1
|
Bulumulla C, Krasley AT, Cristofori-Armstrong B, Valinsky WC, Walpita D, Ackerman D, Clapham DE, Beyene AG. Visualizing synaptic dopamine efflux with a 2D composite nanofilm. eLife 2022; 11:78773. [PMID: 35786443 PMCID: PMC9363124 DOI: 10.7554/elife.78773] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/01/2022] [Indexed: 11/23/2022] Open
Abstract
Chemical neurotransmission constitutes one of the fundamental modalities of communication between neurons. Monitoring release of these chemicals has traditionally been difficult to carry out at spatial and temporal scales relevant to neuron function. To understand chemical neurotransmission more fully, we need to improve the spatial and temporal resolutions of measurements for neurotransmitter release. To address this, we engineered a chemi-sensitive, two-dimensional composite nanofilm that facilitates visualization of the release and diffusion of the neurochemical dopamine with synaptic resolution, quantal sensitivity, and simultaneously from hundreds of release sites. Using this technology, we were able to monitor the spatiotemporal dynamics of dopamine release in dendritic processes, a poorly understood phenomenon. We found that dopamine release is broadcast from a subset of dendritic processes as hotspots that have a mean spatial spread of ≈ 3.2 µm (full width at half maximum [FWHM]) and are observed with a mean spatial frequency of one hotspot per ≈ 7.5 µm of dendritic length. Major dendrites of dopamine neurons and fine dendritic processes, as well as dendritic arbors and dendrites with no apparent varicose morphology participated in dopamine release. Remarkably, these release hotspots co-localized with Bassoon, suggesting that Bassoon may contribute to organizing active zones in dendrites, similar to its role in axon terminals. To form the vast and complex network necessary for an organism to sense and react to the world, neurons must connect at highly specialized junctions. Individual cells communicate at these ‘synapses’ by releasing chemical signals (or neurotransmitters) such as dopamine, a molecule involved in learning and motivation. Despite the central role that synapses play in the brain, it remains challenging to measure exactly where neurotransmitters are released and how far they travel from their release site. Currently, most tools available to scientists only allow bulk measurements of neurotransmitter release. To tackle this limitation, Bulumulla et al. developed a new way to measure neurotransmitter release from neurons, harnessing a technique which uses fluorescent nanosensors that glow brighter when exposed to dopamine. These sensors form a very thin film upon which neurons can grow; when the cells release dopamine, the sensors ‘light up’ as they encounter the molecule. Dubbed DopaFilm, the technology reveals exactly where the neurotransmitter comes from and how it spreads between cells in real time. In particular, the approach showed that dopamine emerges from 'hot spots' at specific sites in cells; it also helped Bulumulla et al. study how dopamine is released from subcellular compartments that have previously not been well characterized. Improving the sensors so that the film could detect other neurotransmitters besides dopamine would broaden the use of this approach. In the future, combining this technology with other types of imaging should enable studies of individual synapses with intricate detail.
Collapse
|
2
|
Harper CB, Smillie KJ. Current molecular approaches to investigate pre-synaptic dysfunction. J Neurochem 2021; 157:107-129. [PMID: 33544872 DOI: 10.1111/jnc.15316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/19/2022]
Abstract
Over the course of the last few decades it has become clear that many neurodevelopmental and neurodegenerative disorders have a synaptic defect, which contributes to pathogenicity. A rise in new techniques, and in particular '-omics'-based methods providing large datasets, has led to an increase in potential proteins and pathways implicated in synaptic function and related disorders. Additionally, advancements in imaging techniques have led to the recent discovery of alternative modes of synaptic vesicle recycling. This has resulted in a lack of clarity over the precise role of different pathways in maintaining synaptic function and whether these new pathways are dysfunctional in neurodevelopmental and neurodegenerative disorders. A greater understanding of the molecular detail of pre-synaptic function in health and disease is key to targeting new proteins and pathways for novel treatments and the variety of new techniques currently available provides an ideal opportunity to investigate these functions. This review focuses on techniques to interrogate pre-synaptic function, concentrating mainly on synaptic vesicle recycling. It further examines techniques to determine the underlying molecular mechanism of pre-synaptic dysfunction and discusses methods to identify molecular targets, along with protein-protein interactions and cellular localization. In combination, these techniques will provide an expanding and more complete picture of pre-synaptic function. With the application of recent technological advances, we are able to resolve events with higher spatial and temporal resolution, leading research towards a greater understanding of dysfunction at the presynapse and the role it plays in pathogenicity.
Collapse
Affiliation(s)
- Callista B Harper
- Centre for Discovery Brain Sciences, University of Edinburgh, Scotland, UK
| | - Karen J Smillie
- Centre for Discovery Brain Sciences, University of Edinburgh, Scotland, UK
| |
Collapse
|
3
|
BDNF impact on synaptic dynamics: extra or intracellular long-term release differently regulates cultured hippocampal synapses. Mol Brain 2020; 13:43. [PMID: 32183860 PMCID: PMC7079446 DOI: 10.1186/s13041-020-00582-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/09/2020] [Indexed: 01/21/2023] Open
Abstract
Brain Derived Neurotrophic Factor (BDNF) signalling contributes to the formation, maturation and plasticity of Central Nervous System (CNS) synapses. Acute exposure of cultured brain circuits to BDNF leads to up-regulation of glutamatergic neuro-transmission, by the accurate tuning of pre and post synaptic features, leading to structural and functional synaptic changes. Chronic BDNF treatment has been comparatively less investigated, besides it may represent a therapeutic option to obtain rescue of post-injury alterations of synaptic networks. In this study, we used a paradigm of BDNF long-term (4 days) incubation to assess in hippocampal neurons in culture, the ability of such a treatment to alter synapses. By patch clamp recordings we describe the augmented function of excitatory neurotransmission and we further explore by live imaging the presynaptic changes brought about by long-term BDNF. In our study, exogenous long-term BDNF exposure of post-natal neurons did not affect inhibitory neurotransmission. We further compare, by genetic manipulations of cultured neurons and BDNF release, intracellular overexpression of this neurotrophin at the same developmental age. We describe for the first-time differences in synaptic modulation by BDNF with respect to exogenous or intracellular release paradigms. Such a finding holds the potential of influencing the design of future therapeutic strategies.
Collapse
|
4
|
Coleman J, Jouannot O, Ramakrishnan SK, Zanetti MN, Wang J, Salpietro V, Houlden H, Rothman JE, Krishnakumar SS. PRRT2 Regulates Synaptic Fusion by Directly Modulating SNARE Complex Assembly. Cell Rep 2019; 22:820-831. [PMID: 29346777 PMCID: PMC5792450 DOI: 10.1016/j.celrep.2017.12.056] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 11/12/2017] [Accepted: 12/17/2017] [Indexed: 11/25/2022] Open
Abstract
Mutations in proline-rich transmembrane protein 2 (PRRT2) are associated with a range of paroxysmal neurological disorders. PRRT2 predominantly localizes to the pre-synaptic terminals and is believed to regulate neurotransmitter release. However, the mechanism of action is unclear. Here, we use reconstituted single vesicle and bulk fusion assays, combined with live cell imaging of single exocytotic events in PC12 cells and biophysical analysis, to delineate the physiological role of PRRT2. We report that PRRT2 selectively blocks the trans SNARE complex assembly and thus negatively regulates synaptic vesicle priming. This inhibition is actualized via weak interactions of the N-terminal proline-rich domain with the synaptic SNARE proteins. Furthermore, we demonstrate that paroxysmal dyskinesia-associated mutations in PRRT2 disrupt this SNARE-modulatory function and with efficiencies corresponding to the severity of the disease phenotype. Our findings provide insights into the molecular mechanisms through which loss-of-function mutations in PRRT2 result in paroxysmal neurological disorders.
Collapse
Affiliation(s)
- Jeff Coleman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ouardane Jouannot
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sathish K Ramakrishnan
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Maria N Zanetti
- Department of Clinical and Experimental Epilepsy, University College London, London WC1N 3BG, UK
| | - Jing Wang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Vincenzo Salpietro
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Henry Houlden
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London WC1N 3BG, UK
| | - James E Rothman
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Clinical and Experimental Epilepsy, University College London, London WC1N 3BG, UK.
| | - Shyam S Krishnakumar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Clinical and Experimental Epilepsy, University College London, London WC1N 3BG, UK.
| |
Collapse
|
5
|
Flores JA, Ramírez-Ponce MP, Montes MÁ, Balseiro-Gómez S, Acosta J, Álvarez de Toledo G, Alés E. Proteoglycans involved in bidirectional communication between mast cells and hippocampal neurons. J Neuroinflammation 2019; 16:107. [PMID: 31109355 PMCID: PMC6528191 DOI: 10.1186/s12974-019-1504-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/08/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Mast cells (MCs) in the brain can respond to environmental cues and relay signals to neurons that may directly influence neuronal electrical activity, calcium signaling, and neurotransmission. MCs also express receptors for neurotransmitters and consequently can be activated by them. Here, we developed a coculture model of peritoneal MCs, incubated together with dissociated hippocampal neurons for the study of cellular mechanisms involved in the mast cell-neuron interactions. METHODS Calcium imaging was used to simultaneously record changes in intracellular calcium [Ca2+]i in neurons and MCs. To provide insight into the contribution of MCs on neurotransmitter release in rat hippocampal neurons, we used analysis of FM dye release, evoked by a cocktail of mediators from MCs stimulated by heat. RESULTS Bidirectional communication is set up between MCs and hippocampal neurons. Neuronal depolarization caused intracellular calcium [Ca2+]i oscillations in MCs that produced a quick response in neurons. Furthermore, activation of MCs with antigen or the secretagogue compound 48/80 also resulted in a neuronal [Ca2+]i response. Moreover, local application onto neurons of the MC mediator cocktail elicited Ca2+ transients and a synaptic release associated with FM dye destaining. Neuronal response was partially blocked by D-APV, a N-methyl-D-aspartate receptor (NMDAR) antagonist, and was inhibited when the cocktail was pre-digested with chondroitinase ABC, which induces enzymatic removal of proteoglycans of chondroitin sulfate (CS). CONCLUSIONS MC-hippocampal neuron interaction affects neuronal [Ca2+]i and exocytosis signaling through a NMDAR-dependent mechanism.
