkers JM. Autaptic Cultures: Methods and Applications. Front Synaptic Neurosci 2020; 12:18. [PMID: 32425765 PMCID: PMC7203343 DOI: 10.3389/fnsyn.2020.00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/01/2020] [Indexed: 11/13/2022] Open
Abstract
Neurons typically form daisy chains of synaptic connections with other neurons, but they can also form synapses with themselves. Although such self-synapses, or autapses, are comparatively rare in vivo, they are surprisingly common in dissociated neuronal cultures. At first glance, autapses in culture seem like a mere curiosity. However, by providing a simple model system in which a single recording electrode gives simultaneous access to the pre- and postsynaptic compartments, autaptic cultures have proven to be invaluable in facilitating important and elegant experiments in the area of synaptic neuroscience. Here, I provide detailed protocols for preparing and recording from autaptic cultures (also called micro-island or microdot cultures). Variations on the basic procedure are presented, as well as practical tips for optimizing the outcomes. I also illustrate the utility of autaptic cultures by reviewing the types of experiments that have used them over the past three decades. These examples serve to highlight the power and elegance of this simple model system, and will hopefully inspire new experiments for the interrogation of synaptic function.
Collapse
Affiliation(s)
- John M Bekkers
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
109
|
Khan MM, Regehr WG. Loss of Doc2b does not influence transmission at Purkinje cell to deep nuclei synapses under physiological conditions. eLife 2020; 9:55165. [PMID: 32347796 PMCID: PMC7190354 DOI: 10.7554/elife.55165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/09/2020] [Indexed: 12/26/2022] Open
Abstract
Doc2a and Doc2b are high-affinity calcium-binding proteins that interact with SNARE proteins and phospholipids. Experiments performed on cultured cells indicated that Doc2 proteins promote spontaneous vesicle fusion and asynchronous neurotransmitter release, regulate vesicle priming, mediate augmentation, and regulate transmission during sustained activity. Here, we assess the role of Doc2 proteins in synaptic transmission under physiological conditions at mature synapses made by Purkinje cells onto neurons in the deep cerebellar nuclei (PC to DCN synapses). PCs express Doc2b but not Doc2a. Surprisingly, spontaneous neurotransmitter release, synaptic strength, the time course of evoked release, responses evoked by sustained high-frequency stimulation, and short-term plasticity were normal in Doc2b KO mice. Thus, in stark contrast to numerous functions previously proposed for Doc2, here we find that Doc2b removal does not influence transmission at PC-to-DCN synapses, indicating that conclusions based on studies of Doc2b in cultured cells do not necessarily generalize to mature synapses under physiological conditions.
Collapse
Affiliation(s)
- Mehak M Khan
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, United States
| |
Collapse
|
110
|
Guan Z, Quiñones-Frías MC, Akbergenova Y, Littleton JT. Drosophila Synaptotagmin 7 negatively regulates synaptic vesicle release and replenishment in a dosage-dependent manner. eLife 2020; 9:e55443. [PMID: 32343229 PMCID: PMC7224696 DOI: 10.7554/elife.55443] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/28/2020] [Indexed: 01/03/2023] Open
Abstract
Synchronous neurotransmitter release is triggered by Ca2+ binding to the synaptic vesicle protein Synaptotagmin 1, while asynchronous fusion and short-term facilitation is hypothesized to be mediated by plasma membrane-localized Synaptotagmin 7 (SYT7). We generated mutations in Drosophila Syt7 to determine if it plays a conserved role as the Ca2+ sensor for these processes. Electrophysiology and quantal imaging revealed evoked release was elevated 2-fold. Syt7 mutants also had a larger pool of readily-releasable vesicles, faster recovery following stimulation, and intact facilitation. Syt1/Syt7 double mutants displayed more release than Syt1 mutants alone, indicating SYT7 does not mediate the residual asynchronous release remaining in the absence of SYT1. SYT7 localizes to an internal membrane tubular network within the peri-active zone, but does not enrich at active zones. These findings indicate the two Ca2+ sensor model of SYT1 and SYT7 mediating all phases of neurotransmitter release and facilitation is not applicable at Drosophila synapses.
Collapse
Affiliation(s)
- Zhuo Guan
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Monica C Quiñones-Frías
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
111
|
Park SJ, Park H, Kim MG, Zhang S, Park SE, Kim S, Chung C. Inositol Pyrophosphate Metabolism Regulates Presynaptic Vesicle Cycling at Central Synapses. iScience 2020; 23:101000. [PMID: 32252022 PMCID: PMC7132149 DOI: 10.1016/j.isci.2020.101000] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/17/2020] [Accepted: 03/18/2020] [Indexed: 11/27/2022] Open
Abstract
The coordination of synaptic vesicle exocytosis and endocytosis supports neurotransmitter release from presynaptic terminals. Although inositol pyrophosphates, such as 5-diphosphoinositol pentakisphosphate (5-IP7), are versatile signaling metabolites in many biological events, physiological actions of 5-IP7 on synaptic membrane vesicle trafficking remain unclear. Here, we investigated the role of 5-IP7 in synaptic transmission in hippocampal brain slices from inositol hexakisphosphate kinase 1 (Ip6k1)-knockout mice. We found that presynaptic release probability was significantly increased in Ip6k1-knockout neurons, implying enhanced activity-dependent synaptic vesicle exocytosis. Expression of wild-type but not catalytically inactive IP6K1 in the Ip6k1-knockout hippocampus restored the altered presynaptic release probability. Moreover, Ip6k1-knockout neurons were insensitive to folimycin, a vacuolar ATPase inhibitor, and dynasore, a dynamin inhibitor, suggesting marked impairment in synaptic endocytosis during exocytosis. Our findings collectively establish that IP6K1 and its product, 5-IP7, act as key physiological determinants for inhibition of presynaptic vesicle exocytosis and stimulation of endocytosis at central synapses.
Collapse
Affiliation(s)
- Seung Ju Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea
| | - Min-Gyu Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seungjae Zhang
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea
| | - Seung Eun Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seyun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; KAIST Institute for the BioCentury, KAIST, Daejeon 34141, Korea.
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
112
|
Ruiter M, Kádková A, Scheutzow A, Malsam J, Söllner TH, Sørensen JB. An Electrostatic Energy Barrier for SNARE-Dependent Spontaneous and Evoked Synaptic Transmission. Cell Rep 2020; 26:2340-2352.e5. [PMID: 30811985 DOI: 10.1016/j.celrep.2019.01.103] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/05/2018] [Accepted: 01/28/2019] [Indexed: 12/22/2022] Open
Abstract
Information transfer across CNS synapses depends on the very low basal vesicle fusion rate and the ability to rapidly upregulate that rate upon Ca2+ influx. We show that local electrostatic repulsion participates in creating an energy barrier, which limits spontaneous synaptic transmission. The barrier amplitude is increased by negative charges and decreased by positive charges on the SNARE-complex surface. Strikingly, the effect of charges on the barrier is additive and this extends to evoked transmission, but with a shallower charge dependence. Action potential-driven synaptic release is equivalent to the abrupt addition of ∼35 positive charges to the fusion machine. Within an electrostatic model for triggering, the Ca2+ sensor synaptotagmin-1 contributes ∼18 charges by binding Ca2+, while also modulating the fusion barrier at rest. Thus, the energy barrier for synaptic vesicle fusion has a large electrostatic component, allowing synaptotagmin-1 to act as an electrostatic switch and modulator to trigger vesicle fusion.
Collapse
Affiliation(s)
- Marvin Ruiter
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark
| | - Anna Kádková
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Scheutzow
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Jörg Malsam
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Thomas H Söllner
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | - Jakob B Sørensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, 2200 Copenhagen N, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
113
|
Hempel T, Plattner N, Noé F. Coupling of Conformational Switches in Calcium Sensor Unraveled with Local Markov Models and Transfer Entropy. J Chem Theory Comput 2020; 16:2584-2593. [PMID: 32196329 DOI: 10.1021/acs.jctc.0c00043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Proteins often have multiple switching domains that are coupled to each other and to the binding of ligands in order to realize signaling functions. Here we investigate the C2A domain of Synaptotagmin-1 (Syt-1), a calcium sensor in the neurotransmitter release machinery and a model system for the large family of C2 membrane binding domains. We combine extensive molecular dynamics (MD) simulations with Markov modeling in order to model conformational switching domains, their states, and their dependence on bound calcium ions. Then, we use transfer entropy to characterize how the switching domains are coupled via directed or allosteric mechanisms and give rise to the calcium sensing function of the protein. Our proposed switching mechanism contributes to the understanding of the neurotransmitter release machinery. Furthermore, the methodological approach we develop serves as a template to analyze conformational switching domains and the broad study of their coupling in macromolecular machines.
