151
|
Salter EW, Sunstrum JK, Matovic S, Inoue W. Chronic stress dampens excitatory synaptic gain in the paraventricular nucleus of the hypothalamus. J Physiol 2018; 596:4157-4172. [PMID: 29901836 DOI: 10.1113/jp275669] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Glutamatergic synaptic inputs to corticotrophin-releasing hormone (CRH) secreting neurons in the paraventricular nucleus of the hypothalamus (PVN) are required for stress-induced activation of the hypothalamic-pituitary-adrenal (HPA) axis. These synapses also undergo stress-induced plasticity, thereby influencing HPA axis stress adaptation. By using patch clamp electrophysiology, we show that, in adult non-stressed mice, action potentials at these glutamatergic afferents elicit multiquantal transmission to the postsynaptic PVN-CRH neurons (i.e. synaptic multiplicity). Mechanistically, synaptic multiplicity results from multivesicular release at common synaptic sites, which is facilitated upon elevation of release probability, effectively increasing the upper limit of the dynamic range of synaptic transmission. Following chronic variable stress, functional PVN glutamate synapse number increases, although its synaptic multiplicity paradoxically decreases. These two contrasting synaptic changes can, respectively, increase the baseline excitatory drive while also limiting the capacity for potentiation, and may preferentially increase the baseline excitatory drive onto PVN-CRH neurons. ABSTRACT The activation of the hypothalamic-pituitary-adrenal (HPA) axis relies on excitation of neuroendocrine neurons in the paraventricular nucleus of the hypothalamus (PVN) that secrete corticotrophin-releasing hormone (CRH). Afferent glutamate synapses onto these PVN-CRH neurons convey critical excitatory inputs during stress, and also undergo stress-induced plasticity, highlighting their roles in both stress activation and adaptation of the HPA axis. In the present study, using whole-cell patch clamp recordings from PVN-CRH neurons in brain slices from adult mice, we found that the amplitude of action potential-dependent spontaneous EPSCs (sEPSCs) was larger than that of action potential independent miniature EPSCs (mEPSCs), suggesting that action potentials at individual axons recruited multiquantal transmission onto the same postsynaptic neurons (i.e. synaptic multiplicity). The large, putative multiquantal sEPSCs had fast rise times similar to mEPSCs, and were abolished by replacing extracellular Ca2+ with Sr2+ , indicating Ca2+ -dependent synchronous release of multiple vesicles. Application of a low affinity, fast dissociating competitive AMPA receptor antagonist γ-d-glutamylglycine revealed that synaptic multiplicity resulted from multivesicular release targeting a common population of postsynaptic receptors. High-frequency afferent stimulation facilitated synaptic multiplicity, effectively increasing the upper limit of the dynamic range of synaptic transmission. Finally, we found that chronic variable stress (CVS), a stress model known to cause basal HPA axis hyperactivity, increased sEPSCs frequency but paradoxically decreased synaptic multiplicity. These results suggest that the CVS-induced synaptic changes may elevate the baseline excitatory drive at the same time as limiting the capacity for potentiation, and may contribute to the basal HPA axis hyperactivity.
Collapse
Affiliation(s)
- Eric W Salter
- Neuroscience Program, University of Western Ontario, London, Ontario, Canada
| | - Julia K Sunstrum
- Neuroscience Program, University of Western Ontario, London, Ontario, Canada
| | - Sara Matovic
- Neuroscience Program, University of Western Ontario, London, Ontario, Canada
| | - Wataru Inoue
- Neuroscience Program, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,Robarts Research Institute, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
152
|
Delgado JY, Selvin PR. A Revised View on the Role of Surface AMPAR Mobility in Tuning Synaptic Transmission: Limitations, Tools, and Alternative Views. Front Synaptic Neurosci 2018; 10:21. [PMID: 30079019 PMCID: PMC6062754 DOI: 10.3389/fnsyn.2018.00021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022] Open
Abstract
Calcium dynamics in presynaptic terminals regulate the response dynamics of most central excitatory synapses. However, this dogma has been challenged by the hypothesis that mobility of the postsynaptic alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid subtype glutamate receptors (AMPAR) plays a role in tuning fast excitatory synaptic transmission. In this review, we reevaluate the factors regulating postsynaptic AMPAR mobility, reassess the modeling parameters, analyze the experimental tools, and end by providing alternative ideas stemming from recent results. In particular, newer methods of labeling AMPARs with small fluorophores in live neurons, combined with super-resolution microscopy and sub-second dynamics, lends support to the idea that AMPARs are primarily within the synapse, are greatly constrained, and have much slower mobility than previously thought. We discuss new experiments which may be necessary to readdress the role of postsynaptic AMPAR mobility in tuning fast excitatory synaptic transmission.