Collapse
Affiliation(s)
- Juan Antonio Flores
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - María Pilar Ramírez-Ponce
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - María Ángeles Montes
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Santiago Balseiro-Gómez
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
- Present Address: Department of Neuroscience, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06510 USA
| | - Jorge Acosta
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Guillermo Álvarez de Toledo
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| | - Eva Alés
- Dpto. de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009 Sevilla, Spain
| |
Collapse
|
6
|
Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM. Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: An approach for their use in neural regeneration therapies. PLoS One 2019; 14:e0213032. [PMID: 30856179 PMCID: PMC6437714 DOI: 10.1371/journal.pone.0213032] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/13/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can trans/differentiate to neural precursors and/or
mature neurons and promote neuroprotection and neurogenesis. The above could
greatly benefit neurodegenerative disorders as well as in the treatment of
post-traumatic and hereditary diseases of the central nervous system (CNS). In
order to attain an ideal source of adult MSCs for the treatment of CNS diseases,
adipose tissue, bone marrow, skin and umbilical cord derived MSCs were isolated
and studied to explore differences with regard to neural differentiation
capacity. In this study, we demonstrated that MSCs from several tissues can
differentiate into neuron-like cells and differentially express progenitors and
mature neural markers. Adipose tissue MSCs exhibited significantly higher
expression of neural markers and had a faster proliferation rate. Our results
suggest that adipose tissue MSCs are the best candidates for the use in
neurological diseases.
Collapse
Affiliation(s)
| | - Pablo Caviedes
- Program of Molecular & Clinical Pharmacology, ICBM, Faculty of
Medicine, Universidad de Chile, Santiago, Chile
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de
Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y
Matemáticas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Mardones
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- Orthopedic Department, Clinica Las Condes, Santiago,
Chile
| | - José J. Minguell
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
| | - Ana Maria Vega-Letter
- Program of Traslational Immunology ICIM, Faculty of Medicine, Clinica
Alemana Universidad del Desarrollo, Santiago, Chile
| | - Claudio M. Jofre
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- * E-mail:
| |
Collapse
|
7
|
Durán E, Montes MÁ, Jemal I, Satterfield R, Young S, Álvarez de Toledo G. Synaptotagmin-7 controls the size of the reserve and resting pools of synaptic vesicles in hippocampal neurons. Cell Calcium 2018; 74:53-60. [PMID: 29957297 DOI: 10.1016/j.ceca.2018.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/04/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023]
Abstract
Continuous neurotransmitter release is subjected to synaptic vesicle availability, which in turn depends on vesicle recycling and the traffic of vesicles between pools. We studied the role of Synaptotagmin-7 (Syt-7) in synaptic vesicle accessibility for release in hippocampal neurons in culture. Synaptic boutons from Syt-7 knockout (KO) mice displayed normal basal secretion with no alteration in the RRP size or the probability of release. However, stronger stimuli revealed an increase in the size of the reserve and resting vesicle pools in Syt-7 KO boutons compared with WT. These data suggest that Syt-7 plays a significant role in the vesicle pool homeostasis and, consequently, in the availability of vesicles for synaptic transmission during strong stimulation, probably, by facilitating advancing synaptic vesicles to the readily releasable pool.
Collapse
Affiliation(s)
- Elisa Durán
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - María Ángeles Montes
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Imane Jemal
- Dpto. Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Rachel Satterfield
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute, Jupiter, FL 33458. USA
| | - Samuel Young
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute, Jupiter, FL 33458. USA
| | | |
Collapse
|
8
|
Cousin MA, Gordon SL, Smillie KJ. Using FM Dyes to Monitor Clathrin-Mediated Endocytosis in Primary Neuronal Culture. Methods Mol Biol 2018; 1847:239-249. [PMID: 30129022 DOI: 10.1007/978-1-4939-8719-1_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This protocol utilizes lipophilic FM dyes to monitor membrane recycling in real time. FM dyes are virtually nonfluorescent in solution but when membrane bound are intensely fluorescent, combined with the flexibility of different emission wavelengths make these dyes an excellent choice for investigating clathrin-mediated endocytosis, among other membrane trafficking and recycling pathways.
Collapse
Affiliation(s)
- Michael A Cousin
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Sarah L Gordon
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Karen J Smillie
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
9
|
Ferro M, Lamanna J, Ripamonti M, Racchetti G, Arena A, Spadini S, Montesano G, Cortese R, Zimarino V, Malgaroli A. Functional mapping of brain synapses by the enriching activity-marker SynaptoZip. Nat Commun 2017; 8:1229. [PMID: 29089485 PMCID: PMC5663910 DOI: 10.1038/s41467-017-01335-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/08/2017] [Indexed: 01/27/2023] Open
Abstract
Ideally, elucidating the role of specific brain circuits in animal behavior would require the ability to measure activity at all involved synapses, possibly with unrestricted field of view, thus even at those boutons deeply located into the brain. Here, we introduce and validate an efficient scheme reporting synaptic vesicle cycling in vivo. This is based on SynaptoZip, a genetically encoded molecule deploying in the vesicular lumen a bait moiety designed to capture upon exocytosis a labeled alien peptide, Synbond. The resulting signal is cumulative and stores the number of cycling events occurring at individual synapses. Since this functional signal is enduring and measurable both online and ex post, SynaptoZip provides a unique method for the analysis of the history of synaptic activity in regions several millimeters below the brain surface. We show its broad applicability by reporting stimulus-evoked and spontaneous circuit activity in wide cortical fields, in anesthetized and freely moving animals. Visualization of synaptic activity in the living brain is challenging. This study devises a simple and efficient scheme that reports synaptic vesicle recycling in vivo using SynaptoZip, a genetically encoded sensor of past synaptic activities.
Collapse
Affiliation(s)
- Mattia Ferro
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Jacopo Lamanna
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Maddalena Ripamonti
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Gabriella Racchetti
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Alessandro Arena
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy.,Department of Physiology, Institute of Basal Medical Sciences, University of Oslo, Oslo, 0315, Norway
| | - Sara Spadini
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Giovanni Montesano
- Università Vita-Salute San Raffaele, Milan, 20132, Italy.,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy.,Dipartimento Testa-Collo, San Paolo Hospital, University of Milan, Milan, 20122, Italy
| | | | - Vincenzo Zimarino
- Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy
| | - Antonio Malgaroli
- Università Vita-Salute San Raffaele, Milan, 20132, Italy. .,Neurobiology of Learning Unit, Division of Neuroscience, Scientific Institute Ospedale San Raffaele, Milan, 20132, Italy.
| |
Collapse
|
10
|
Complexin Mutants Reveal Partial Segregation between Recycling Pathways That Drive Evoked and Spontaneous Neurotransmission. J Neurosci 2017; 37:383-396. [PMID: 28077717 DOI: 10.1523/jneurosci.1854-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/21/2022] Open
Abstract
Synaptic vesicles fuse at morphological specializations in the presynaptic terminal termed active zones (AZs). Vesicle fusion can occur spontaneously or in response to an action potential. Following fusion, vesicles are retrieved and recycled within nerve terminals. It is still unclear whether vesicles that fuse spontaneously or following evoked release share similar recycling mechanisms. Genetic deletion of the SNARE-binding protein complexin dramatically increases spontaneous fusion, with the protein serving as the synaptic vesicle fusion clamp at Drosophila synapses. We examined synaptic vesicle recycling pathways at complexin null neuromuscular junctions, where spontaneous release is dramatically enhanced. We combined loading of the lipophilic dye FM1-43 with photoconversion, electron microscopy, and electrophysiology to monitor evoked and spontaneous recycling vesicle pools. We found that the total number of recycling vesicles was equal to those retrieved through spontaneous and evoked pools, suggesting that retrieval following fusion is partially segregated for spontaneous and evoked release. In addition, the kinetics of FM1-43 destaining and synaptic depression measured in the presence of the vesicle-refilling blocker bafilomycin indicated that spontaneous and evoked recycling pools partially intermix during the release process. Finally, FM1-43 photoconversion combined with electron microscopy analysis indicated that spontaneous recycling preferentially involves synaptic vesicles in the vicinity of AZs, whereas vesicles recycled following evoked release involve a larger intraterminal pool. Together, these results suggest that spontaneous and evoked vesicles use separable recycling pathways and then partially intermix during subsequent rounds of fusion. SIGNIFICANCE STATEMENT Neurotransmitter release involves fusion of synaptic vesicles with the plasma membrane in response to an action potential, or spontaneously in the absence of stimulation. Upon fusion, vesicles are retrieved and recycled, and it is unclear whether recycling pathways for evoked and spontaneous vesicles are segregated after fusion. We addressed this question by taking advantage of preparations lacking the synaptic protein complexin, which have elevated spontaneous release that enables reliable tracking of the spontaneous recycling pool. Our results suggest that spontaneous and evoked recycling pathways are segregated during the retrieval process but can partially intermix during stimulation.