Collapse
Affiliation(s)
- Tim Hempel
- Department of Mathematics and Computer Science, FU Berlin, Arnimallee 6, 14195 Berlin, Germany.,Department of Physics, FU Berlin, Arnimallee 6, 14195 Berlin, Germany
| | - Nuria Plattner
- Department of Mathematics and Computer Science, FU Berlin, Arnimallee 6, 14195 Berlin, Germany
| | - Frank Noé
- Department of Mathematics and Computer Science, FU Berlin, Arnimallee 6, 14195 Berlin, Germany.,Department of Physics, FU Berlin, Arnimallee 6, 14195 Berlin, Germany.,Department of Chemistry, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
114
|
Wang C, Tu J, Zhang S, Cai B, Liu Z, Hou S, Zhong Q, Hu X, Liu W, Li G, Liu Z, He L, Diao J, Zhu ZJ, Li D, Liu C. Different regions of synaptic vesicle membrane regulate VAMP2 conformation for the SNARE assembly. Nat Commun 2020; 11:1531. [PMID: 32210233 PMCID: PMC7093461 DOI: 10.1038/s41467-020-15270-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 02/25/2020] [Indexed: 01/16/2023] Open
Abstract
Vesicle associated membrane protein 2 (VAMP2/synaptobrevin2), a core SNARE protein residing on synaptic vesicles (SVs), forms helix bundles with syntaxin-1 and SNAP25 for the SNARE assembly. Prior to the SNARE assembly, the structure of VAMP2 is unclear. Here, by using in-cell NMR spectroscopy, we describe the dynamic membrane association of VAMP2 SNARE motif in mammalian cells, and the structural change of VAMP2 upon the change of intracellular lipid environment. We analyze the lipid compositions of the SV membrane by mass-spectrometry-based lipidomic profiling, and further reveal that VAMP2 forms distinctive conformations in different membrane regions. In contrast to the non-raft region, the membrane region of cholesterol-rich lipid raft markedly weakens the membrane association of VAMP2 SNARE motif, which releases the SNARE motif and facilitates the SNARE assembly. Our work reveals the regulation of different membrane regions on VAMP2 structure and sheds light on the spatial regulation of SNARE assembly.
Collapse
Affiliation(s)
- Chuchu Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jia Tu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Bin Cai
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zhenying Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shouqiao Hou
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qinglu Zhong
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Xiao Hu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Wenbin Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian, 116023, China
| | - Zhijun Liu
- National Facility for Protein Science in Shanghai, ZhangJiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200030, China. .,Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| |
Collapse
|
115
|
Sathe G, Mangalaparthi KK, Jain A, Darrow J, Troncoso J, Albert M, Moghekar A, Pandey A. Multiplexed Phosphoproteomic Study of Brain in Patients with Alzheimer's Disease and Age-Matched Cognitively Healthy Controls. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 24:216-227. [PMID: 32182160 DOI: 10.1089/omi.2019.0191] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder caused by neuronal loss that results in cognitive and functional impairment. Formation of neurofibrillary tangles composed of abnormal hyperphosphorylation of tau protein is one of the major pathological hallmarks of AD. Importantly, several neurodegenerative disorders, including AD, are associated with abnormal protein phosphorylation events. However, little is known thus far on global protein phosphorylation changes in AD. We report a phosphoproteomics study examining the frontal gyrus of people with AD and age-matched cognitively normal subjects, using tandem mass tag (TMT) multiplexing technology along with immobilized metal affinity chromatography to enrich phosphopeptides. We identified 4631 phosphopeptides corresponding to 1821 proteins with liquid chromatography-mass spectrometry (MS)/MS analysis on an Orbitrap Fusion Lumos Tribrid mass spectrometer. Of these, 504 phosphopeptides corresponding to 350 proteins were significantly altered in the AD brain: 389 phosphopeptides increased whereas 115 phosphopeptides decreased phosphorylation. We observed significant changes in phosphorylation of known as well as novel molecules. Using targeted parallel reaction monitoring experiments, we validated the phosphorylation of microtubule-associated protein tau and myristoylated alanine-rich protein kinase C substrate (MARCKS) in control and AD (Control = 6, AD = 11) brain samples. In conclusion, our study provides new evidence on alteration of RNA processing and splicing, neurogenesis and neuronal development, and metabotropic glutamate receptor 5 (GRM5) calcium signaling pathways in the AD brain, and it thus offers new insights to accelerate diagnostics and therapeutics innovation in AD.
Collapse
Affiliation(s)
- Gajanan Sathe
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.,Institute of Bioinformatics, Bangalore, India.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | | | - Ankit Jain
- Institute of Bioinformatics, Bangalore, India
| | - Jacqueline Darrow
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Juan Troncoso
- Department of Pathology and Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Akhilesh Pandey
- Center for Molecular Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.,Institute of Bioinformatics, Bangalore, India.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Manipal Academy of Higher Education (MAHE), Manipal, India.,Department of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
116
|
Calcium ions modulate the structure of the intrinsically disordered Nucleobindin-2 protein. Int J Biol Macromol 2020; 154:1091-1104. [PMID: 32184136 DOI: 10.1016/j.ijbiomac.2020.03.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 01/05/2023]
Abstract
Nucleobindin-2 (Nucb2) is a widely expressed multi-domain protein. Nucb2 participates in many physiological processes, i.e. calcium level maintenance, feeding regulation in the hypothalamus, emotion and stress regulation, and many others. To date, this protein has not been structurally characterized. We describe the first comparative structural analysis of two homologs, a Gallus gallus and a Homo sapiens Nucb2. The in silico analysis suggested that apo-Nucb2s contain a mosaic-like structure, consisting of intertwined disordered and ordered regions. Surprisingly, the hydrogen-deuterium exchange mass spectrometry results revealed that Nucb2 is divided into two parts: an N-terminal half with a stable mosaic-like structure and a disordered C-terminal half. However, the presence of Ca2+ induces the formation of a mosaic-like structure in the C-terminal half of the Nucb2s. The Ca2+ also affects the tertiary and quaternary structure of Nucb2s. The presence of Ca2+ leads to an overall compaction of the Nucb2 molecule, resulting in structural change that is propagated along the molecule, which in turn affects the quaternary structure of the protein. Intrinsic disorder, and the mosaic-like Ca2+ dependent structure of Nucb2s, might be seen as the molecular factors responsible for their multifunctionality. Thus, Nucb2s might function as the versatile Ca2+ sensor involved in signal transduction.
Collapse
|
117
|
Magdziarek M, Bolembach AA, Stepien KP, Quade B, Liu X, Rizo J. Re-examining how Munc13-1 facilitates opening of syntaxin-1. Protein Sci 2020; 29:1440-1458. [PMID: 32086964 DOI: 10.1002/pro.3844] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/06/2022]
Abstract
Munc13-1 is crucial for neurotransmitter release and, together with Munc18-1, orchestrates assembly of the neuronal SNARE complex formed by syntaxin-1, SNAP-25, and synaptobrevin. Assembly starts with syntaxin-1 folded into a self-inhibited closed conformation that binds to Munc18-1. Munc13-1 is believed to catalyze the opening of syntaxin-1 to facilitate SNARE complex formation. However, different types of Munc13-1-syntaxin-1 interactions have been reported to underlie this activity, and the critical nature of Munc13-1 for release may arise because of its key role in bridging the vesicle and plasma membranes. To shed light into the mechanism of action of Munc13-1, we have used NMR spectroscopy, SNARE complex assembly experiments, and liposome fusion assays. We show that point mutations in a linker region of syntaxin-1 that forms intrinsic part of the closed conformation strongly impair stimulation of SNARE complex assembly and liposome fusion mediated by Munc13-1 fragments, even though binding of this linker region to Munc13-1 is barely detectable. Conversely, the syntaxin-1 SNARE motif clearly binds to Munc13-1, but a mutation that disrupts this interaction does not affect SNARE complex assembly or liposome fusion. We also show that Munc13-1 cannot be replaced by an artificial tethering factor to mediate liposome fusion. Overall, these results emphasize how very weak interactions can play fundamental roles in promoting conformational transitions and strongly support a model whereby the critical nature of Munc13-1 for neurotransmitter release arises not only from its ability to bridge two membranes but also from an active role in opening syntaxin-1 via interactions with the linker.