Collapse
Affiliation(s)
- Jary Y Delgado
- Department of Neurobiology, The University of Chicago, Chicago, IL, United States
| | - Paul R Selvin
- Department of Physics, Biophysics, and the Center for the Physics of Living Cells, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
153
|
Akbergenova Y, Cunningham KL, Zhang YV, Weiss S, Littleton JT. Characterization of developmental and molecular factors underlying release heterogeneity at Drosophila synapses. eLife 2018; 7:38268. [PMID: 29989549 PMCID: PMC6075867 DOI: 10.7554/elife.38268] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/30/2018] [Indexed: 12/14/2022] Open
Abstract
Neurons communicate through neurotransmitter release at specialized synaptic regions known as active zones (AZs). Using biosensors to visualize single synaptic vesicle fusion events at Drosophila neuromuscular junctions, we analyzed the developmental and molecular determinants of release probability (Pr) for a defined connection with ~300 AZs. Pr was heterogeneous but represented a stable feature of each AZ. Pr remained stable during high frequency stimulation and retained heterogeneity in mutants lacking the Ca2+ sensor Synaptotagmin 1. Pr correlated with both presynaptic Ca2+ channel abundance and Ca2+ influx at individual release sites. Pr heterogeneity also correlated with glutamate receptor abundance, with high Pr connections developing receptor subtype segregation. Intravital imaging throughout development revealed that AZs acquire high Pr during a multi-day maturation period, with Pr heterogeneity largely reflecting AZ age. The rate of synapse maturation was activity-dependent, as both increases and decreases in neuronal activity modulated glutamate receptor field size and segregation. To send a message to its neighbor, a neuron releases chemicals called neurotransmitters into the gap – or synapse – between them. The neurotransmitter molecules bind to proteins on the receiver neuron called receptors. But what causes the sender neuron to release neurotransmitter in the first place? The process starts when an electrical impulse called an action potential arrives at the sender cell. Its arrival causes channels in the membrane of the sender neuron to open, so that calcium ions flood into the cell. The calcium ions interact with packages of neurotransmitter molecules, known as synaptic vesicles. This causes some of the vesicles to empty their contents into the synapse. But this process is not particularly reliable. Only a small fraction of action potentials cause vesicles to fuse with the synaptic membrane. How likely this is to occur varies greatly between neurons, and even between synapses formed by the same neuron. Synapses that are likely to release neurotransmitter are said to be strong. They are good at passing messages from the sender neuron to the receiver. Synapses with a low probability of release are said to be weak. But what exactly differs between strong and weak synapses? Akbergenova et al. studied synapses between motor neurons and muscle cells in the fruit fly Drosophila. Each motor neuron forms several hundred synapses. Some of these synapses are 50 times more likely to release neurotransmitter than others. Using calcium imaging and genetics, Akbergenova et al. showed that sender cells at strong synapses have more calcium channels than sender cells at weak synapses. The subtypes and arrangement of receptor proteins also differ between the receiver neurons of strong versus weak synapses. Finally, studies in larvae revealed that newly formed synapses all start out weak and then gradually become stronger. How fast this strengthening occurs depends on how active the neuron at the synapse is. This study has shown, in unprecedented detail, key molecular factors that make some fruit fly synapses more likely to release neurotransmitter than others. Many proteins at synapses of mammals resemble those at fruit fly synapses. This means that similar factors may also explain differences in synaptic strength in the mammalian brain. Changes in the strength of synapses underlie the ability to learn. Furthermore, many neurological and psychiatric disorders result from disruption of synapses. Understanding the molecular basis of synapses will thus provide clues to the origins of certain brain diseases.