Collapse
|
11
|
Lee H, Liu W, Brown AS, Landgraf R, Wilson JN. Fluorescent Kinase Probes Enabling Identification and Dynamic Imaging of HER2(+) Cells. Anal Chem 2016; 88:11310-11313. [PMID: 27934102 DOI: 10.1021/acs.analchem.6b03836] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The human epidermal growth factor receptor, EGFR/ERBB/HER, family of receptor tyrosine kinases is central to many signaling pathways and a validated chemotherapy target in multiple cancers. While EGFR/ERBB-targeted therapies, including monoclonal antibodies, e.g., trastuzumab, and small molecule kinase inhibitors, such as lapatinib, have been developed, rapid identification and classification of cancer cells is key to identifying the best treatment regime. We report ERBB2 (also HER2) targeting kinase probes that exhibit a "turn-on" emission response upon binding. These live cell compatible probes differentiate ERBB2(+) cells from low-level, ERBB2(-) cells by targeting the intracellular ATP-binding pocket of ERBB2 with therapeutic inhibitor-like specificity. Beyond kinase expression levels, probe signal is linked to the phosphotyrosine-correlated activation state of the ERBB2 population. Additionally, the rapid signaling capability of the probes can report changes in activation state in live cells providing a unique type of complementary information to immunohistochemical assays of receptor kinase populations.
Collapse
Affiliation(s)
- Heajin Lee
- Department of Chemistry, University of Miami , Coral Gables, Florida 33146, United States
| | - Wenjun Liu
- Department of Biochemistry and Molecular Biology, University of Miami , Miami, Florida 33101, United States
| | - Adrienne S Brown
- Department of Chemistry, University of Miami , Coral Gables, Florida 33146, United States
| | - Ralf Landgraf
- Department of Biochemistry and Molecular Biology, University of Miami , Miami, Florida 33101, United States
| | - James N Wilson
- Department of Chemistry, University of Miami , Coral Gables, Florida 33146, United States
| |
Collapse
|
12
|
Rauti R, Lozano N, León V, Scaini D, Musto M, Rago I, Ulloa Severino FP, Fabbro A, Casalis L, Vázquez E, Kostarelos K, Prato M, Ballerini L. Graphene Oxide Nanosheets Reshape Synaptic Function in Cultured Brain Networks. ACS NANO 2016; 10:4459-71. [PMID: 27030936 DOI: 10.1021/acsnano.6b00130] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Graphene offers promising advantages for biomedical applications. However, adoption of graphene technology in biomedicine also poses important challenges in terms of understanding cell responses, cellular uptake, or the intracellular fate of soluble graphene derivatives. In the biological microenvironment, graphene nanosheets might interact with exposed cellular and subcellular structures, resulting in unexpected regulation of sophisticated biological signaling. More broadly, biomedical devices based on the design of these 2D planar nanostructures for interventions in the central nervous system require an accurate understanding of their interactions with the neuronal milieu. Here, we describe the ability of graphene oxide nanosheets to down-regulate neuronal signaling without affecting cell viability.
Collapse
Affiliation(s)
- Rossana Rauti
- Life Science Department, University of Trieste , 34127 Trieste, Italy
| | - Neus Lozano
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Veronica León
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Denis Scaini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | - Mattia Musto
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| | - Ilaria Rago
- ELETTRA Synchrotron Light Source , 34149 Trieste, Italy
| | | | - Alessandra Fabbro
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
| | | | - Ester Vázquez
- Departamento de Química Orgánica, Facultad de Ciencias y Tecnologías Químicas-IRICA, Universidad de Castilla La Mancha , 13071 Ciudad Real, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab, School of Medicine and National Graphene Institute, Faculty of Medical & Human Sciences, University of Manchester , M13 9PL Manchester, United Kingdom
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste , 34127 Trieste, Italy
- CIC BiomaGUNE, Parque Tecnológico de San Sebastián, Paseo Miramón, 182, 20009 San Sebastián, Guipúzcoa, Spain
- Basque Foundation for Science , Ikerbasque, Bilbao 48013, Spain
| | - Laura Ballerini
- Life Science Department, University of Trieste , 34127 Trieste, Italy
- International School for Advanced Studies (SISSA) , 34136 Trieste, Italy
| |
Collapse
|
13
|
Slater CR. The functional organization of motor nerve terminals. Prog Neurobiol 2015; 134:55-103. [DOI: 10.1016/j.pneurobio.2015.09.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/28/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
|
14
|
Maiti P, Manna J, McDonald MP. Merging advanced technologies with classical methods to uncover dendritic spine dynamics: A hot spot of synaptic plasticity. Neurosci Res 2015; 96:1-13. [PMID: 25728560 DOI: 10.1016/j.neures.2015.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 02/17/2015] [Accepted: 02/19/2015] [Indexed: 01/08/2023]
Abstract
The structure of dendritic spines determines synaptic efficacy, a plastic process that mediates information processing in the vertebrate nervous system. Aberrant spine morphology, including alterations in shape, size, and number, are common in different brain diseases. Because of this, accurate and unbiased characterization of dendritic spine structure is vital to our ability to explore and understand their involvement in neuronal development, synaptic plasticity, and synaptic failure in neurological diseases. Investigators have attempted to elucidate the precise structure and function of dendritic spines for more than a hundred years, but their fundamental role in synaptic plasticity and neurological diseases remains elusive. Limitations and ambiguities in imaging techniques have exacerbated the challenges of acquiring accurate information about spines and spine features. However, recent advancements in molecular biology, protein engineering, immuno-labeling techniques, and the use of super-resolution nano-microscopy along with powerful image analysis software have provided a better understanding of dendritic spine architecture. Here we describe the pros and cons of the classical staining techniques used to study spine morphology, and the alteration of dendritic spines in various neuropathological conditions. Finally, we highlight recent advances in super-resolved nanoscale microscopy, and their potentials and pitfalls when used to explore dendritic spine dynamics.
Collapse
Affiliation(s)
- Panchanan Maiti
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Michael P McDonald
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
15
|
Abstract
Secretory carrier membrane protein 5 (SCAMP5), a recently identified candidate gene for autism, is brain specific and highly abundant in synaptic vesicles (SVs), but its function is currently unknown. Here, we found that knockdown (KD) of endogenous SCAMP5 by SCAMP5-specific shRNAs in cultured rat hippocampal neurons resulted in a reduction in total vesicle pool size as well as in recycling pool size, but the recycling/resting pool ratio was significantly increased. SCAMP5 KD slowed endocytosis after stimulation, but impaired it severely during strong stimulation. We also found that KD dramatically lowered the threshold of activity at which SV endocytosis became unable to compensate for the ongoing exocytosis occurring during a stimulus. Reintroducing shRNA-resistant SCAMP5 reversed these endocytic defects. Therefore, our results suggest that SCAMP5 functions during high neuronal activity when a heavy load is imposed on endocytosis. Our data also raise the possibility that the reduction in expression of SCAMP5 in autistic patients may be related to the synaptic dysfunction observed in autism.
Collapse
|
16
|
Nimmervoll B, Flucher BE, Obermair GJ. Dominance of P/Q-type calcium channels in depolarization-induced presynaptic FM dye release in cultured hippocampal neurons. Neuroscience 2013; 253:330-40. [PMID: 24012836 PMCID: PMC3824072 DOI: 10.1016/j.neuroscience.2013.08.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/09/2013] [Accepted: 08/27/2013] [Indexed: 11/26/2022]
Abstract
We analyzed depolarization-induced synaptic FM dye release in hippocampal neurons. We pharmacologically isolated the contribution of voltage-gated Ca2+ channels. 85% of synapses utilize N- and P/Q-type channels, 15% only P/Q-type channels. In both groups of synapses release kinetics are determined by P/Q-type channels. We propose a more direct coupling of P/Q-type channels to synaptic release.
Neurotransmitter release probability is related by high power to the local concentration of calcium in presynaptic terminals, which in turn is controlled by voltage-gated calcium channels. P/Q- and N-type channels trigger synaptic transmission in the majority of neurons of the central nervous system. However, whether and under which conditions both channel types act cooperatively or independently is still insufficiently understood. Previous studies suggested either a dominance of N- or P/Q-type channels, or a synergistic action of both channels, depending on the experimental paradigms. Thus, to provide insight into the properties of neurotransmitter release in cultured mouse hippocampal neurons, we used quantitative analysis of FM dye release from presynaptic boutons induced by high potassium membrane depolarization. Increasing extracellular potassium concentrations revealed a sigmoid dependence of FM dye release to the stimulation strength. Individual and combined application of the P/Q- and N-type channel-specific blockers ω-agatoxin-IVA and ω-conotoxin-GVIA, respectively, allowed us to specifically isolate the contribution of both channel types to release triggered with 40 mM KCl. Analysis of the release kinetics and the fractional release amplitude demonstrate that, whereas in only 15% of the synapses release depended exclusively on P/Q-type channels, the majority of synapses (85%) contained both N- and P/Q-type channels. Nevertheless, the kinetics of FM dye release in synapses containing both channel types was determined by the P/Q-type channels. Together, our data suggest a more direct coupling of P/Q-type channels to synaptic release compared to N-type channels, which may explain the high prevalence of neurological P/Q-type channelopathies.