Collapse
Affiliation(s)
- Magdalena Magdziarek
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Agnieszka A Bolembach
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Karolina P Stepien
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiaoxia Liu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
118
|
Bowers MR, Reist NE. Synaptotagmin: Mechanisms of an electrostatic switch. Neurosci Lett 2020; 722:134834. [DOI: 10.1016/j.neulet.2020.134834] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 02/09/2023]
|
119
|
Phatarpekar PV, Billadeau DD. Molecular regulation of the plasma membrane-proximal cellular steps involved in NK cell cytolytic function. J Cell Sci 2020; 133:133/5/jcs240424. [PMID: 32086255 DOI: 10.1242/jcs.240424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells, cytolytic lymphocytes of the innate immune system, play a crucial role in the immune response against infection and cancer. NK cells kill target cells through exocytosis of lytic granules that contain cytotoxic proteins, such as perforin and granzymes. Formation of a functional immune synapse, i.e. the interface between the NK cell and its target cell enhances lysis through accumulation of polymerized F-actin at the NK cell synapse, leading to convergence of lytic granules to the microtubule organizing center (MTOC) and its subsequent polarization along microtubules to deliver the lytic granules to the synapse. In this review, we focus on the molecular mechanisms regulating the cellular processes that occur after the lytic granules are delivered to the cytotoxic synapse. We outline how - once near the synapse - the granules traverse the clearings created by F-actin remodeling to dock, tether and fuse with the plasma membrane in order to secrete their lytic content into the synaptic cleft through exocytosis. Further emphasis is given to the role of Ca2+ mobilization during degranulation and, whenever applicable, we compare these mechanisms in NK cells and cytotoxic T lymphocytes (CTLs) as adaptive immune system effectors.
Collapse
Affiliation(s)
- Prasad V Phatarpekar
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
120
|
Pan YZ, Liu X, Rizo J. Analysis of asymmetry in lipid and content mixing assays with reconstituted proteoliposomes containing the neuronal SNAREs. Sci Rep 2020; 10:2907. [PMID: 32076023 PMCID: PMC7031292 DOI: 10.1038/s41598-020-59740-7] [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: 12/02/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
Reconstitution assays with proteoliposomes provide a powerful tool to elucidate the mechanism of neurotransmitter release, but it is important to understand how these assays report on membrane fusion, and recent studies with yeast vacuolar SNAREs uncovered asymmetry in the results of lipid mixing assays. We have investigated whether such asymmetry also occurs in reconstitution assays with the neuronal SNAREs, using syntaxin-1-SNAP-25-containing liposomes and liposomes containing synaptobrevin (T and V liposomes, respectively), and fluorescent probes to monitor lipid and content mixing simultaneously. Switching the fluorescent probes placed on the T and V liposomes, we observed a striking asymmetry in both lipid and content mixing stimulated by a fragment spanning the two C2 domains of synaptotagmin-1, or by a peptide that spans the C-terminal half of the synaptobrevin SNARE motif. However, no such asymmetry was observed in assays performed in the presence of Munc18-1, Munc13-1, NSF and αSNAP, which coordinate the assembly-disassembly cycle of neuronal SNARE complexes. Our results show that switching fluorescent probes between the two types of liposomes provides a useful approach to better understand the reactions that occur between liposomes and detect heterogenous behavior in these reactions.
Collapse
Affiliation(s)
- Yun-Zu Pan
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, United States. .,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, United States. .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States.
| |
Collapse
|
121
|
Katti S, Igumenova TI. Interference of pH buffer with Pb 2+-peripheral domain interactions: obstacle or opportunity? Metallomics 2020; 12:164-172. [PMID: 32051983 DOI: 10.1039/d0mt00002g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Pb2+ is a xenobiotic metal ion that competes for Ca2+-binding sites in proteins. Using the peripheral Ca2+-sensing domains of Syt1, we show that the chelating pH buffer Bis-Tris enables identification and functional characterization of high-affinity Pb2+ sites that are likely to be targeted by bioavailable Pb2+.
Collapse
Affiliation(s)
- Sachin Katti
- Department of Biochemistry and Biophysics, Texas A&M University, 300 Olsen Boulevard, College Station, TX 77843, USA.
| | | |
Collapse
|
122
|
Bowers MR, Reist NE. The C2A domain of synaptotagmin is an essential component of the calcium sensor for synaptic transmission. PLoS One 2020; 15:e0228348. [PMID: 32032373 PMCID: PMC7006929 DOI: 10.1371/journal.pone.0228348] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/13/2020] [Indexed: 01/27/2023] Open
Abstract
The synaptic vesicle protein, synaptotagmin, is the principle Ca2+ sensor for synaptic transmission. Ca2+ influx into active nerve terminals is translated into neurotransmitter release by Ca2+ binding to synaptotagmin’s tandem C2 domains, triggering the fast, synchronous fusion of multiple synaptic vesicles. Two hydrophobic residues, shown to mediate Ca2+-dependent membrane insertion of these C2 domains, are required for this process. Previous research suggested that one of its tandem C2 domains (C2B) is critical for fusion, while the other domain (C2A) plays only a facilitatory role. However, the function of the two hydrophobic residues in C2A have not been adequately tested in vivo. Here we show that these two hydrophobic residues are absolutely required for synaptotagmin to trigger vesicle fusion. Using in vivo electrophysiological recording at the Drosophila larval neuromuscular junction, we found that mutation of these two key C2A hydrophobic residues almost completely abolished neurotransmitter release. Significantly, mutation of both hydrophobic residues resulted in more severe deficits than those seen in synaptotagmin null mutants. Thus, we report the most severe phenotype of a C2A mutation to date, demonstrating that the C2A domain is absolutely essential for synaptotagmin’s function as the electrostatic switch. The postulated role of synaptotagmin’s C2A domain in triggering neurotransmitter release has fluctuated wildly over the years. Early biochemical experiments suggested that the C2A domain was essential, while the C2B domain was superfluous. Then, functional experiments measuring neurotransmitter release in vivo following disruptions in Ca2+ binding suggested that C2B was essential, while C2A was superfluous. Subsequently, the use of more refined mutations to disrupt Ca2+ binding indicated that C2A played a facilitatory role. Here we show two hydrophobic residues of the C2A domain are absolutely required for synaptotagmin-triggered neurotransmitter release. Thus, after over twenty years of research, we now demonstrate that the C2A domain of synaptotagmin is an essential component of the Ca2+ sensor for triggering synaptic transmission in vivo.
Collapse
Affiliation(s)
- Matthew R. Bowers
- Department of Biomedical Sciences, Molecular, Cellular, Integrative Neurosciences Program, Colorado State University, Fort Collins, CO, United States of America
| | - Noreen E. Reist
- Department of Biomedical Sciences, Molecular, Cellular, Integrative Neurosciences Program, Colorado State University, Fort Collins, CO, United States of America
- * E-mail:
| |
Collapse
|
123
|
Deficiency of SCAMP5 leads to pediatric epilepsy and dysregulation of neurotransmitter release in the brain. Hum Genet 2020; 139:545-555. [PMID: 32020363 DOI: 10.1007/s00439-020-02123-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/28/2020] [Indexed: 10/25/2022]
Abstract
Secretory carrier membrane proteins (SCAMPs) play an important role in exocytosis in animals, but the precise function of SCAMPs in human disease is unknown. In this study, we identified a homozygous mutation, SCAMP5 R91W, in a Chinese consanguineous family with pediatric epilepsy and juvenile Parkinson's disease. Scamp5 R91W mutant knock-in mice showed typical early-onset epilepsy similar to that in humans. Single-neuron electrophysiological recordings showed that the R91W mutation significantly increased the frequency of miniature excitatory postsynaptic currents (mEPSCs) at a resting state and also increased the amplitude of evoked EPSCs. The R91W mutation affected the interaction between SCAMP5 and synaptotagmin 1 and may affect the function of the SNARE complex, the machinery required for vesicular trafficking and neurotransmitter release. Our work shows that dysfunction of SCAMP5 shifted the excitation/inhibition balance of the neuronal network in the brain, and the deficiency of SCAMP5 leads to pediatric epilepsy.
Collapse
|
124
|
Katti S, Nyenhuis SB, Her B, Cafiso DS, Igumenova TI. Partial Metal Ion Saturation of C2 Domains Primes Synaptotagmin 1-Membrane Interactions. Biophys J 2020; 118:1409-1423. [PMID: 32075747 DOI: 10.1016/j.bpj.2020.01.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/15/2020] [Accepted: 01/27/2020] [Indexed: 12/29/2022] Open
Abstract
Synaptotagmin 1 (Syt1) is an integral membrane protein whose phospholipid-binding tandem C2 domains, C2A and C2B, act as Ca2+ sensors of neurotransmitter release. Our objective was to understand the role of individual metal-ion binding sites of these domains in the membrane association process. We used Pb2+, a structural and functional surrogate of Ca2+, to generate the protein states with well-defined protein-metal ion stoichiometry. NMR experiments revealed that binding of one divalent metal ion per C2 domain results in loss of conformational plasticity of the loop regions, potentially pre-organizing them for additional metal-ion and membrane-binding events. In C2A, a divalent metal ion in site 1 is sufficient to drive its weak association with phosphatidylserine-containing membranes, whereas in C2B, it enhances the interactions with the signaling lipid phosphatidylinositol-4,5-bisphosphate. In full-length Syt1, both Pb2+-complexed C2 domains associate with phosphatidylserine-containing membranes. Electron paramagnetic resonance experiments show that the extent of membrane insertion correlates with the occupancy of the C2 metal ion sites. Together, our results indicate that upon partial metal ion saturation of the intra-loop region, Syt1 adopts a dynamic, partially membrane-bound state. The properties of this state, such as conformationally restricted loop regions and positioning of C2 domains in close proximity to anionic lipid headgroups, "prime" Syt1 for cooperative binding of a full complement of metal ions and deeper membrane insertion.