Collapse
Affiliation(s)
- Yulia Akbergenova
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Karen L Cunningham
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Yao V Zhang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Shirley Weiss
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
154
|
Grillo FW, Neves G, Walker A, Vizcay-Barrena G, Fleck RA, Branco T, Burrone J. A Distance-Dependent Distribution of Presynaptic Boutons Tunes Frequency-Dependent Dendritic Integration. Neuron 2018; 99:275-282.e3. [PMID: 29983327 PMCID: PMC6078905 DOI: 10.1016/j.neuron.2018.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 03/23/2018] [Accepted: 06/08/2018] [Indexed: 11/28/2022]
Abstract
How presynaptic inputs and neurotransmitter release dynamics are distributed along a dendritic tree is not well established. Here, we show that presynaptic boutons that form onto basal dendrites of CA1 pyramidal neurons display a decrease in active zone (AZ) size with distance from the soma, resulting in a distance-dependent increase in short-term facilitation. Our findings suggest that the spatial distribution of short-term facilitation serves to compensate for the electrotonic attenuation of subthreshold distal inputs during repeated stimulation and fine-tunes the preferred input frequency of dendritic domains. Presynaptic inputs decrease in size with distance along CA1 basal dendrites Release probability decreases with distance along basal dendrites Short-term facilitation increases with distance along basal dendrites Increased synaptic facilitation offsets passive decay and boosts supralinear events
Collapse
Affiliation(s)
- Federico W Grillo
- Centre for Developmental Neurobiology, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK
| | - Guilherme Neves
- Centre for Developmental Neurobiology, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK
| | - Alison Walker
- Centre for Developmental Neurobiology, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK
| | - Roland A Fleck
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK
| | - Tiago Branco
- The Sainsbury Wellcome Centre, University College London, 25 Howland Street, London, W1T 4JG, UK
| | - Juan Burrone
- Centre for Developmental Neurobiology, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, Kings College London, New Hunts House, Guys Hospital Campus, London, SE1 1UL, UK.
| |
Collapse
|
155
|
Schroeder A, Vanderlinden J, Vints K, Ribeiro LF, Vennekens KM, Gounko NV, Wierda KD, de Wit J. A Modular Organization of LRR Protein-Mediated Synaptic Adhesion Defines Synapse Identity. Neuron 2018; 99:329-344.e7. [PMID: 29983322 DOI: 10.1016/j.neuron.2018.06.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/04/2018] [Accepted: 06/14/2018] [Indexed: 10/28/2022]
Abstract
Pyramidal neurons express rich repertoires of leucine-rich repeat (LRR)-containing adhesion molecules with similar synaptogenic activity in culture. The in vivo relevance of this molecular diversity is unclear. We show that hippocampal CA1 pyramidal neurons express multiple synaptogenic LRR proteins that differentially distribute to the major excitatory inputs on their apical dendrites. At Schaffer collateral (SC) inputs, FLRT2, LRRTM1, and Slitrk1 are postsynaptically localized and differentially regulate synaptic structure and function. FLRT2 controls spine density, whereas LRRTM1 and Slitrk1 exert opposing effects on synaptic vesicle distribution at the active zone. All LRR proteins differentially affect synaptic transmission, and their combinatorial loss results in a cumulative phenotype. At temporoammonic (TA) inputs, LRRTM1 is absent; FLRT2 similarly controls functional synapse number, whereas Slitrk1 function diverges to regulate postsynaptic AMPA receptor density. Thus, LRR proteins differentially control synaptic architecture and function and act in input-specific combinations and a context-dependent manner to specify synaptic properties.