Collapse
Affiliation(s)
- B Nimmervoll
- Division of Physiology, Medical University Innsbruck, Fritz-Pregl-Str. 3, 6020 Innsbruck, Austria
| | | | | |
Collapse
|
17
|
Hallermann S, Silver RA. Sustaining rapid vesicular release at active zones: potential roles for vesicle tethering. Trends Neurosci 2013; 36:185-94. [PMID: 23164531 PMCID: PMC4748400 DOI: 10.1016/j.tins.2012.10.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 10/18/2012] [Accepted: 10/21/2012] [Indexed: 12/12/2022]
Abstract
Rapid information processing in our nervous system relies on high-frequency fusion of transmitter-filled vesicles at chemical synapses. Some sensory synapses possess prominent electron-dense ribbon structures that provide a scaffold for tethering synaptic vesicles at the active zone (AZ), enabling sustained vesicular release. Here, we review functional data indicating that some central and neuromuscular synapses can also sustain vesicle-fusion rates that are comparable to those of ribbon-type sensory synapses. Comparison of the ultrastructure across these different types of synapses, together with recent work showing that cytomatrix proteins can tether vesicles and speed vesicle reloading, suggests that filamentous structures may play a key role in vesicle supply. We discuss potential mechanisms by which vesicle tethering could contribute to sustained high rates of vesicle fusion across ribbon-type, central, and neuromuscular synapses.
Collapse
Affiliation(s)
- Stefan Hallermann
- European Neuroscience Institute Göttingen, Grisebachstrasse 5, 37077 Göttingen, Germany.
| | | |
Collapse
|
18
|
Sevrain D, Le Grand Y, Buhé V, Jeanmaire C, Pauly G, Carré JL, Misery L, Lebonvallet N. Two-photon microscopy of dermal innervation in a human re-innervated model of skin. Exp Dermatol 2013; 22:290-1. [DOI: 10.1111/exd.12108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 11/29/2022]
Affiliation(s)
- David Sevrain
- Laboratoire de Spectrométrie et Optique Laser; Université de Brest; UEB; Brest; France
| | - Yann Le Grand
- Laboratoire de Spectrométrie et Optique Laser; Université de Brest; UEB; Brest; France
| | - Virginie Buhé
- Laboratoire des Neurosciences de Brest; Université de Brest; LNB EA4685 UEB; Brest; France
| | - Christine Jeanmaire
- BASF Beauty Creations; BASF Beauty Care Solutions France SAS; Pulnoy; France
| | - Gilles Pauly
- BASF Beauty Creations; BASF Beauty Care Solutions France SAS; Pulnoy; France
| | - Jean-Luc Carré
- Laboratoire des Neurosciences de Brest; Université de Brest; LNB EA4685 UEB; Brest; France
| | - Laurent Misery
- Laboratoire des Neurosciences de Brest; Université de Brest; LNB EA4685 UEB; Brest; France
| | | |
Collapse
|
19
|
Automated condition-invariable neurite segmentation and synapse classification using textural analysis-based machine-learning algorithms. J Neurosci Methods 2012; 213:84-98. [PMID: 23261652 DOI: 10.1016/j.jneumeth.2012.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 11/24/2022]
Abstract
High-resolution live-cell imaging studies of neuronal structure and function are characterized by large variability in image acquisition conditions due to background and sample variations as well as low signal-to-noise ratio. The lack of automated image analysis tools that can be generalized for varying image acquisition conditions represents one of the main challenges in the field of biomedical image analysis. Specifically, segmentation of the axonal/dendritic arborizations in brightfield or fluorescence imaging studies is extremely labor-intensive and still performed mostly manually. Here we describe a fully automated machine-learning approach based on textural analysis algorithms for segmenting neuronal arborizations in high-resolution brightfield images of live cultured neurons. We compare performance of our algorithm to manual segmentation and show that it combines 90% accuracy, with similarly high levels of specificity and sensitivity. Moreover, the algorithm maintains high performance levels under a wide range of image acquisition conditions indicating that it is largely condition-invariable. We further describe an application of this algorithm to fully automated synapse localization and classification in fluorescence imaging studies based on synaptic activity. Textural analysis-based machine-learning approach thus offers a high performance condition-invariable tool for automated neurite segmentation.
Collapse
|
20
|
Chen T, Wang X, von Wangenheim D, Zheng M, Šamaj J, Ji W, Lin J. Probing and tracking organelles in living plant cells. PROTOPLASMA 2012; 249 Suppl 2:S157-S167. [PMID: 22183127 DOI: 10.1007/s00709-011-0364-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 12/06/2011] [Indexed: 05/31/2023]
Abstract
Intracellular organelle movements and positioning play pivotal roles in enabling plants to proliferate life efficiently and to survive diverse environmental stresses. The elaborate dissection of organelle dynamics and their underlying mechanisms (e.g., the role of the cytoskeleton in organelle movements) largely depends on the advancement and efficiency of organelle tracking systems. Here, we provide an overview of some recently developed tools for labeling and tracking organelle dynamics in living plant cells.
Collapse
Affiliation(s)
- Tong Chen
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Mancuso JJ, Chen Y, Li X, Xue Z, Wong STC. Methods of dendritic spine detection: from Golgi to high-resolution optical imaging. Neuroscience 2012; 251:129-40. [PMID: 22522468 DOI: 10.1016/j.neuroscience.2012.04.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 03/30/2012] [Accepted: 04/05/2012] [Indexed: 12/18/2022]
Abstract
Dendritic spines, the bulbous protrusions that form the postsynaptic half of excitatory synapses, are one of the most prominent features of neurons and have been imaged and studied for over a century. In that time, changes in the number and morphology of dendritic spines have been correlated to the developmental process as well as the pathophysiology of a number of neurodegenerative diseases. Due to the sheer scale of synaptic connectivity in the brain, work to date has merely scratched the surface in the study of normal spine function and pathology. This review will highlight traditional approaches to the imaging of dendritic spines and newer approaches made possible by advances in microscopy, protein engineering, and image analysis. The review will also describe recent work that is leading researchers toward the possibility of a systematic and comprehensive study of spine anatomy throughout the brain.
Collapse
Affiliation(s)
- J J Mancuso
- Department of Systems Medicine and Bioengineering, The Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX 77030, USA; Ting Tsung and Wei Fong Chao Center for Bioinformatics Research and Imaging in Neurosciences, USA
| | | | | | | | | |
Collapse
|
22
|
Koch M, Holt M. Coupling exo- and endocytosis: an essential role for PIP₂ at the synapse. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:1114-32. [PMID: 22387937 DOI: 10.1016/j.bbalip.2012.02.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Revised: 02/12/2012] [Accepted: 02/13/2012] [Indexed: 12/24/2022]
Abstract
Chemical synapses are specialist points of contact between two neurons, where information transfer takes place. Communication occurs through the release of neurotransmitter substances from small synaptic vesicles in the presynaptic terminal, which fuse with the presynaptic plasma membrane in response to neuronal stimulation. However, as neurons in the central nervous system typically only possess ~200 vesicles, high levels of release would quickly lead to a depletion in the number of vesicles, as well as leading to an increase in the area of the presynaptic plasma membrane (and possible misalignment with postsynaptic structures). Hence, synaptic vesicle fusion is tightly coupled to a local recycling of synaptic vesicles. For a long time, however, the exact molecular mechanisms coupling fusion and subsequent recycling remained unclear. Recent work now indicates a unique role for the plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP(2)), acting together with the vesicular protein synaptotagmin, in coupling these two processes. In this work, we review the evidence for such a mechanism and discuss both the possible advantages and disadvantages for vesicle recycling (and hence signal transduction) in the nervous system. This article is part of a Special Issue entitled Lipids and Vesicular Transport.
Collapse
Affiliation(s)
- Marta Koch
- Laboratory of Neurogenetics, VIB Center for the Biology of Disease and K.U. Leuven Center for Human Genetics, O&N4 Herestraat 49, 3000 Leuven, Belgium
| | | |
Collapse
|
23
|
Optogenetic reporters: Fluorescent protein-based genetically encoded indicators of signaling and metabolism in the brain. PROGRESS IN BRAIN RESEARCH 2012; 196:235-63. [PMID: 22341329 DOI: 10.1016/b978-0-444-59426-6.00012-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Fluorescent protein technology has evolved to include genetically encoded biosensors that can monitor levels of ions, metabolites, and enzyme activities as well as protein conformation and even membrane voltage. They are well suited to live-cell microscopy and quantitative analysis, and they can be used in multiple imaging modes, including one- or two-photon fluorescence intensity or lifetime microscopy. Although not nearly complete, there now exists a substantial set of genetically encoded reporters that can be used to monitor many aspects of neuronal and glial biology, and these biosensors can be used to visualize synaptic transmission and activity-dependent signaling in vitro and in vivo. In this review, we present an overview of design strategies for engineering biosensors, including sensor designs using circularly permuted fluorescent proteins and using fluorescence resonance energy transfer between fluorescent proteins. We also provide examples of indicators that sense small ions (e.g., pH, chloride, zinc), metabolites (e.g., glutamate, glucose, ATP, cAMP, lipid metabolites), signaling pathways (e.g., G protein-coupled receptors, Rho GTPases), enzyme activities (e.g., protein kinase A, caspases), and reactive species. We focus on examples where these genetically encoded indicators have been applied to brain-related studies and used with live-cell fluorescence microscopy.
Collapse
|
24
|
Hempel CM, Sivula M, Levenson JM, Rose DM, Li B, Sirianni AC, Xia E, Ryan TA, Gerber DJ, Cottrell JR. A system for performing high throughput assays of synaptic function. PLoS One 2011; 6:e25999. [PMID: 21998743 PMCID: PMC3187845 DOI: 10.1371/journal.pone.0025999] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 09/15/2011] [Indexed: 11/20/2022] Open
Abstract
Unbiased, high-throughput screening has proven invaluable for dissecting complex biological processes. Application of this general approach to synaptic function would have a major impact on neuroscience research and drug discovery. However, existing techniques for studying synaptic physiology are labor intensive and low-throughput. Here, we describe a new high-throughput technology for performing assays of synaptic function in primary neurons cultured in microtiter plates. We show that this system can perform 96 synaptic vesicle cycling assays in parallel with high sensitivity, precision, uniformity, and reproducibility and can detect modulators of presynaptic function. By screening libraries of pharmacologically defined compounds on rat forebrain cultures, we have used this system to identify novel effects of compounds on specific aspects of presynaptic function. As a system for unbiased compound as well as genomic screening, this technology has significant applications for basic neuroscience research and for the discovery of novel, mechanism-based treatments for central nervous system disorders.