Collapse
Affiliation(s)
- Sachin Katti
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Sarah B Nyenhuis
- Department of Chemistry and Biophysics Program, University of Virginia, Charlottesville, Virginia
| | - Bin Her
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - David S Cafiso
- Department of Chemistry and Biophysics Program, University of Virginia, Charlottesville, Virginia
| | - Tatyana I Igumenova
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas.
| |
Collapse
|
125
|
Synaptotagmin 1 Is Involved in Neuropathic Pain and Electroacupuncture-Mediated Analgesic Effect. Int J Mol Sci 2020; 21:ijms21030968. [PMID: 32024024 PMCID: PMC7037106 DOI: 10.3390/ijms21030968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 01/26/2020] [Accepted: 01/29/2020] [Indexed: 01/09/2023] Open
Abstract
Numerous studies have verified that electroacupuncture (EA) can relieve neuropathic pain through a variety of mechanisms. Synaptotagmin 1 (Syt-1), a synaptic vesicle protein for regulating exocytosis of neurotransmitters, was found to be affected by EA stimulation. However, the roles of Syt-1 in neuropathic pain and EA-induced analgesic effect remain unclear. Here, the effect of Syt-1 on nociception was assessed through an antibody blockade, siRNA silencing, and lentivirus-mediated overexpression of spinal Syt-1 in rats with spared nerve injury (SNI). EA was used for stimulating bilateral "Sanjinjiao" and "Zusanli" acupoints of the SNI rats to evaluate its effect on nociceptive thresholds and spinal Syt-1 expression. The mechanically and thermally nociceptive behaviors were assessed with paw withdrawal threshold (PWT) and paw withdrawal latency (PWL) at different temperatures, respectively, at day 0, 7, 8, 14, and 20. Syt-1 mRNA and protein levels were determined with qRT-PCR and Western blot, respectively, and its distribution was observed with the immunohistochemistry method. The results demonstrated Syt-1 antibody blockade and siRNA silencing increased ipsilateral PWTs and PWLs of SNI rats, while Syt-1 overexpression decreased ipsilateral PWTs and PWLs of rats. EA significantly attenuated nociceptive behaviors and down-regulated spinal Syt-1 protein levels (especially in laminae I-II), which were reversed by Syt-1 overexpression. Our findings firstly indicate that Syt-1 is involved in the development of neuropathic pain and that EA attenuates neuropathic pain, probably through suppressing Syt-1 protein expression in the spinal cord.
Collapse
|
126
|
Mes D, Palstra AP, Henkel CV, Mayer I, Vindas MA. Swimming exercise enhances brain plasticity in fish. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191640. [PMID: 32218982 PMCID: PMC7029906 DOI: 10.1098/rsos.191640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/04/2019] [Indexed: 06/10/2023]
Abstract
It is well-established that sustained exercise training can enhance brain plasticity and boost cognitive performance in mammals, but this phenomenon has not received much attention in fish. The aim of this study was to determine whether sustained swimming exercise can enhance brain plasticity in juvenile Atlantic salmon. Brain plasticity was assessed by both mapping the whole telencephalon transcriptome and conducting telencephalic region-specific microdissections on target genes. We found that 1772 transcripts were differentially expressed between the exercise and control groups. Gene ontology (GO) analysis identified 195 and 272 GO categories with a significant overrepresentation of up- or downregulated transcripts, respectively. A multitude of these GO categories was associated with neuronal excitability, neuronal signalling, cell proliferation and neurite outgrowth (i.e. cognition-related neuronal markers). Additionally, we found an increase in proliferating cell nuclear antigen (pcna) after both three and eight weeks of exercise in the equivalent to the hippocampus in fish. Furthermore, the expression of the neural plasticity markers synaptotagmin (syt) and brain-derived neurotrophic factor (bdnf) were also increased due to exercise in the equivalent to the lateral septum in fish. In conclusion, this is the first time that swimming exercise has been directly linked to increased telencephalic neurogenesis and neural plasticity in a teleost, and our results pave the way for future studies on exercise-induced neuroplasticity in fish.
Collapse
Affiliation(s)
- Daan Mes
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Arjan P. Palstra
- Wageningen University and Research Animal Breeding and Genomics, Wageningen Livestock Research, Wageningen, The Netherlands
| | - Christiaan V. Henkel
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Ian Mayer
- Department of Production Animal Clinical Sciences, Norwegian University of Life Sciences, Oslo, Norway
| | - Marco A. Vindas
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences, Oslo, Norway
- Uni Environment, Uni Research AS, Bergen, Norway
| |
Collapse
|
127
|
Synaptotagmin-1 and Doc2b Exhibit Distinct Membrane-Remodeling Mechanisms. Biophys J 2019; 118:643-656. [PMID: 31952804 DOI: 10.1016/j.bpj.2019.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Synaptotagmin-1 (Syt1) is a calcium sensor protein that is critical for neurotransmission and is therefore extensively studied. Here, we use pairs of optically trapped beads coated with SNARE-free synthetic membranes to investigate Syt1-induced membrane remodeling. This activity is compared with that of Doc2b, which contains a conserved C2AB domain and induces membrane tethering and hemifusion in this cell-free model. We find that the soluble C2AB domain of Syt1 strongly affects the probability and strength of membrane-membrane interactions in a strictly Ca2+- and protein-dependent manner. Single-membrane loading of Syt1 yielded the highest probability and force of membrane interactions, whereas in contrast, Doc2b was more effective after loading both membranes. A lipid-mixing assay with confocal imaging reveals that both Syt1 and Doc2b are able to induce hemifusion; however, significantly higher Syt1 concentrations are required. Consistently, both C2AB fragments cause a reduction in the membrane-bending modulus, as measured by a method based on atomic force microscopy. This lowering of the energy required for membrane deformation may contribute to Ca2+-induced fusion.
Collapse
|
128
|
Abstract
Synaptotagmin 1 (Syt1) is an abundant and important presynaptic vesicle protein that binds Ca2+ for the regulation of synaptic vesicle exocytosis. Our previous study reported its localization and function on spindle assembly in mouse oocyte meiotic maturation. The present study was designed to investigate the function of Syt1 during mouse oocyte activation and subsequent cortical granule exocytosis (CGE) using confocal microscopy, morpholinol-based knockdown and time-lapse live cell imaging. By employing live cell imaging, we first studied the dynamic process of CGE and calculated the time interval between [Ca2+]i rise and CGE after oocyte activation. We further showed that Syt1 was co-localized to cortical granules (CGs) at the oocyte cortex. After oocyte activation with SrCl2, the Syt1 distribution pattern was altered significantly, similar to the changes seen for the CGs. Knockdown of Syt1 inhibited [Ca2+]i oscillations, disrupted the F-actin distribution pattern and delayed the time of cortical reaction. In summary, as a synaptic vesicle protein and calcium sensor for exocytosis, Syt1 acts as an essential regulator in mouse oocyte activation events including the generation of Ca2+ signals and CGE.