Collapse
Affiliation(s)
- Anna Schroeder
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Katlijn Vints
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium; Electron Microscopy Platform & VIB BioImaging Core, Herestraat 49, 3000 Leuven, Belgium
| | - Luís F Ribeiro
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Natalia V Gounko
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium; Electron Microscopy Platform & VIB BioImaging Core, Herestraat 49, 3000 Leuven, Belgium
| | - Keimpe D Wierda
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
156
|
Kulik Á, Booker SA, Vida I. Differential distribution and function of GABABRs in somato-dendritic and axonal compartments of principal cells and interneurons in cortical circuits. Neuropharmacology 2018; 136:80-91. [DOI: 10.1016/j.neuropharm.2017.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/24/2022]
|
157
|
Jiang N, Kim HJ, Chozinski TJ, Azpurua JE, Eaton BA, Vaughan JC, Parrish JZ. Superresolution imaging of Drosophila tissues using expansion microscopy. Mol Biol Cell 2018; 29:1413-1421. [PMID: 29688792 PMCID: PMC6014096 DOI: 10.1091/mbc.e17-10-0583] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The limited resolving power of conventional diffraction-limited microscopy hinders analysis of small, densely packed structural elements in cells. Expansion microscopy (ExM) provides an elegant solution to this problem, allowing for increased resolution with standard microscopes via physical expansion of the specimen in a swellable polymer hydrogel. Here, we apply, validate, and optimize ExM protocols that enable the study of Drosophila embryos, larval brains, and larval and adult body walls. We achieve a lateral resolution of ∼70 nm in Drosophila tissues using a standard confocal microscope, and we use ExM to analyze fine intracellular structures and intercellular interactions. First, we find that ExM reveals features of presynaptic active zone (AZ) structure that are observable with other superresolution imaging techniques but not with standard confocal microscopy. We further show that synapses known to exhibit age-dependent changes in activity also exhibit age-dependent changes in AZ structure. Finally, we use the significantly improved axial resolution of ExM to show that dendrites of somatosensory neurons are inserted into epithelial cells at a higher frequency than previously reported in confocal microscopy studies. Altogether, our study provides a foundation for the application of ExM to Drosophila tissues and underscores the importance of tissue-specific optimization of ExM procedures.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Biology, University of Washington, Seattle, WA 98195
| | - Hyeon-Jin Kim
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Tyler J Chozinski
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Jorge E Azpurua
- Department of Physiology, University of Texas Health Sciences Center, San Antonio, TX 78229
| | - Benjamin A Eaton
- Department of Physiology, University of Texas Health Sciences Center, San Antonio, TX 78229
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA 98195
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA 98195
| |
Collapse
|
158
|
Theis AK, Rózsa B, Katona G, Schmitz D, Johenning FW. Voltage Gated Calcium Channel Activation by Backpropagating Action Potentials Downregulates NMDAR Function. Front Cell Neurosci 2018; 12:109. [PMID: 29755321 PMCID: PMC5932410 DOI: 10.3389/fncel.2018.00109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/03/2018] [Indexed: 12/20/2022] Open
Abstract
The majority of excitatory synapses are located on dendritic spines of cortical glutamatergic neurons. In spines, compartmentalized Ca2+ signals transduce electrical activity into specific long-term biochemical and structural changes. Action potentials (APs) propagate back into the dendritic tree and activate voltage gated Ca2+ channels (VGCCs). For spines, this global mode of spine Ca2+ signaling is a direct biochemical feedback of suprathreshold neuronal activity. We previously demonstrated that backpropagating action potentials (bAPs) result in long-term enhancement of spine VGCCs. This activity-dependent VGCC plasticity results in a large interspine variability of VGCC Ca2+ influx. Here, we investigate how spine VGCCs affect glutamatergic synaptic transmission. We combined electrophysiology, two-photon Ca2+ imaging and two-photon glutamate uncaging in acute brain slices from rats. T- and R-type VGCCs were the dominant depolarization-associated Ca2+conductances in dendritic spines of excitatory layer 2 neurons and do not affect synaptic excitatory postsynaptic potentials (EPSPs) measured at the soma. Using two-photon glutamate uncaging, we compared the properties of glutamatergic synapses of single spines that express different levels of VGCCs. While VGCCs contributed to EPSP mediated Ca2+ influx, the amount of EPSP mediated Ca2+ influx is not determined by spine VGCC expression. On a longer timescale, the activation of VGCCs by bAP bursts results in downregulation of spine NMDAR function.