Collapse
Affiliation(s)
- Chris M. Hempel
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | - Michael Sivula
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | | | - David M. Rose
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | - Bing Li
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | - Ana C. Sirianni
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | - Eva Xia
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | - Timothy A. Ryan
- Department of Biochemistry, Weill Cornell Medical College, New York, New York, United States of America
| | - David J. Gerber
- Galenea Corporation, Cambridge, Massachusetts, United States of America
| | | |
Collapse
|
25
|
Liu D, Martina JA, Wu XS, Hammer JA, Long EO. Two modes of lytic granule fusion during degranulation by natural killer cells. Immunol Cell Biol 2011; 89:728-38. [PMID: 21483445 PMCID: PMC3257049 DOI: 10.1038/icb.2010.167] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lytic granules in cytotoxic lymphocytes, which include T cells and natural killer (NK) cells, are secretory lysosomes that release their content upon fusion with the plasma membrane (PM), a process known as degranulation. Although vesicle exocytosis has been extensively studied in endocrine and neuronal cells, much less is known about the fusion of lytic granules in cytotoxic lymphocytes. Here, we used total internal reflection fluorescence microscopy to examine lytic granules labeled with fluorescently tagged Fas ligand (FasL) in the NK cell line NKL stimulated with phorbol ester and ionomycin and in primary NK cells activated by physiological receptor–ligand interactions. Two fusion modes were observed: complete fusion, characterized by loss of granule content and rapid diffusion of FasL at the PM; and incomplete fusion, characterized by transient fusion pore opening and retention of FasL at the fusion site. The pH-sensitive green fluorescence protein (pHluorin) fused to the lumenal domain of FasL was used to visualize fusion pore opening with a time resolution of 30 ms. Upon incomplete fusion, pHluorin emission lasted several seconds in the absence of noticeable diffusion. Thus, we conclude that lytic granules in NK cells undergo both complete and incomplete fusion with the PM, and propose that incomplete fusion may promote efficient recycling of lytic granule membrane after the release of cytotoxic effector molecules.
Collapse
Affiliation(s)
- Dongfang Liu
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | | | | | | | | |
Collapse
|
26
|
Peled ES, Isacoff EY. Optical quantal analysis of synaptic transmission in wild-type and rab3-mutant Drosophila motor axons. Nat Neurosci 2011; 14:519-26. [PMID: 21378971 DOI: 10.1038/nn.2767] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 01/21/2011] [Indexed: 02/06/2023]
Abstract
Synaptic transmission from a neuron to its target cells occurs via neurotransmitter release from dozens to thousands of presynaptic release sites whose strength and plasticity can vary considerably. We report an in vivo imaging method that monitors real-time synaptic transmission simultaneously at many release sites with quantal resolution. We applied this method to the model glutamatergic system of the Drosophila melanogaster larval neuromuscular junction. We find that, under basal conditions, about half of release sites have a very low release probability, but these are interspersed with sites with as much as a 50-fold higher probability. Paired-pulse stimulation depresses high-probability sites, facilitates low-probability sites, and recruits previously silent sites. Mutation of the small GTPase Rab3 substantially increases release probability but still leaves about half of the sites silent. Our findings suggest that basal synaptic strength and short-term plasticity are regulated at the level of release probability at individual sites.
Collapse
Affiliation(s)
- Einat S Peled
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | | |
Collapse
|
27
|
Frequency-dependent modes of synaptic vesicle endocytosis and exocytosis at adult mouse neuromuscular junctions. J Neurosci 2011; 31:1093-105. [PMID: 21248134 DOI: 10.1523/jneurosci.2800-10.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During locomotion, adult rodent lumbar motoneurons fire in high-frequency (80-100 Hz) 1-2 s bursts every several seconds, releasing between 10,000 and 20,000 vesicles per burst. The estimated total vesicle pool size indicates that all vesicles would be used within 30 s; thus, a mechanism for rapid endocytosis and vesicle recycling is necessary to maintain effective transmission and motor behavior. However, whether such rapid recycling exists at mouse neuromuscular junctions (NMJs) or how it is regulated has been unclear. Here, we show that much less FM1-43 dye is lost per stimulus with 100 Hz stimulation than with 10 Hz stimulation even when the same number of vesicles undergo exocytosis. Electrophysiological data using folimycin show this lesser amount of dye loss is caused in part by the rapid reuse of vesicles. We showed previously that a myosin light chain kinase (MLCK)-myosin II pathway was required for effective transmission at 100 Hz. Here, we confirm the activation of MLCK, based on increased nerve terminal phospho-MLC immunostaining, with 100 Hz but not with 10 Hz stimulation. We further demonstrate that activation of MLCK, by increased extracellular Ca(2+), by PKC (protein kinase C) activation, or by a MLCK agonist peptide, reduces the amount of dye lost even with 10 Hz stimulation. MLCK activation at 10 Hz also resulted in more vesicles being rapidly reused. Thus, MLCK activation by 100 Hz stimulation switches the mechanism of vesicle cycling to a rapid-reuse mode and is required to sustain effective transmission in adult mouse NMJs.
Collapse
|
28
|
Branco T, Marra V, Staras K. Examining size-strength relationships at hippocampal synapses using an ultrastructural measurement of synaptic release probability. J Struct Biol 2010; 172:203-10. [PMID: 19895891 PMCID: PMC3084449 DOI: 10.1016/j.jsb.2009.10.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 09/25/2009] [Accepted: 10/27/2009] [Indexed: 11/19/2022]
Abstract
Release probability (p(r)) is a fundamental presynaptic parameter which is critical in defining synaptic strength. Knowledge of how synapses set and regulate their p(r) is a fundamental step in understanding synaptic transmission and communication between neurons. Despite its importance, p(r) is difficult to measure directly at single synapses. One important strategy to achieve this has relied on the application of fluorescence-based imaging methods, but this is always limited by the lack of detailed information on the morphological and structural properties of the individual synapses under study, and thus precludes an investigation of the relationship between p(r) and synaptic anatomy. Here we outline a powerful methodology based on using FM-styryl dyes, photoconversion and correlative ultrastructural analysis in dissociated hippocampal cultured neurons, which provides both a direct readout of p(r) as well as nanoscale detail on synaptic organization and structure. We illustrate the value of this approach by investigating, at the level of individual reconstructed terminals, the relationship between release probability and defined vesicle pools. We show that in our population of synapses, p(r) is highly variable, and while it is positively correlated with the number of vesicles docked at the active zone it shows no relationship with the total number of synaptic vesicles. The lack of a direct correlation between total synaptic size and performance in these terminals suggests that factors other than the absolute magnitude of the synapse are the most important determinants of synaptic efficacy.
Collapse
Affiliation(s)
- Tiago Branco
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Vincenzo Marra
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Kevin Staras
- School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| |
Collapse
|
29
|
Lucido AL, Gopalakrishnan G, Yam PT, Colman DR, Lennox RB. Isolation of functional presynaptic complexes from CNS neurons: a cell-free preparation for the study of presynaptic compartments In vitro. ACS Chem Neurosci 2010; 1:535-41. [PMID: 22777142 DOI: 10.1021/cn100048z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 06/17/2010] [Indexed: 11/30/2022] Open
Abstract
The difficulty in developing successful treatments to facilitate nerve regeneration has prompted a number of new in vitro experimental methods. We have recently shown that functional presynaptic boutons can be formed when neuronal cells are cocultured with surface-modified artificial substrates including poly(d-lysine)-coated beads and supported lipid bilayer-coated beads (Lucido(2009) J. Neurosci.29, 12449-12466; Gopalakrishnan(2010) ACS Chem. Neurosci.1, 86-94). We demonstrate here, using confocal microscopy combined with immunocytochemistry, that it is possible to isolate such in vitro presynaptic endings in an exclusive fashion onto glass substrates through a simple "sandwich/lift-off" technique (Perez(2006) Adv. Funct. Mater.16, 306-312). Isolated presynaptic complexes are capable of releasing and recycling neurotransmitter in response to an external chemical trigger. These bead-presynaptic complexes are facile to prepare and are readily dispersible in solution. They are thus compatible with many experimental methods whose focus is the study of the neuronal presynaptic compartment.