Collapse
|
129
|
Li Q, Zhang S, Hu M, Xu M, Jiang X. Silencing of synaptotagmin 13 inhibits tumor growth through suppressing proliferation and promoting apoptosis of colorectal cancer cells. Int J Mol Med 2019; 45:234-244. [PMID: 31939613 PMCID: PMC6889939 DOI: 10.3892/ijmm.2019.4412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
The treatment of colorectal cancer is currently hampered by the lack of early detection technology. The identification of molecular biomarkers for colorectal cancer is crucial for improving prognosis. Synaptotagmin (SYT) 13 has been reported to be associated with several human tumors, but its role in colorectal cancer remains elusive. In the present study, immunohistochemistry was utilized to detect the expression of SYT13 in colorectal cancer tissues and cells. MTT, colony formation, wound healing and Transwell assays were conducted to evaluate the effect of SYT13 knockdown on the biological behavior of RKO and HCT116 cells. Cell apoptosis and cell cycle profiles were detected by FACS. A mouse xenograft model was constructed to investigate the effect of SYT13 on colorectal cancer in vivo. The results indicated that SYT13 was upregulated in colorectal tumor tissues compared with paracancerous tissues. Silencing of SYT13 inhibited the proliferation, colony formation, migration and invasion ability of RKO and HCT116 cells. Moreover, SYT13 knockdown arrested the cell cycle in the G2 phase, thus inducing cell apoptosis. The in vivo experiments also demonstrated the inhibitory effect of SYT13 on tumor growth. In conclusion, the present study demonstrated that SYT13 may act as a promoter in the development and progression of colorectal cancer and, therefore, may be of value as a target for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Qin Li
- Department of Gastroenterology, Shanghai East Hospital, Tongji University, Shanghai 200123, P.R. China
| | - Shun Zhang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai 200123, P.R. China
| | - Miao Hu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University, Shanghai 200123, P.R. China
| | - Ming Xu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University, Shanghai 200123, P.R. China
| | - Xiaohua Jiang
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University, Shanghai 200123, P.R. China
| |
Collapse
|
130
|
Herbison R, Evans S, Doherty JF, Algie M, Kleffmann T, Poulin R. A molecular war: convergent and ontogenetic evidence for adaptive host manipulation in related parasites infecting divergent hosts. Proc Biol Sci 2019; 286:20191827. [PMID: 31744433 DOI: 10.1098/rspb.2019.1827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mermithids (phylum Nematoda) and hairworms (phylum Nematomorpha) somehow drive their arthropod hosts into water, which is essential for the worms' survival after egression. The mechanisms behind this behavioural change have been investigated in hairworms, but not in mermithids. Establishing a similar mechanistic basis for host behavioural change between these two distantly related parasitic groups would provide strong convergent evidence for adaptive manipulation and insight into how these parasites modify and/or create behaviour. Here, we search for this convergence, and also contrast changes in physiology between hosts infected with immature and mature mermithids to provide the first ontogenetic evidence for adaptive manipulation by disentangling host response and pathology from the parasite's apparent manipulative effects. We used SWATH-mass spectrometry on brains of Forficula auricularia (earwig) and Bellorchestia quoyana (sandhopper), infected with the mermithids Mermis nigrescens and Thaumamermis zealandica, respectively, at both immature and mature stages of infection, to quantify proteomic changes resulting from mermithid infection. Across both hosts (and hairworm-infected hosts, from earlier studies), the general function of dysregulated proteins was conserved. Proteins involved in energy generation/mobilization were dysregulated, corroborating reports of erratic/hyperactive behaviour in infected hosts. Dysregulated proteins involved in axon/dendrite and synapse modulation were also common to all hosts, suggesting neuronal manipulation is involved in inducing positive hydrotaxis. Furthermore, downregulation of CamKII and associated proteins suggest manipulation of memory also contributes to the behavioural shift.
Collapse
Affiliation(s)
- Ryan Herbison
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Steven Evans
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | | | - Michael Algie
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Torsten Kleffmann
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Robert Poulin
- Department of Zoology, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
131
|
Endocytic Adaptor Proteins in Health and Disease: Lessons from Model Organisms and Human Mutations. Cells 2019; 8:cells8111345. [PMID: 31671891 PMCID: PMC6912373 DOI: 10.3390/cells8111345] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Cells need to exchange material and information with their environment. This is largely achieved via cell-surface receptors which mediate processes ranging from nutrient uptake to signaling responses. Consequently, their surface levels have to be dynamically controlled. Endocytosis constitutes a powerful mechanism to regulate the surface proteome and to recycle vesicular transmembrane proteins that strand at the plasma membrane after exocytosis. For efficient internalization, the cargo proteins need to be linked to the endocytic machinery via adaptor proteins such as the heterotetrameric endocytic adaptor complex AP-2 and a variety of mostly monomeric endocytic adaptors. In line with the importance of endocytosis for nutrient uptake, cell signaling and neurotransmission, animal models and human mutations have revealed that defects in these adaptors are associated with several diseases ranging from metabolic disorders to encephalopathies. This review will discuss the physiological functions of the so far known adaptor proteins and will provide a comprehensive overview of their links to human diseases.
Collapse
|
132
|
Abstract
Ca2+ binding proteins (CBP) are of key importance for calcium to play its role as a pivotal second messenger. CBP bind Ca2+ in specific domains, contributing to the regulation of its concentration at the cytosol and intracellular stores. They also participate in numerous cellular functions by acting as Ca2+ transporters across cell membranes or as Ca2+-modulated sensors, i.e. decoding Ca2+ signals. Since CBP are integral to normal physiological processes, possible roles for them in a variety of diseases has attracted growing interest in recent years. In addition, research on CBP has been reinforced with advances in the structural characterization of new CBP family members. In this chapter we have updated a previous review on CBP, covering in more depth potential participation in physiopathological processes and candidacy for pharmacological targets in many diseases. We review intracellular CBP that contain the structural EF-hand domain: parvalbumin, calmodulin, S100 proteins, calcineurin and neuronal Ca2+ sensor proteins (NCS). We also address intracellular CBP lacking the EF-hand domain: annexins, CBP within intracellular Ca2+ stores (paying special attention to calreticulin and calsequestrin), proteins that contain a C2 domain (such as protein kinase C (PKC) or synaptotagmin) and other proteins of interest, such as regucalcin or proprotein convertase subtisilin kexins (PCSK). Finally, we summarise the latest findings on extracellular CBP, classified according to their Ca2+ binding structures: (i) EF-hand domains; (ii) EGF-like domains; (iii) ɣ-carboxyl glutamic acid (GLA)-rich domains; (iv) cadherin domains; (v) Ca2+-dependent (C)-type lectin-like domains; (vi) Ca2+-binding pockets of family C G-protein-coupled receptors.
Collapse
|
133
|
Kou R, Kobayashi Y, Inoue S, Tsuchizawa T, Ueno Y, Suzuki S, Hibino H, Yamamoto T, Nakajima H, Yamada K. Dopamine detection on activated reaction field consisting of graphene-integrated silicon photonic cavity. OPTICS EXPRESS 2019; 27:32058-32068. [PMID: 31684425 DOI: 10.1364/oe.27.032058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/07/2019] [Indexed: 06/10/2023]
Abstract
Graphene is widely recognized as an outstanding and multi-functional material in various application fields such as electronics, photonics, mechanics, and life sciences. We propose a neurotransmitter sensor with ultra-small volume for detecting the photonic light-matter response. Such detection can be achieved using surface-activated monolayer graphene sheets and CMOS-compatible silicon-photonic circuits. Patterned pieces of CVD-grown graphene are integrated on the top of a silicon micro-ring resonator, which induce the adsorption of catecholamine molecules originated from the π-stacking effect. We used dopamine to demonstrate such detection and examine the sensitivity of graphene-dopamine coupling. To avoid high optical insertion loss and degradation of resonance characteristics caused by a graphene's extremely high optical absorption coefficient in the near infrared region, a ring resonator with adjusted coupling design is used to compensate for the drawbacks. Owing to the advanced nano-sensing platform and measurement system, an activated graphene-sensing surface of only ∼30 µm2/ch enables π coupling to dopamine with enough sensitivity to detect less than 10-µM solution concentration. The detection mechanism through the surface reaction is also verified by optical simulation and atomic force microscopy measurement, revealing that the flowing dopamine molecules can only occupy the outermost surface of graphene. We expect this sensor to contribute to the development of an innovative label-free and disposable bio-sensing platform with accurate, sensitive, and fast response.
Collapse
|
134
|
Varga K, Jiang ZJ, Gong LW. Phosphatidylserine is critical for vesicle fission during clathrin-mediated endocytosis. J Neurochem 2019; 152:48-60. [PMID: 31587282 DOI: 10.1111/jnc.14886] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/25/2019] [Accepted: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Phosphatidylserine (PS), a negatively charged phospholipid present predominantly at the inner leaflet of the plasma membrane, has been widely implicated in many cellular processes including membrane trafficking. Along this line, PS has been demonstrated to be important for endocytosis, however, the involved mechanisms remain uncertain. By monitoring clathrin-mediated endocytosis (CME) of single vesicles in mouse chromaffin cells using cell-attached capacitance measurements that offer millisecond time resolution, we demonstrate in the present study that the fission-pore duration is reduced by PS addition, indicating a stimulatory role of PS in regulating the dynamics of vesicle fission during CME. Furthermore, our results show that the PS-mediated effect on the fission-pore duration is Ca2+ -dependent and abolished in the absence of synaptotagmin 1 (Syt1), implying that Syt1 is necessary for the stimulatory role of PS in vesicle fission during CME. Consistently, a Syt1 mutant with a defective PS-Syt1 interaction increases the fission-pore duration. Taken together, our study suggests that PS-Syt1 interaction may be critical in regulating fission dynamics during CME.