Collapse
Affiliation(s)
- Anne-Kathrin Theis
- Neuroscience Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Balázs Rózsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.,Faculty of Information Technology and Bionics, Pázmány Péter University, Budapest, Hungary
| | - Gergely Katona
- Faculty of Information Technology and Bionics, Pázmány Péter University, Budapest, Hungary
| | - Dietmar Schmitz
- Neuroscience Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany.,Einstein Center for Neuroscience, Berlin, Germany.,Bernstein Center for Computational Neuroscience, Berlin, Germany.,Cluster of Excellence "Neurocure", Berlin, Germany.,DZNE-German Center for Neurodegenerative Disease, Berlin, Germany
| | - Friedrich W Johenning
- Neuroscience Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany.,Einstein Center for Neuroscience, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
159
|
Li X, Goel P, Chen C, Angajala V, Chen X, Dickman DK. Synapse-specific and compartmentalized expression of presynaptic homeostatic potentiation. eLife 2018; 7:34338. [PMID: 29620520 PMCID: PMC5927770 DOI: 10.7554/elife.34338] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/04/2018] [Indexed: 01/23/2023] Open
Abstract
Postsynaptic compartments can be specifically modulated during various forms of synaptic plasticity, but it is unclear whether this precision is shared at presynaptic terminals. Presynaptic homeostatic plasticity (PHP) stabilizes neurotransmission at the Drosophila neuromuscular junction, where a retrograde enhancement of presynaptic neurotransmitter release compensates for diminished postsynaptic receptor functionality. To test the specificity of PHP induction and expression, we have developed a genetic manipulation to reduce postsynaptic receptor expression at one of the two muscles innervated by a single motor neuron. We find that PHP can be induced and expressed at a subset of synapses, over both acute and chronic time scales, without influencing transmission at adjacent release sites. Further, homeostatic modulations to CaMKII, vesicle pools, and functional release sites are compartmentalized and do not spread to neighboring pre- or post-synaptic structures. Thus, both PHP induction and expression mechanisms are locally transmitted and restricted to specific synaptic compartments. Everything we think and do is the result of communication between neurons. This communication takes place at junctions called synapses. When two nerve cells or neurons communicate at a synapse, the output terminal of the first cell releases a chemical called a neurotransmitter. This binds to receiver proteins, or receptors, on the second cell. When this communication is interrupted, synapses can adapt to maintain a stable dialogue between them. This can occur in two ways. Either the first neuron starts to release more neurotransmitter from its output terminal, or the second neuron produces extra receptors with which to detect the neurotransmitter. But how specific are these changes? The brain contains far more synapses than neurons because each neuron can form synapses with many other cells. Can a neuron adjust how much of the neurotransmitter it releases at some of its synapses while leaving the others unchanged? Li et al. have now addressed this question by studying a special type of synapse that forms between neurons and muscles, known as a neuromuscular junction. At one particular neuromuscular junction in fruit flies, a single neuron splits into two output terminals, each of which forms a synapse with a different muscle. Li et al. show that when the number of neurotransmitter receptors in one of the muscles is artificially reduced, the associated output terminal compensates by increasing its neurotransmitter release. By contrast, the other output terminal remains unaffected. This suggests that a neuron can induce remarkably specific changes in a subset of its synapses. This discovery paves the way towards identifying the smallest possible unit of change that can occur in the neurons’ ability to communicate. This unit may in turn be the smallest change that can support learning. Such knowledge will help us understand how the nervous system processes and stabilizes information transfer, both in health and after injury or disease.