Collapse
Affiliation(s)
- Anna Lisa Lucido
- Montreal Neurological Institute & Hospital, 3801 University Street
- McGill Program in NeuroEngineering
| | - Gopakumar Gopalakrishnan
- Montreal Neurological Institute & Hospital, 3801 University Street
- Department of Chemistry, 801 Sherbrooke Street West
- McGill Program in NeuroEngineering
- FQRNT Centre for Self-Assembled Chemical Structures (CSACS)
| | - Patricia T. Yam
- Montreal Neurological Institute & Hospital, 3801 University Street
- McGill Program in NeuroEngineering
| | - David R. Colman
- Montreal Neurological Institute & Hospital, 3801 University Street
- McGill Program in NeuroEngineering
| | - R. Bruce Lennox
- Department of Chemistry, 801 Sherbrooke Street West
- McGill Program in NeuroEngineering
- FQRNT Centre for Self-Assembled Chemical Structures (CSACS)
| |
Collapse
|
30
|
Staras K, Branco T. Sharing vesicles between central presynaptic terminals: implications for synaptic function. Front Synaptic Neurosci 2010; 2:20. [PMID: 21423506 PMCID: PMC3059672 DOI: 10.3389/fnsyn.2010.00020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 05/27/2010] [Indexed: 11/13/2022] Open
Abstract
Presynaptic terminals in hippocampal neurons house functionally distinct vesicle pools, the size, structure and biochemical features of which are major determinants of presynaptic strength and performance. In classical models of synaptic function these vesicle pools are synapse-specific, but accumulating evidence is now demonstrating that some vesicles are laterally mobile along axons and readily shared in a functional manner across adjacent terminals. In effect then, these mobile vesicles represent a further class of synapse-spanning vesicle pool, or "superpool". Here we outline the characteristics of this additional pool type, discussing its structural organization within axons and presynaptic terminals as well as its relationship with conventional vesicle pools. We draw comparisons between extrasynaptic vesicle dynamics and the growing literature on extrasynaptic mobility of non-vesicular synaptic elements which, taken together, raise important questions about the operational independence of adjacent release sites. We also examine the functional implications of lateral vesicle sharing, from the notion that extrasynaptic vesicles can contribute to the release capabilities of individual terminals, to its potential role as a substrate for facilitating changes in synaptic weight as a basis for plasticity.
Collapse
Affiliation(s)
- Kevin Staras
- School of Life Sciences, University of Sussex Brighton, UK
| | | |
Collapse
|
31
|
Opazo F, Punge A, Bückers J, Hoopmann P, Kastrup L, Hell SW, Rizzoli SO. Limited Intermixing of Synaptic Vesicle Components upon Vesicle Recycling. Traffic 2010; 11:800-12. [DOI: 10.1111/j.1600-0854.2010.01058.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Clayton EL, Cousin MA. Quantitative monitoring of activity-dependent bulk endocytosis of synaptic vesicle membrane by fluorescent dextran imaging. J Neurosci Methods 2009; 185:76-81. [PMID: 19766140 DOI: 10.1016/j.jneumeth.2009.09.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 09/08/2009] [Accepted: 09/09/2009] [Indexed: 11/20/2022]
Abstract
Activity-dependent bulk endocytosis (ADBE) is the dominant synaptic vesicle (SV) retrieval mode in central nerve terminals during periods of intense neuronal activity. Despite this fact there are very few real time assays that report the activity of this critical SV retrieval mode. In this paper we report a simple and quantitative assay of ADBE using uptake of large flourescent dextrans as fluid phase markers. We show that almost all dextran uptake occurs in nerve terminals, using co-localisation with the fluorescent probe FM1-43. We also demonstrate that accumulated dextran cannot be unloaded by neuronal stimulation, indicating its specific loading into bulk endosomes and not SVs. Quantification of dextran uptake was achieved by using thresholding analysis to count the number of loaded nerve terminals, since monitoring the average fluorescence intensity of these nerve terminals did not accurately report the extent of ADBE. Using this analysis we showed that dextran uptake occurs very soon after stimulation and that it does not persist when stimulation terminates. Thus we have devised a simple and quantitative method to monitor ADBE in living neurones, which will be ideal for real time screening of small molecule inhibitors of this key SV retrieval mode.
Collapse
Affiliation(s)
- Emma Louise Clayton
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, UK
| | | |
Collapse
|
33
|
Stella SL, Hu WD, Brecha NC. Adenosine suppresses exocytosis from cone terminals of the salamander retina. Neuroreport 2009; 20:923-9. [PMID: 19491713 DOI: 10.1097/wnr.0b013e32832ca4b0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the retina, adenosine is released in the dark and has been shown to inhibit Ca2+ influx through voltage-gated Ca2+ channels in cones. Therefore, we tested whether adenosine can inhibit exocytosis from isolated cone photoreceptors. Simultaneous measurements of membrane exocytosis and Ca2+ were made from cones using the activity-dependent dye, Synaptored-C2, and the Ca2+ indicator dye, Fluo-4. Adenosine suppressed exocytosis in cones, indicating that transmitter release is also reduced from cone terminals, and further supports an inhibitory mechanism for modulating transmitter release onto second-order neurons. Furthermore, this raises the possibility that adenosine might be neuroprotective for photoreceptors and second-order neurons by suppressing Ca2+ levels in cones and reducing exocytosis of L-glutamate, respectively.
Collapse
Affiliation(s)
- Salvatore L Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1763, USA.
| | | | | |
Collapse
|
34
|
Calì C, Marchaland J, Spagnuolo P, Gremion J, Bezzi P. Regulated exocytosis from astrocytes physiological and pathological related aspects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 85:261-93. [PMID: 19607976 DOI: 10.1016/s0074-7742(09)85020-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Astrocytes have traditionally been considered ancillary, satellite cells of the nervous system. However, it is a very recent acquisition that glial cells generate signaling loops which are integral to the brain circuitry and participate, interactively with neuronal networks, in the processing of information. Such a conceptual breakthrough makes this field of investigation one of the hottest in neuroscience, as it calls for a revision of past theories of brain function as well as for new strategies of experimental exploration of brain function. Glial cells are electrically not excitable, and it was only the use of optical recording techniques together with calcium sensitive dyes, that allowed the chemical excitability of glial cells to become apparent. Studies using these new techniques have shown for the first time that glial cells are activated by surrounding synaptic activity and translate neuronal signals into their own calcium code. Intracellular calcium concentration([Ca2+]i) elevations in glial cells have then shown to underlie spatial transfer of information in the glial network, accompanied by release of chemical transmitters (gliotransmitters) such as glutamate and back-signaling to neurons. As a consequence, optical imaging techniques applied to cell cultures or intact tissue have become a state-of-the-art technology for studying glial cell signaling. The molecular mechanisms leading to release of "gliotransmitters," especially glutamate, from glia are under debate. Accumulating evidence clearly indicates that astrocytes secrete numerous transmitters by Ca(2+)-dependent exocytosis. This review will discuss the mechanisms underlying the release of chemical transmitters from astrocytes with a particular emphasis to the regulated exocytosis processes.
Collapse
Affiliation(s)
- Corrado Calì
- Department of Cellular Biology and Morphology (DBCM), Faculty of Medicine, University of Lausanne, rue du Bugnon 9, 1005 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
35
|
Xia X, Lessmann V, Martin TFJ. Imaging of evoked dense-core-vesicle exocytosis in hippocampal neurons reveals long latencies and kiss-and-run fusion events. J Cell Sci 2008; 122:75-82. [PMID: 19066284 DOI: 10.1242/jcs.034603] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Evoked neuropeptide secretion in the central nervous system occurs slowly, but the basis for slow release is not fully understood. Whereas exocytosis of single synaptic vesicles in neurons and of dense-core vesicles (DCVs) in endocrine cells have been directly visualized, single DCV exocytic events in neurons of the central nervous system have not been previously studied. We imaged DCV exocytosis in primary cultured hippocampal neurons using fluorescent propeptide cargo and total internal reflectance fluorescence microscopy. The majority of Ca(2+)-triggered exocytic events occurred from immobile plasma-membrane-proximal DCVs in the cell soma, whereas there were few events in the neurites. Strikingly, DCVs in the cell soma exhibited 50-fold greater release probabilities than those in neurites. Latencies to depolarization-evoked fusion for DCVs were surprisingly long, occurring with an average time constant (tau) of 16 seconds for DCVs in the soma and even longer for DCVs in neurites. All of the single DCV release events exhibited rapid fusion-pore openings and closures, the kinetics of which were highly dependent upon Ca(2+) levels. These ;kiss-and-run' events were associated with limited cargo secretion. Thus, the slow evoked release of neuropeptides could be attributed to very prolonged latencies from stimulation to fusion and transient fusion-pore openings that might limit cargo secretion.
Collapse
Affiliation(s)
- Xiaofeng Xia
- Department of Biochemistry, University of Wisconsin, Madison, WI 53706, USA
| | | | | |
Collapse
|
36
|
Ben Gedalya T, Loeb V, Israeli E, Altschuler Y, Selkoe DJ, Sharon R. Alpha-synuclein and polyunsaturated fatty acids promote clathrin-mediated endocytosis and synaptic vesicle recycling. Traffic 2008; 10:218-34. [PMID: 18980610 DOI: 10.1111/j.1600-0854.2008.00853.x] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Alpha-synuclein (alphaS) is an abundant neuronal cytoplasmic protein implicated in Parkinson's disease (PD), but its physiological function remains unknown. Consistent with its having structural motifs shared with class A1 apolipoproteins, alphaS can reversibly associate with membranes and help regulate membrane fatty acid composition. We previously observed that variations in alphaS expression level in dopaminergic cultured cells or brains are associated with changes in polyunsaturated fatty acid (PUFA) levels and altered membrane fluidity. We now report that alphaS acts with PUFAs to enhance the internalization of the membrane-binding dye, FM 1-43. Specifically, alphaS expression coupled with exposure to physiological levels of certain PUFAs enhanced clathrin-mediated endocytosis in neuronal and non-neuronal cultured cells. Moreover, alphaS expression and PUFA-enhanced basal and -evoked synaptic vesicle (SV) endocytosis in primary hippocampal cultures of wild type (wt) and genetically depleted alphaS mouse brains. We suggest that alphaS and PUFAs normally function in endocytic mechanisms and are specifically involved in SV recycling upon neuronal stimulation.