Collapse
Affiliation(s)
- Kelly Varga
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Biological Sciences, University of North Texas at Dallas, Dallas, Texas, USA
| | - Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
135
|
Zhang L, Fan B, Zheng Y, Lou Y, Cui Y, Wang K, Zhang T, Tan X. Identification SYT13 as a novel biomarker in lung adenocarcinoma. J Cell Biochem 2019; 121:963-973. [PMID: 31625195 DOI: 10.1002/jcb.29224] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Liyan Zhang
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| | - Bijun Fan
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| | - Yu Zheng
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| | - Yueyan Lou
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| | - Yongqi Cui
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| | - Ke Wang
- Key Lab of Reproduction Regulation of NPFPC, Shanghai Institute of Planned Parenthood Research (SIPPR) Fudan University Reproduction and Development Institution Shanghai China
| | - Tiancheng Zhang
- Key Lab of Reproduction Regulation of NPFPC, Shanghai Institute of Planned Parenthood Research (SIPPR) Fudan University Reproduction and Development Institution Shanghai China
| | - Xiaoming Tan
- Department of Respiratory Medicine, South Campus, Renji Hospital, School of Medicine Shanghai Jiaotong University Shanghai China
| |
Collapse
|
136
|
Deák F, Anderson RE, Fessler JL, Sherry DM. Novel Cellular Functions of Very Long Chain-Fatty Acids: Insight From ELOVL4 Mutations. Front Cell Neurosci 2019; 13:428. [PMID: 31616255 PMCID: PMC6763723 DOI: 10.3389/fncel.2019.00428] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 09/06/2019] [Indexed: 12/22/2022] Open
Abstract
Elongation of Very Long chain fatty acids-4 (ELOVL4) protein is a member of the ELOVL family of fatty acid elongases that is collectively responsible for catalyzing formation of long chain fatty acids. ELOVL4 is the only family member that catalyzes production of Very Long Chain Saturated Fatty Acids (VLC-SFA) and Very Long Chain Polyunsaturated Fatty Acids (VLC-PUFA) with chain lengths ≥28 carbons. ELOVL4 and its VLC-SFA and VLC-PUFA products are emerging as important regulators of synaptic signaling and neuronal survival in the central nervous system (CNS). Distinct sets of mutations in ELOVL4 cause three different neurological diseases in humans. Heterozygous inheritance of one set of autosomal dominant ELOVL4 mutations that leads to truncation of the ELOVL4 protein causes Stargardt-like macular dystrophy (STGD3), an aggressive juvenile-onset retinal degeneration. Heterozygous inheritance of a different set of autosomal dominant ELOVL4 mutations that leads to a full-length protein with single amino acid substitutions causes spinocerebellar ataxia 34 (SCA34), a late-onset neurodegenerative disease characterized by gait ataxia and cerebellar atrophy. Homozygous inheritance of a different set of ELOVL4 mutations causes a more severe disease with infantile onset characterized by seizures, spasticity, intellectual disability, ichthyosis, and premature death. ELOVL4 is expressed widely in the CNS and is found primarily in neurons. ELOVL4 is expressed in cell-specific patterns within different regions of the CNS that are likely to be related to disease symptoms. In the retina, ELOVL4 is expressed exclusively in photoreceptors and produces VLC-PUFA that are incorporated into phosphatidylcholine and enriched in the light sensitive membrane disks of the photoreceptor outer segments. VLC-PUFA are enzymatically converted into "elovanoid" compounds that appear to provide paracrine signals that promote photoreceptor and neuronal survival. In the brain, the main ELOVL4 products are VLC-SFA that are incorporated into sphingolipids and enriched in synaptic vesicles, where they regulate kinetics of presynaptic neurotransmitter release. Understanding the function of ELOVL4 and its VLC-SFA and VLC-PUFA products will advance our understanding of basic mechanisms in neural signaling and has potential for developing novel therapies for seizure and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ferenc Deák
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Robert E Anderson
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jennifer L Fessler
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - David M Sherry
- Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
137
|
Burgoyne RD, Helassa N, McCue HV, Haynes LP. Calcium Sensors in Neuronal Function and Dysfunction. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a035154. [PMID: 30833454 DOI: 10.1101/cshperspect.a035154] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Calcium signaling in neurons as in other cell types can lead to varied changes in cellular function. Neuronal Ca2+ signaling processes have also become adapted to modulate the function of specific pathways over a wide variety of time domains and these can have effects on, for example, axon outgrowth, neuronal survival, and changes in synaptic strength. Ca2+ also plays a key role in synapses as the trigger for fast neurotransmitter release. Given its physiological importance, abnormalities in neuronal Ca2+ signaling potentially underlie many different neurological and neurodegenerative diseases. The mechanisms by which changes in intracellular Ca2+ concentration in neurons can bring about diverse responses is underpinned by the roles of ubiquitous or specialized neuronal Ca2+ sensors. It has been established that synaptotagmins have key functions in neurotransmitter release, and, in addition to calmodulin, other families of EF-hand-containing neuronal Ca2+ sensors, including the neuronal calcium sensor (NCS) and the calcium-binding protein (CaBP) families, play important physiological roles in neuronal Ca2+ signaling. It has become increasingly apparent that these various Ca2+ sensors may also be crucial for aspects of neuronal dysfunction and disease either indirectly or directly as a direct consequence of genetic variation or mutations. An understanding of the molecular basis for the regulation of the targets of the Ca2+ sensors and the physiological roles of each protein in identified neurons may contribute to future approaches to the development of treatments for a variety of human neuronal disorders.
Collapse
Affiliation(s)
- Robert D Burgoyne
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hannah V McCue
- Centre for Genomic Research, University of Liverpool, Liverpool, United Kingdom
| | - Lee P Haynes
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
138
|
Chakrabarti R, Wichmann C. Nanomachinery Organizing Release at Neuronal and Ribbon Synapses. Int J Mol Sci 2019; 20:E2147. [PMID: 31052288 PMCID: PMC6539712 DOI: 10.3390/ijms20092147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
A critical aim in neuroscience is to obtain a comprehensive view of how regulated neurotransmission is achieved. Our current understanding of synapses relies mainly on data from electrophysiological recordings, imaging, and molecular biology. Based on these methodologies, proteins involved in a synaptic vesicle (SV) formation, mobility, and fusion at the active zone (AZ) membrane have been identified. In the last decade, electron tomography (ET) combined with a rapid freezing immobilization of neuronal samples opened a window for understanding the structural machinery with the highest spatial resolution in situ. ET provides significant insights into the molecular architecture of the AZ and the organelles within the presynaptic nerve terminal. The specialized sensory ribbon synapses exhibit a distinct architecture from neuronal synapses due to the presence of the electron-dense synaptic ribbon. However, both synapse types share the filamentous structures, also commonly termed as tethers that are proposed to contribute to different steps of SV recruitment and exocytosis. In this review, we discuss the emerging views on the role of filamentous structures in SV exocytosis gained from ultrastructural studies of excitatory, mainly central neuronal compared to ribbon-type synapses with a focus on inner hair cell (IHC) ribbon synapses. Moreover, we will speculate on the molecular entities that may be involved in filament formation and hence play a crucial role in the SV cycle.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
- Collaborative Research Center 1286 "Quantitative Synaptology", 37099 Göttingen, Germany.
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
139
|
Brunger AT, Choi UB, Lai Y, Leitz J, White KI, Zhou Q. The pre-synaptic fusion machinery. Curr Opin Struct Biol 2019; 54:179-188. [PMID: 30986753 PMCID: PMC6939388 DOI: 10.1016/j.sbi.2019.03.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 11/26/2022]
Abstract
Here, we review recent insights into the neuronal presynaptic fusion machinery that releases neurotransmitter molecules into the synaptic cleft upon stimulation. The structure of the pre-fusion state of the SNARE/complexin-1/synaptotagmin-1 synaptic protein complex suggests a new model for the initiation of fast Ca2+-triggered membrane fusion. Functional studies have revealed roles of the essential factors Munc18 and Munc13, demonstrating that a part of their function involves the proper assembly of synaptic protein complexes. Near-atomic resolution structures of the NSF/αSNAP/SNARE complex provide first glimpses of the molecular machinery that disassembles the SNARE complex during the synaptic vesicle cycle. These structures show how this machinery captures the SNARE substrate and provide clues as to a possible processing mechanism.