Collapse
Affiliation(s)
- Xiling Li
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Neuroscience Graduate Program, University of Southern California, California, United States
| | - Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, United States.,Graduate Program in Molecular and Computational Biology, University of Southern California, California, United States
| | - Catherine Chen
- Department of Neurobiology, University of Southern California, Los Angeles, United States
| | | | - Xun Chen
- Neuroscience Graduate Program, University of Southern California, California, United States
| | | |
Collapse
|
160
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
161
|
Variations in Ca 2+ Influx Can Alter Chelator-Based Estimates of Ca 2+ Channel-Synaptic Vesicle Coupling Distance. J Neurosci 2018; 38:3971-3987. [PMID: 29563180 DOI: 10.1523/jneurosci.2061-17.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 02/23/2018] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
The timing and probability of synaptic vesicle fusion from presynaptic terminals is governed by the distance between voltage-gated Ca2+ channels (VGCCs) and Ca2+ sensors for exocytosis. This VGCC-sensor coupling distance can be determined from the fractional block of vesicular release by exogenous Ca2+ chelators, which depends on biophysical factors that have not been thoroughly explored. Using numerical simulations of Ca2+ reaction and diffusion, as well as vesicular release, we examined the contributions of conductance, density, and open duration of VGCCs, and the influence of endogenous Ca2+ buffers on the inhibition of exocytosis by EGTA. We found that estimates of coupling distance are critically influenced by the duration and amplitude of Ca2+ influx at active zones, but relatively insensitive to variations of mobile endogenous buffer. High concentrations of EGTA strongly inhibit vesicular release in close proximity (20-30 nm) to VGCCs if the flux duration is brief, but have little influence for longer flux durations that saturate the Ca2+ sensor. Therefore, the diversity in presynaptic action potential duration is sufficient to alter EGTA inhibition, resulting in errors potentially as large as 300% if Ca2+ entry durations are not considered when estimating VGCC-sensor coupling distances.SIGNIFICANT STATEMENT The coupling distance between voltage-gated Ca2+ channels and Ca2+ sensors for exocytosis critically determines the timing and probability of neurotransmitter release. Perfusion of presynaptic terminals with the exogenous Ca2+ chelator EGTA has been widely used for both qualitative and quantitative estimates of this distance. However, other presynaptic terminal parameters such as the amplitude and duration of Ca2+ entry can also influence EGTA inhibition of exocytosis, thus confounding conclusions based on EGTA alone. Here, we performed reaction-diffusion simulations of Ca2+-driven synaptic vesicle fusion, which delineate the critical parameters influencing an accurate prediction of coupling distance. Our study provides guidelines for characterizing and understanding how variability in coupling distance across chemical synapses could be estimated accurately.
Collapse
|
162
|
Lou X. Sensing Exocytosis and Triggering Endocytosis at Synapses: Synaptic Vesicle Exocytosis-Endocytosis Coupling. Front Cell Neurosci 2018; 12:66. [PMID: 29593500 PMCID: PMC5861208 DOI: 10.3389/fncel.2018.00066] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/29/2022] Open
Abstract
The intact synaptic structure is critical for information processing in neural circuits. During synaptic transmission, rapid vesicle exocytosis increases the size of never terminals and endocytosis counteracts the increase. Accumulating evidence suggests that SV exocytosis and endocytosis are tightly connected in time and space during SV recycling, and this process is essential for synaptic function and structural stability. Research in the past has illustrated the molecular details of synaptic vesicle (SV) exocytosis and endocytosis; however, the mechanisms that timely connect these two fundamental events are poorly understood at central synapses. Here we discuss recent progress in SV recycling and summarize several emerging mechanisms by which synapses can “sense” the occurrence of exocytosis and timely initiate compensatory endocytosis. They include Ca2+ sensing, SV proteins sensing, and local membrane stress sensing. In addition, the spatial organization of endocytic zones adjacent to active zones provides a structural basis for efficient coupling between SV exocytosis and endocytosis. Through linking different endocytosis pathways with SV fusion, these mechanisms ensure necessary plasticity and robustness of nerve terminals to meet diverse physiological needs.
Collapse
Affiliation(s)
- Xuelin Lou
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
163
|
Domínguez-Álvaro M, Montero-Crespo M, Blazquez-Llorca L, Insausti R, DeFelipe J, Alonso-Nanclares L. Three-dimensional analysis of synapses in the transentorhinal cortex of Alzheimer's disease patients. Acta Neuropathol Commun 2018; 6:20. [PMID: 29499755 PMCID: PMC5834884 DOI: 10.1186/s40478-018-0520-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 12/11/2022] Open
Abstract
Synaptic dysfunction or loss in early stages of Alzheimer’s disease (AD) is thought to be a major structural correlate of cognitive dysfunction. Early loss of episodic memory, which occurs at the early stage of AD, is closely associated with the progressive degeneration of medial temporal lobe (MTL) structures of which the transentorhinal cortex (TEC) is the first affected area. However, no ultrastructural studies have been performed in this region in human brain samples from AD patients. In the present study, we have performed a detailed three-dimensional (3D) ultrastructural analysis using focused ion beam/scanning electron microscopy (FIB/SEM) to investigate possible synaptic alterations in the TEC of patients with AD. Surprisingly, the analysis of the density, morphological features and spatial distribution of synapses in the neuropil showed no significant differences between AD and control samples. However, light microscopy studies showed that cortical thickness of the TEC was severely reduced in AD samples, but there were no changes in the volume occupied by neuronal and glial cell bodies, blood vessels, and neuropil. Thus, the present results indicate that there is a dramatic loss of absolute number of synapses, while the morphology of synaptic junctions and synaptic spatial distribution are maintained. How these changes affect cognitive impairment in AD remains to be elucidated.