Collapse
Affiliation(s)
- Tziona Ben Gedalya
- Department of Cellular Biochemistry and Human Genetics, Faculty of Medicine, Hebrew University, Ein-Kerem, Jerusalem 91120, Israel
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The formation of memories relies in part on plastic changes at synapses between neurons. Although the mechanisms of synaptic plasticity have been studied extensively over several decades, many aspects of this process remain controversial. The cellular locus of expression of long-term potentiation (LTP), a major form of synaptic plasticity, is one of the most important unresolved phenomena. In this article, some recent advances in this area made possible by the development of new imaging tools are summarized. These studies have demonstrated that LTP is compound in nature and consists of both presynaptic and postsynaptic components. Some features of presynaptic and postsynaptic changes during compound LTP are also reviewed.
Collapse
Affiliation(s)
- Jay A Blundon
- Department of Development Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105-2794, USA
| | | |
Collapse
|
38
|
Royle SJ, Granseth B, Odermatt B, Derevier A, Lagnado L. Imaging phluorin-based probes at hippocampal synapses. Methods Mol Biol 2008; 457:293-303. [PMID: 19066036 PMCID: PMC3474192 DOI: 10.1007/978-1-59745-261-8_22] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Accurate measurement of synaptic vesicle exocytosis and endocytosis is crucial to understanding the molecular basis of synaptic transmission. The fusion of a pH-sensitive green fluorescent protein (pHluorin) to various synaptic vesicle proteins has allowed the study of synaptic vesicle recycling in real time. Two such probes, synaptopHluorin and sypHy, have been imaged at synapses of hippocampal neurons in culture. The combination of these reporters with techniques for molecular interference, such as RNAi allows for the study of molecules involved in synaptic vesicle recycling. Here the authors describe methods for the culture and transfection of hippocampal neurons, imaging of pHluorin-based probes at synapses and analysis of pHluorin signals down to the resolution of individual synaptic vesicles.
Collapse
Affiliation(s)
- Stephen J Royle
- School of Biomedical Sciences, University of Liverpool, Liverpool, UK
| | | | | | | | | |
Collapse
|
39
|
He L, Wu LG. The debate on the kiss-and-run fusion at synapses. Trends Neurosci 2007; 30:447-55. [PMID: 17765328 DOI: 10.1016/j.tins.2007.06.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 06/08/2007] [Accepted: 06/13/2007] [Indexed: 11/17/2022]
Abstract
It has long been proposed that following vesicle fusion, a small pore might open and close rapidly without full dilation. Such 'kiss-and-run' vesicle fusion can in principle result in rapid vesicle recycling and influence the size and the kinetics of the resulting synaptic current. However, the existence of kiss-and-run remains highly controversial, as revealed by recent imaging and electrophysiological studies at several synapses, including hippocampal synapses, neuromuscular junctions and retinal bipolar synapses. Only a minor fraction of fusion events has been shown to be kiss-and-run, as determined using cell-attached capacitance recordings in endocrine cells, pituitary nerve terminals and calyx-type synapses. Further work is needed to determine whether kiss-and-run is a major mode of fusion and has a major role in controlling synaptic strength at synapses.
Collapse
Affiliation(s)
- Liming He
- National Institute of Neurological Disorders and Stroke, 35 Convent Drive, Bldg 35, Rm. 2B-1012, Bethesda, MD 20892, USA
| | | |
Collapse
|
40
|
Stella SL, Hu WD, Vila A, Brecha NC. Adenosine inhibits voltage-dependent Ca2+ influx in cone photoreceptor terminals of the tiger salamander retina. J Neurosci Res 2007; 85:1126-37. [PMID: 17304584 PMCID: PMC3737423 DOI: 10.1002/jnr.21210] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Endogenous adenosine has already been shown to inhibit transmitter release from the rod synapse by suppressing Ca(2+) influx through voltage-gated Ca(2+) channels. However, it is not clear how adenosine modulates the cone synapse. Cone photoreceptors, like rod photoreceptors, also possess L-type Ca(2+) channels that regulate the release of L-glutamate. To assess the impact of adenosine on Ca(2+) influx though voltage-gated Ca(2+) channels in cone terminals, whole-cell perforated-patch clamp recording and Ca(2+) imaging with fluo-4 were used on isolated cones and salamander retinal slices. Synaptic markers (VAMP and piccolo) and activity-dependent dye labeling revealed that tiger salamander cone terminals contain a broad, vesicle-filled cytoplasmic extension at the base of the somatic compartment, which is unlike rod terminals that contain one or more thin axons, each terminating in a large bulbous synaptic terminal. The spatiotemporal Ca(2+) responses of the cone terminals do not differ significantly from the Ca(2+) responses of the soma or inner segment like that observed in rods. Whole-cell recording of cone I(Ca) and Ca(2+) imaging of synaptic terminals in cones demonstrate that adenosine inhibited both I(Ca) and the depolarization-evoked Ca(2+) increase in cone terminals in a dose-dependent manner from 1 to 50 muM. These results indicate that, as in rods, adenosine's ability to suppress voltage-dependent Ca(2+) channels at the cone synapse will limit the amount of L-glutamate released. Therefore, adenosine has an inhibitory effect on L-glutamate release at the first synapse, which likely favors elevated adenosine levels in the dark or during dark-adapted conditions.
Collapse
Affiliation(s)
- Salvatore L Stella
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
41
|
Jensen V, Walaas SI, Hilfiker S, Ruiz A, Hvalby Ø. A delayed response enhancement during hippocampal presynaptic plasticity in mice. J Physiol 2007; 583:129-43. [PMID: 17569738 PMCID: PMC2277251 DOI: 10.1113/jphysiol.2007.131300] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
High frequency afferent stimulation of chemical synapses often induces short-term increases in synaptic efficacy, due to increased release probability and/or increased supply of readily releasable synaptic vesicles. This may be followed by synaptic depression, often caused by vesicle depletion. We here describe an additional, novel type of delayed and transient response enhancement phase which occurred during prolonged stimulation at 5-20 Hz frequency of excitatory glutamatergic synapses in slices from the adult mouse CA1 hippocampal region. This second enhancement phase, which was most clearly defined at physiological temperatures and essentially absent at 24 degrees C, was dependent on the presence of F-actin filaments and synapsins I and/or II, and could not be ascribed to changes in presynaptic action potentials, inhibitory neurotransmission or glutamate receptor desensitization. Time course studies showed that the delayed response phase interrupted the synaptic decay 3-4 s after stimulus train initiation and continued, when examined at 5-10 Hz frequencies, for approximately 75 stimuli before decay. The novel response enhancement, probably deriving from a restricted pool of synaptic vesicles, may allow maintenance of synaptic efficacy during prolonged periods of excitatory synaptic activity.
Collapse
Affiliation(s)
- Vidar Jensen
- Molecular Neurobiology Research Group (MONERG), PO Box 1104, Faculty of Medicine, University of Oslo, N-0317 Blindern, Oslo, Norway
| | | | | | | | | |
Collapse
|
42
|
De Proost I, Pintelon I, Brouns I, Timmermans JP, Adriaensen D. Selective visualisation of sensory receptors in the smooth muscle layer of ex-vivo airway whole-mounts by styryl pyridinium dyes. Cell Tissue Res 2007; 329:421-31. [PMID: 17522895 DOI: 10.1007/s00441-007-0431-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 04/25/2007] [Indexed: 02/01/2023]
Abstract
Recently, we established the location, morphology and neurochemical coding of vagal smooth-muscle-associated airway receptors (SMARs) in rat lungs. These receptors were characterised as branching laminar terminals that originated from myelinated nerve fibres and were intercalated between airway smooth-muscle bundles. To allow the direct physiological examination of these receptors, the present investigation aimed at visualising SMARs in airway whole-mounts of rat and mouse lungs ex vivo. Short incubation with various styryl pyridinium dyes (AM1-43, FM2-10, FM4-64 or 4-Di-2-ASP) gave a highly selective fluorescent visualisation of both laminar nerve terminals and myelinated fibres from which they originated throughout the intrapulmonary airway tree in mouse and in rat. The reliable and specific labelling of SMARs ex vivo with these lipophilic membrane dyes was confirmed via immunostaining for protein gene-product 9.5 and vesicular glutamate transporters. Similar to the intrapulmonary location of NEBs, these SMARs appeared to be even more explicitly located near airway bifurcations. Both the trachealis muscle and the smooth-muscle bundles of extrapulmonary bronchi were also shown to contain laminar nerve terminals that were morphologically similar to the SMARs reported in the intrapulmonary airways. Thus, this study provides an in-vitro model enabling, for the first time, the fast and reliable visualisation of SMARs and the myelinated nerve fibres from which they originate in airway whole-mount preparations ex vivo. As such, this model opens up further perspectives and creates a valid basis for direct physiological measurement and manipulation of the individually identified airway receptors.
Collapse
Affiliation(s)
- Ian De Proost
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Groenenborgerlaan 171, BE-2020 Antwerp, Belgium
| | | | | | | | | |
Collapse
|
43
|
Abstract
FM dyes have been used to label and then monitor synaptic vesicles, secretory granules and other endocytic structures in a variety of preparations. Here, we describe the general procedure for using FM dyes to study endosomal trafficking in general, and synaptic vesicle recycling in particular. The dye, dissolved in normal saline solution, is added to a chamber containing the preparation to be labeled. Stimulation evokes exocytosis, and compensatory endocytosis that follows traps FM dye inside the retrieved vesicles. The extracellular dye is then washed from the chamber, and labeled endocytic structures are examined with a fluorescence microscope. Fluorescence intensity provides a direct measure of the labeled vesicle number, a good measure of the amount of exocytosis. If the preparation is stimulated again, without dye in the chamber, dimming of the preparation provides a measure of exocytosis of labeled vesicles. With a synaptic preparation on hand, this protocol requires 1 day.