Collapse
Affiliation(s)
- Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA.
| | - Ucheor B Choi
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Ying Lai
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Jeremy Leitz
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Kristopher Ian White
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| | - Qiangjun Zhou
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA; Department of Structural Biology, Stanford University, Stanford, USA; Department of Photon Science, Stanford University, Stanford, USA; Howard Hughes Medical Institute, Stanford University, Stanford, USA
| |
Collapse
|
140
|
Kula B, Chen T, Kukley M. Glutamatergic signaling between neurons and oligodendrocyte lineage cells: Is it synaptic or non‐synaptic? Glia 2019; 67:2071-2091. [DOI: 10.1002/glia.23617] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Bartosz Kula
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Graduate Training Centre for NeuroscienceUniversity of Tübingen Tübingen Germany
| | - Ting‐Jiun Chen
- Center for Neuroscience ResearchChildren's Research Institute, Children's National Medical Center Washington District of Columbia
| | - Maria Kukley
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Research Institute for OphthalmologyUniversity Hospital Tübingen Tübingen Germany
| |
Collapse
|
141
|
Shimojo M, Madara J, Pankow S, Liu X, Yates J, Südhof TC, Maximov A. Synaptotagmin-11 mediates a vesicle trafficking pathway that is essential for development and synaptic plasticity. Genes Dev 2019; 33:365-376. [PMID: 30808661 PMCID: PMC6411015 DOI: 10.1101/gad.320077.118] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/21/2018] [Indexed: 11/25/2022]
Abstract
Synaptotagmin-11 (Syt11) is a Synaptotagmin isoform that lacks an apparent ability to bind calcium, phospholipids, or SNARE proteins. While human genetic studies have linked mutations in the Syt11 gene to schizophrenia and Parkinson's disease, the localization or physiological role of Syt11 remain unclear. We found that in neurons, Syt11 resides on abundant vesicles that differ from synaptic vesicles and resemble trafficking endosomes. These vesicles recycle via the plasma membrane in an activity-dependent manner, but their exocytosis is slow and desynchronized. Constitutive knockout mice lacking Syt11 died shortly after birth, suggesting Syt11-mediated membrane transport is required for survival. In contrast, selective ablation of Syt11 in excitatory forebrain neurons using a conditional knockout did not affect life span but impaired synaptic plasticity and memory. Syt11-deficient neurons displayed normal secretion of fast neurotransmitters and peptides but exhibited a reduction of long-term synaptic potentiation. Hence, Syt11 is an essential component of a neuronal vesicular trafficking pathway that differs from the well-characterized synaptic vesicle trafficking pathway but is also essential for life.
Collapse
Affiliation(s)
- Masafumi Shimojo
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
| | - Joseph Madara
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
| | - Sandra Pankow
- Department of Molecular Medicine, Scripps Research, La Jolla, California 92037, USA
| | - Xinran Liu
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
| | - John Yates
- Department of Molecular Medicine, Scripps Research, La Jolla, California 92037, USA
| | - Thomas C Südhof
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, California 94035, USA
| | - Anton Maximov
- Department of Neuroscience, Scripps Research, La Jolla, California 92037, USA
- The Dorris Neuroscience, Scripps Research, La Jolla, California 92037, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center at Dallas, Dallas 75235, Texas, USA
| |
Collapse
|
142
|
Quade B, Camacho M, Zhao X, Orlando M, Trimbuch T, Xu J, Li W, Nicastro D, Rosenmund C, Rizo J. Membrane bridging by Munc13-1 is crucial for neurotransmitter release. eLife 2019; 8:42806. [PMID: 30816091 PMCID: PMC6407922 DOI: 10.7554/elife.42806] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/22/2019] [Indexed: 11/13/2022] Open
Abstract
Munc13-1 plays a crucial role in neurotransmitter release. We recently proposed that the C-terminal region encompassing the C1, C2B, MUN and C2C domains of Munc13-1 (C1C2BMUNC2C) bridges the synaptic vesicle and plasma membranes through interactions involving the C2C domain and the C1-C2B region. However, the physiological relevance of this model has not been demonstrated. Here we show that C1C2BMUNC2C bridges membranes through opposite ends of its elongated structure. Mutations in putative membrane-binding sites of the C2C domain disrupt the ability of C1C2BMUNC2C to bridge liposomes and to mediate liposome fusion in vitro. These mutations lead to corresponding disruptive effects on synaptic vesicle docking, priming, and Ca2+-triggered neurotransmitter release in mouse neurons. Remarkably, these effects include an almost complete abrogation of release by a single residue substitution in this 200 kDa protein. These results show that bridging the synaptic vesicle and plasma membranes is a central function of Munc13-1.
Collapse
Affiliation(s)
- Bradley Quade
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marcial Camacho
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Xiaowei Zhao
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marta Orlando
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Thorsten Trimbuch
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Junjie Xu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Wei Li
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Daniela Nicastro
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Christian Rosenmund
- Institut für Neurophysiologie, Charité - Universitätsmedizin, Berlin, Germany.,NeuroCure Cluster of Excellence, Berlin, Germany
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
143
|
Batool S, Raza H, Zaidi J, Riaz S, Hasan S, Syed NI. Synapse formation: from cellular and molecular mechanisms to neurodevelopmental and neurodegenerative disorders. J Neurophysiol 2019; 121:1381-1397. [PMID: 30759043 DOI: 10.1152/jn.00833.2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The precise patterns of neuronal assembly during development determine all functional outputs of a nervous system; these may range from simple reflexes to learning, memory, cognition, etc. To understand how brain functions and how best to repair it after injury, disease, or trauma, it is imperative that we first seek to define fundamental steps mediating this neuronal assembly. To acquire the sophisticated ensemble of highly specialized networks seen in a mature brain, all proliferated and migrated neurons must extend their axonal and dendritic processes toward targets, which are often located at some distance. Upon contact with potential partners, neurons must undergo dramatic structural changes to become either a pre- or a postsynaptic neuron. This connectivity is cemented through specialized structures termed synapses. Both structurally and functionally, the newly formed synapses are, however, not static as they undergo consistent changes in order for an animal to meet its behavioral needs in a changing environment. These changes may be either in the form of new synapses or an enhancement of their synaptic efficacy, referred to as synaptic plasticity. Thus, synapse formation is not restricted to neurodevelopment; it is a process that remains active throughout life. As the brain ages, either the lack of neuronal activity or cell death render synapses dysfunctional, thus giving rise to neurodegenerative disorders. This review seeks to highlight salient steps that are involved in a neuron's journey, starting with the establishment, maturation, and consolidation of synapses; we particularly focus on identifying key players involved in the synaptogenic program. We hope that this endeavor will not only help the beginners in this field to understand how brain networks are assembled in the first place but also shed light on various neurodevelopmental, neurological, neurodegenerative, and neuropsychiatric disorders that involve synaptic inactivity or dysfunction.
Collapse
Affiliation(s)
- Shadab Batool
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada.,Department of Neuroscience, University of Calgary, Alberta, Canada
| | - Hussain Raza
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Jawwad Zaidi
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Saba Riaz
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Sean Hasan
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Naweed I Syed
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada.,Department of Cell Biology & Anatomy, University of Calgary, Alberta, Canada
| |
Collapse
|
144
|
Tran HT, Anderson LH, Knight JD. Membrane-Binding Cooperativity and Coinsertion by C2AB Tandem Domains of Synaptotagmins 1 and 7. Biophys J 2019; 116:1025-1036. [PMID: 30795874 DOI: 10.1016/j.bpj.2019.01.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/21/2018] [Accepted: 01/30/2019] [Indexed: 02/04/2023] Open
Abstract
Synaptotagmin-1 (Syt-1) and synaptotagmin-7 (Syt-7) contain analogous tandem C2 domains, C2A and C2B, which together sense Ca2+ to bind membranes and promote the stabilization of exocytotic fusion pores. Syt-1 triggers fast release of neurotransmitters, whereas Syt-7 functions in processes that involve lower Ca2+ concentrations such as hormone secretion. Syt-1 C2 domains are reported to bind membranes cooperatively, based on the observation that they penetrate farther into membranes as the C2AB tandem than as individual C2 domains. In contrast, we previously suggested that the two C2 domains of Syt-7 bind membranes independently, based in part on measurements of their liposome dissociation kinetics. Here, we investigated C2A-C2B interdomain cooperativity with Syt-1 and Syt-7 using directly comparable measurements. Equilibrium Ca2+ titrations demonstrate that the Syt-7 C2AB tandem binds liposomes lacking phosphatidylinositol-4,5-bisphosphate (PIP2) with greater Ca2+ sensitivity than either of its individual domains and binds to membranes containing PIP2 even in the absence of Ca2+. Stopped-flow kinetic measurements show differences in cooperativity between Syt-1 and Syt-7: Syt-1 C2AB dissociates from PIP2-free liposomes much more slowly than either of its individual C2 domains, indicating cooperativity, whereas the major population of Syt-7 C2AB has a dissociation rate comparable to its C2A domain, suggesting a lack of cooperativity. A minor subpopulation of Syt-7 C2AB dissociates at a slower rate, which could be due to a small cooperative component and/or liposome clustering. Measurements using an environment-sensitive fluorescent probe indicate that the Syt-7 C2B domain inserts deeply into membranes as part of the C2AB tandem, similar to the coinsertion previously reported for Syt-1. Overall, coinsertion of C2A and C2B domains is coupled to cooperative energetic effects in Syt-1 to a much greater extent than in Syt-7. The difference can be understood in terms of the relative contributions of C2A and C2B domains toward membrane binding in the two proteins.