Collapse
|
164
|
Single excitatory axons form clustered synapses onto CA1 pyramidal cell dendrites. Nat Neurosci 2018; 21:353-363. [PMID: 29459763 DOI: 10.1038/s41593-018-0084-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 01/05/2018] [Indexed: 01/05/2023]
Abstract
CA1 pyramidal neurons are a major output of the hippocampus and encode features of experience that constitute episodic memories. Feature-selective firing of these neurons results from the dendritic integration of inputs from multiple brain regions. While it is known that synchronous activation of spatially clustered inputs can contribute to firing through the generation of dendritic spikes, there is no established mechanism for spatiotemporal synaptic clustering. Here we show that single presynaptic axons form multiple, spatially clustered inputs onto the distal, but not proximal, dendrites of CA1 pyramidal neurons. These compound connections exhibit ultrastructural features indicative of strong synapses and occur much more commonly in entorhinal than in thalamic afferents. Computational simulations revealed that compound connections depolarize dendrites in a biophysically efficient manner, owing to their inherent spatiotemporal clustering. Our results suggest that distinct afferent projections use different connectivity motifs that differentially contribute to dendritic integration.
Collapse
|
165
|
Tønnesen J, Inavalli VK, Nägerl UV. Super-Resolution Imaging of the Extracellular Space in Living Brain Tissue. Cell 2018; 172:1108-1121.e15. [DOI: 10.1016/j.cell.2018.02.007] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 11/08/2017] [Accepted: 02/01/2018] [Indexed: 01/07/2023]
|
166
|
Study of the Size and Shape of Synapses in the Juvenile Rat Somatosensory Cortex with 3D Electron Microscopy. eNeuro 2018; 5:eN-NWR-0377-17. [PMID: 29387782 PMCID: PMC5790755 DOI: 10.1523/eneuro.0377-17.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/18/2017] [Indexed: 11/21/2022] Open
Abstract
Changes in the size of the synaptic junction are thought to have significant functional consequences. We used focused ion beam milling and scanning electron microscopy (FIB/SEM) to obtain stacks of serial sections from the six layers of the rat somatosensory cortex. We have segmented in 3D a large number of synapses (n = 6891) to analyze the size and shape of excitatory (asymmetric) and inhibitory (symmetric) synapses, using dedicated software. This study provided three main findings. Firstly, the mean synaptic sizes were smaller for asymmetric than for symmetric synapses in all cortical layers. In all cases, synaptic junction sizes followed a log-normal distribution. Secondly, most cortical synapses had disc-shaped postsynaptic densities (PSDs; 93%). A few were perforated (4.5%), while a smaller proportion (2.5%) showed a tortuous horseshoe-shaped perimeter. Thirdly, the curvature was larger for symmetric than for asymmetric synapses in all layers. However, there was no correlation between synaptic area and curvature.
Collapse
|
167
|
Kubota Y, Sohn J, Hatada S, Schurr M, Straehle J, Gour A, Neujahr R, Miki T, Mikula S, Kawaguchi Y. A carbon nanotube tape for serial-section electron microscopy of brain ultrastructure. Nat Commun 2018; 9:437. [PMID: 29382816 PMCID: PMC5789869 DOI: 10.1038/s41467-017-02768-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 12/26/2017] [Indexed: 01/09/2023] Open
Abstract
Automated tape-collecting ultramicrotomy in conjunction with scanning electron microscopy (SEM) is a powerful approach for volume electron microscopy and three-dimensional neuronal circuit analysis. Current tapes are limited by section wrinkle formation, surface scratches and sample charging during imaging. Here we show that a plasma-hydrophilized carbon nanotube (CNT)-coated polyethylene terephthalate (PET) tape effectively resolves these issues and produces SEM images of comparable quality to those from transmission electron microscopy. CNT tape can withstand multiple rounds of imaging, offer low surface resistance across the entire tape length and generate no wrinkles during the collection of ultrathin sections. When combined with an enhanced en bloc staining protocol, CNT tape-processed brain sections reveal detailed synaptic ultrastructure. In addition, CNT tape is compatible with post-embedding immunostaining for light and electron microscopy. We conclude that CNT tape can enable high-resolution volume electron microscopy for brain ultrastructure analysis.