Collapse
Affiliation(s)
- Michael A Gaffield
- Neuroscience Program, University of Colorado Medical School, Aurora, Colorado 80045, USA
| | | |
Collapse
|
44
|
Drew LJ, Wood JN. FM1-43 is a permeant blocker of mechanosensitive ion channels in sensory neurons and inhibits behavioural responses to mechanical stimuli. Mol Pain 2007; 3:1. [PMID: 17207285 PMCID: PMC1779769 DOI: 10.1186/1744-8069-3-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Accepted: 01/06/2007] [Indexed: 01/31/2023] Open
Abstract
The molecular identity and pharmacological properties of mechanically gated ion channels in sensory neurons are poorly understood. We show that FM1-43, a styryl dye used to fluorescently label cell membranes, permeates mechanosensitive ion channels in cultured dorsal root ganglion neurons, resulting in blockade of three previously defined subtypes of mechanically activated currents. Blockade and dye uptake is voltage dependent and regulated by external Ca2+. The structurally related larger dye FM3-25 inhibited mechanically activated currents to a lesser degree and did not permeate the channels. In vivo, FMI-43 decreases pain sensitivity in the Randall-Selitto test and increases the withdrawal threshold from von Frey hairs, together suggesting that the channels expressed at the cell body in culture mediate mechanosensation in the intact animal. These data give further insight into the mechanosensitive ion channels expressed by somatosensory neurons and suggest FM dyes are an interesting tool for studying them.
Collapse
Affiliation(s)
- Liam J Drew
- Dept. of Biology, UCL, Gower Street, London, WC1E 6BT, UK
- Dept. of Physiology and Cellular Biophysics, Columbia University, 630, W168th St, New York, NY10032, USA
| | - John N Wood
- Dept. of Biology, UCL, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
45
|
Tyler WJ, Zhang XL, Hartman K, Winterer J, Muller W, Stanton PK, Pozzo-Miller L. BDNF increases release probability and the size of a rapidly recycling vesicle pool within rat hippocampal excitatory synapses. J Physiol 2006; 574:787-803. [PMID: 16709633 PMCID: PMC1817733 DOI: 10.1113/jphysiol.2006.111310] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Exerting its actions pre-, post- and peri-synaptically, brain-derived neurotrophic factor (BDNF) is one of the most potent modulators of hippocampal synaptic function. Here, we examined the effects of BDNF on a rapidly recycling pool (RRP) of vesicles within excitatory synapses. First, we estimated vesicular release in hippocampal cultures by performing FM4-64 imaging in terminals impinging on enhanced green fluorescent protein (eGFP)-labelled dendritic spines - a hallmark of excitatory synapses. Consistent with a modulation of the RRP, BDNF increased the evoked destaining rate of FM4-64 only during the initial phase of field stimulation. Multiphoton microscopy in acute hippocampal slices confirmed these observations by selectively imaging the RRP, which was loaded with FM1-43 by hyperosmotic shock. Slices exposed to BDNF showed an increase in the evoked and spontaneous rates of FM1-43 destaining from terminals in CA1 stratum radiatum, mostly representing excitatory terminals of Schaffer collaterals. Variance-mean analysis of evoked EPSCs in CA1 pyramidal neurons further confirmed that release probability is increased in BDNF-treated slices, without changes in the number of independent release sites or average postsynaptic quantal amplitude. Because BDNF was absent during dye loading, imaging, destaining and whole-cell recordings, these results demonstrate that BDNF induces a long-lasting enhancement in the probability of transmitter release at hippocampal excitatory synapses by modulating the RRP. Since the endogenous BDNF scavenger TrkB-IgG prevented the enhancement of FM1-43 destaining rate caused by induction of long-term potentiation in acute hippocampal slices, the modulation of a rapidly recycling vesicle pool may underlie the role of BDNF in hippocampal long-term synaptic plasticity.
Collapse
Affiliation(s)
- William J Tyler
- Department of Neurobiology, SHEL-1002, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294-2182, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Schweizer FE, Ryan TA. The synaptic vesicle: cycle of exocytosis and endocytosis. Curr Opin Neurobiol 2006; 16:298-304. [PMID: 16707259 DOI: 10.1016/j.conb.2006.05.006] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 05/05/2006] [Indexed: 11/30/2022]
Abstract
Synaptic vesicles are clustered at the presynaptic terminal where they fuse and recycle in response to stimulation. Vesicles appear to be sorted into pools, but we do not yet understand how physiologically defined pools relate to morphological pools. The advent of dynamic imaging approaches has led to an appreciation of the regulation of vesicle mobility. Newly endocytosed vesicles are highly mobile but appear to become transiently trapped as they re-enter the recycling pool. Recent experiments indicate that endocytosis might have a constant rate, but limited capacity. How endocytosis is linked to exocytosis remains unclear, although calcium emerges as an important player.
Collapse
Affiliation(s)
- Felix E Schweizer
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 650 Charles E. Young Drive South, CHS 63-323, Los Angeles, CA 90095-1763, USA.
| | | |
Collapse
|
47
|
Micheva KD, Smith SJ. Strong effects of subphysiological temperature on the function and plasticity of mammalian presynaptic terminals. J Neurosci 2006; 25:7481-8. [PMID: 16107635 PMCID: PMC6725406 DOI: 10.1523/jneurosci.1801-05.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Most cellular processes are known to be strongly temperature dependent. Nevertheless, a large fraction of studies of mammalian synaptic function have been and are performed near room temperature (i.e., at least 10 degrees C below physiological temperature). Here, we examined the effects of temperature on presynaptic function in primary cultures of rat hippocampal neurons. FM dyes, VAMP (vesicle-associated membrane protein)-GFP (green fluorescent protein) transfection, and HRP uptake were used to quantify various aspects of synaptic vesicle recycling. Our results show that there are very substantial differences in synaptic vesicle recycling at physiological temperature as opposed to the common, lower experimental temperatures. At 37 degrees C, compared with 23 degrees C, the speed of both exocytosis and endocytosis was higher. The size of the recycling vesicle pool (in both number of vesicles and spatial extent) was twofold larger at 37 degrees C. In addition, although repeated 10 Hz electrical stimulation caused an NMDA receptor-dependent enlargement (averaging 170%) of the measurable recycling vesicle pool at 23 degrees C, the same stimulus repetition had no effect at 37 degrees C. These results show that it is potentially misleading to extend conclusions drawn about vesicle function or presynaptic plasticity at lowered experimental temperature to physiological conditions and that much new experimental work at the higher physiological temperature range will be needed to understand the true parameters of presynaptic functions.
Collapse
Affiliation(s)
- Kristina D Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA.
| | | |
Collapse
|
48
|
Evans GJO, Cousin MA. Tyrosine phosphorylation of synaptophysin in synaptic vesicle recycling. Biochem Soc Trans 2006; 33:1350-3. [PMID: 16246116 PMCID: PMC2077014 DOI: 10.1042/bst20051350] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The integral SV (synaptic vesicle) protein synaptophysin was one of the first nerve terminal proteins identified. However its role, if any, in the SV life cycle remains undetermined. One of the most prominent features of synaptophysin is that its cytoplasmic C-terminus largely consists of pentapeptide repeats initiated by a tyrosine residue. Synaptophysin is heavily phosphorylated by tyrosine kinases in the nerve terminal, suggesting that this phosphorylation is central to its function. This review will cover the evidence for tyrosine phosphorylation of synaptophysin and how this phosphorylation may control its function in the SV life cycle.
Collapse
Affiliation(s)
- G J O Evans
- Membrane Biology Group, Centre for Integrative Physiology, George Square, University of Edinburgh, Edinburgh EH8 9XD, UK
| | | |
Collapse
|
49
|
Abstract
The integral SV (synaptic vesicle) protein synaptophysin was one of the first nerve terminal proteins identified. However its role, if any, in the SV life cycle remains undetermined. One of the most prominent features of synaptophysin is that its cytoplasmic C-terminus largely consists of pentapeptide repeats initiated by a tyrosine residue. Synaptophysin is heavily phosphorylated by tyrosine kinases in the nerve terminal, suggesting that this phosphorylation is central to its function. This review will cover the evidence for tyrosine phosphorylation of synaptophysin and how this phosphorylation may control its function in the SV life cycle.
Collapse
|
50
|
Choi SY, Sheng Z, Kramer RH. Imaging light-modulated release of synaptic vesicles in the intact retina: retinal physiology at the dawn of the post-electrode era. Vision Res 2005; 45:3487-95. [PMID: 16185743 DOI: 10.1016/j.visres.2005.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2005] [Revised: 08/08/2005] [Accepted: 08/11/2005] [Indexed: 11/19/2022]
Abstract
Here, we illustrate an optical method for directly measuring the light-regulated synaptic output of neurons in the retina. The method allows simultaneous recording from many retinal neurons in intact flat-mount preparations of the vertebrate retina. These recordings depend on the use of FM1-43, an activity-dependent fluorescent dye that selectively labels synaptic vesicles. Release of the dye, which occurs upon vesicle exocytosis, is detected with 2-photon microscopy. This utilizes an infrared laser to trigger fluorescence excitation of the dye, while minimally perturbing retinal activity by activating phototransduction in rods and cones. Using this approach, one can measure activity of single neurons in the intact retinal network and populations of neurons in different layers of the retina, providing a new way to examine the function of retinal synapses and how visual information is processed.
Collapse
Affiliation(s)
- Sue-Yeon Choi
- Department of Molecular and Cell Biology, University of California, Berkeley, USA
| | | | | |
Collapse
|