Collapse
Affiliation(s)
- Hai T Tran
- Department of Chemistry, University of Colorado Denver, Denver, Colorado
| | - Lauren H Anderson
- Department of Chemistry, University of Colorado Denver, Denver, Colorado
| | - Jefferson D Knight
- Department of Chemistry, University of Colorado Denver, Denver, Colorado.
| |
Collapse
|
145
|
Campbell CL, Saavedra-Rodriguez K, Kubik TD, Lenhart A, Lozano-Fuentes S, Black WC. Vgsc-interacting proteins are genetically associated with pyrethroid resistance in Aedes aegypti. PLoS One 2019; 14:e0211497. [PMID: 30695054 PMCID: PMC6350986 DOI: 10.1371/journal.pone.0211497] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/15/2019] [Indexed: 11/18/2022] Open
Abstract
Association mapping of factors that condition pyrethroid resistance in Aedes aegypti has consistently identified genes in multiple functional groups. Toward better understanding of the mechanisms involved, we examined high throughput sequencing data (HTS) from two Aedes aegypti aegypti collections from Merida, Yucatan, Mexico treated with either permethrin or deltamethrin. Exome capture enrichment for coding regions and the AaegL5 annotation were used to identify genes statistically associated with resistance. The frequencies of single nucleotide polymorphisms (SNPs) were compared between resistant and susceptible mosquito pools using a contingency χ2 analysis. The -log10(χ2p value) was calculated at each SNP site, with a weighted average determined from all sites in each gene. Genes with -log10(χ2p value) ≥ 4.0 and present among all 3 treatment groups were subjected to gene set enrichment analysis (GSEA). We found that several functional groups were enriched compared to all coding genes. These categories were transport, signal transduction and metabolism, in order from highest to lowest statistical significance. Strikingly, 21 genes with demonstrated association to synaptic function were identified. In the high association group (n = 1,053 genes), several genes were identified that also genetically or physically interact with the voltage-gated sodium channel (VGSC). These genes were eg., CHARLATAN (CHL), a transcriptional regulator, several ankyrin-domain proteins, PUMILIO (PUM), a translational repressor, and NEDD4 (E3 ubiquitin-protein ligase). There were 13 genes that ranked among the top 10%: these included VGSC; CINGULIN, a predicted neuronal gap junction protein, and the aedine ortholog of NERVY (NVY), a transcriptional regulator. Silencing of CHL and NVY followed by standard permethrin bottle bioassays validated their association with permethrin resistance. Importantly, VGSC levels were also reduced about 50% in chl- or nvy-dsRNA treated mosquitoes. These results are consistent with the contribution of a variety of neuronal pathways to pyrethroid resistance in Ae. aegypti.
Collapse
Affiliation(s)
- Corey L Campbell
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Karla Saavedra-Rodriguez
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Tristan D Kubik
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Audrey Lenhart
- Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Saul Lozano-Fuentes
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - William C Black
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
146
|
Prinslow EA, Stepien KP, Pan YZ, Xu J, Rizo J. Multiple factors maintain assembled trans-SNARE complexes in the presence of NSF and αSNAP. eLife 2019; 8:38880. [PMID: 30657450 PMCID: PMC6353594 DOI: 10.7554/elife.38880] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 01/17/2019] [Indexed: 11/13/2022] Open
Abstract
Neurotransmitter release requires formation of trans-SNARE complexes between the synaptic vesicle and plasma membranes, which likely underlies synaptic vesicle priming to a release-ready state. It is unknown whether Munc18-1, Munc13-1, complexin-1 and synaptotagmin-1 are important for priming because they mediate trans-SNARE complex assembly and/or because they prevent trans-SNARE complex disassembly by NSF-αSNAP, which can lead to de-priming. Here we show that trans-SNARE complex formation in the presence of NSF-αSNAP requires both Munc18-1 and Munc13-1, as proposed previously, and is facilitated by synaptotagmin-1. Our data also show that Munc18-1, Munc13-1, complexin-1 and likely synaptotagmin-1 contribute to maintaining assembled trans-SNARE complexes in the presence of NSF-αSNAP. We propose a model whereby Munc18-1 and Munc13-1 are critical not only for mediating vesicle priming but also for precluding de-priming by preventing trans-SNARE complex disassembly; in this model, complexin-1 also impairs de-priming, while synaptotagmin-1 may assist in priming and hinder de-priming.
Collapse
Affiliation(s)
- Eric A Prinslow
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Karolina P Stepien
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yun-Zu Pan
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Junjie Xu
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Josep Rizo
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
147
|
Biochemical studies of membrane fusion at the single-particle level. Prog Lipid Res 2019; 73:92-100. [PMID: 30611882 DOI: 10.1016/j.plipres.2019.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 01/21/2023]
Abstract
To study membrane fusion mediated by synaptic proteins, proteoliposomes have been widely used for in vitro ensemble measurements with limited insights into the fusion mechanism. Single-particle techniques have proven to be powerful in overcoming the limitations of traditional ensemble methods. Here, we summarize current single-particle methods in biophysical and biochemical studies of fusion mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and other synaptic proteins, together with their advantages and limitations.
Collapse
|
148
|
Rodrigues MA, Gomes DA, Nathanson MH. Calcium Signaling in Cholangiocytes: Methods, Mechanisms, and Effects. Int J Mol Sci 2018; 19:ijms19123913. [PMID: 30563259 PMCID: PMC6321159 DOI: 10.3390/ijms19123913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Calcium (Ca2+) is a versatile second messenger that regulates a number of cellular processes in virtually every type of cell. The inositol 1,4,5-trisphosphate receptor (ITPR) is the only intracellular Ca2+ release channel in cholangiocytes, and is therefore responsible for Ca2+-mediated processes in these cells. This review will discuss the machinery responsible for Ca2+ signals in these cells, as well as experimental models used to investigate cholangiocyte Ca2+ signaling. We will also discuss the role of Ca2+ in the normal and abnormal regulation of secretion and apoptosis in cholangiocytes, two of the best characterized processes mediated by Ca2+ in this cell type.
Collapse
Affiliation(s)
- Michele Angela Rodrigues
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| | - Dawidson Assis Gomes
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
- Department of Biochemistry and Immunology, Federal University of Minas Gerais. Av. Antônio Carlos, 6627, Belo Horizonte-MG 31270-901, Brazil.
| | - Michael Harris Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| |
Collapse
|
149
|
Madrigal MP, Portalés A, SanJuan MP, Jurado S. Postsynaptic SNARE Proteins: Role in Synaptic Transmission and Plasticity. Neuroscience 2018; 420:12-21. [PMID: 30458218 DOI: 10.1016/j.neuroscience.2018.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 10/03/2018] [Accepted: 11/10/2018] [Indexed: 12/30/2022]
Abstract
Soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) proteins mediate membrane fusion events in eukaryotic cells. Traditionally recognized as major players in regulating presynaptic neurotransmitter release, accumulative evidence over recent years has identified several SNARE proteins implicated in important postsynaptic processes such as neurotransmitter receptor trafficking and synaptic plasticity. Here we analyze the emerging data revealing this novel functional dimension for SNAREs with a focus on the molecular specialization of vesicular recycling and fusion in dendrites compared to those at axon terminals and its impact in synaptic transmission and plasticity.
Collapse
Affiliation(s)
| | - Adrián Portalés
- Instituto de Neurociencias CSIC-UMH, 03550 San Juan de Alicante, Spain
| | | | - Sandra Jurado
- Instituto de Neurociencias CSIC-UMH, 03550 San Juan de Alicante, Spain.
| |
Collapse
|
150
|
McDargh ZA, Polley A, O'Shaughnessy B. SNARE-mediated membrane fusion is a two-stage process driven by entropic forces. FEBS Lett 2018; 592:3504-3515. [PMID: 30346036 DOI: 10.1002/1873-3468.13277] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 11/08/2022]
Abstract
SNARE proteins constitute the core of the exocytotic membrane fusion machinery. Fusion occurs when vesicle-associated and target membrane-associated SNAREs zipper into trans-SNARE complexes ('SNAREpins'), but the number required is controversial and the mechanism of cooperative fusion is poorly understood. We developed a highly coarse-grained molecular dynamics simulation to access the long fusion timescales, which revealed a two-stage process. First, zippering energy was dissipated and cooperative entropic forces assembled the SNAREpins into a ring; second, entropic forces expanded the ring, pressing membranes together and catalyzing fusion. We predict that any number of SNAREs fuses membranes, but fusion is faster with more SNAREs.
Collapse
Affiliation(s)
- Zachary A McDargh
- Department of Chemical Engineering, Columbia University, New York City, NY, USA
| | - Anirban Polley
- Department of Chemical Engineering, Columbia University, New York City, NY, USA
| | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York City, NY, USA
| |
Collapse
|