Collapse
Affiliation(s)
- Yoshiyuki Kubota
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan. .,Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan.
| | - Jaerin Sohn
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan.,Research Fellow of Japan Society for the Promotion of Science (JSPS), 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan
| | - Sayuri Hatada
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan
| | - Meike Schurr
- Department of Connectomics, Max-Planck Institute for Brain Research, Max-von-Laue-Str. 4, D-60438, Frankfurt, Germany
| | - Jakob Straehle
- Department of Connectomics, Max-Planck Institute for Brain Research, Max-von-Laue-Str. 4, D-60438, Frankfurt, Germany
| | - Anjali Gour
- Department of Connectomics, Max-Planck Institute for Brain Research, Max-von-Laue-Str. 4, D-60438, Frankfurt, Germany
| | - Ralph Neujahr
- Carl Zeiss Microscopy GmbH, ZEISS Microscopy Customer Center Europe, Rudolph-Eber-Str. 2, D- 873447, Oberkochen, Germany
| | - Takafumi Miki
- Graduate School of Brain Science, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto, 610-0394, Japan
| | - Shawn Mikula
- Electrons-Photons-Neurons, Max-Planck Institute of Neurobiology, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Yasuo Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), 5-1 Myodaiji-Higashiyama, Okazaki, Aichi, 444-8787, Japan
| |
Collapse
|
168
|
Mitochondrial Ultrastructure Is Coupled to Synaptic Performance at Axonal Release Sites. eNeuro 2018; 5:eN-NWR-0390-17. [PMID: 29383328 PMCID: PMC5788698 DOI: 10.1523/eneuro.0390-17.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/28/2017] [Accepted: 01/06/2018] [Indexed: 01/08/2023] Open
Abstract
Mitochondrial function in neurons is tightly linked with metabolic and signaling mechanisms that ultimately determine neuronal performance. The subcellular distribution of these organelles is dynamically regulated as they are directed to axonal release sites on demand, but whether mitochondrial internal ultrastructure and molecular properties would reflect the actual performance requirements in a synapse-specific manner, remains to be established. Here, we examined performance-determining ultrastructural features of presynaptic mitochondria in GABAergic and glutamatergic axons of mice and human. Using electron-tomography and super-resolution microscopy we found, that these features were coupled to synaptic strength: mitochondria in boutons with high synaptic activity exhibited an ultrastructure optimized for high rate metabolism and contained higher levels of the respiratory chain protein cytochrome-c (CytC) than mitochondria in boutons with lower activity. The strong, cell type-independent correlation between mitochondrial ultrastructure, molecular fingerprints and synaptic performance suggests that changes in synaptic activity could trigger ultrastructural plasticity of presynaptic mitochondria, likely to adjust their performance to the actual metabolic demand.
Collapse
|
169
|
Neef J, Urban NT, Ohn TL, Frank T, Jean P, Hell SW, Willig KI, Moser T. Quantitative optical nanophysiology of Ca 2+ signaling at inner hair cell active zones. Nat Commun 2018; 9:290. [PMID: 29348575 PMCID: PMC5773603 DOI: 10.1038/s41467-017-02612-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/14/2017] [Indexed: 12/17/2022] Open
Abstract
Ca2+ influx triggers the release of synaptic vesicles at the presynaptic active zone (AZ). A quantitative characterization of presynaptic Ca2+ signaling is critical for understanding synaptic transmission. However, this has remained challenging to establish at the required resolution. Here, we employ confocal and stimulated emission depletion (STED) microscopy to quantify the number (20-330) and arrangement (mostly linear 70 nm × 100-600 nm clusters) of Ca2+ channels at AZs of mouse cochlear inner hair cells (IHCs). Establishing STED Ca2+ imaging, we analyze presynaptic Ca2+ signals at the nanometer scale and find confined elongated Ca2+ domains at normal IHC AZs, whereas Ca2+ domains are spatially spread out at the AZs of bassoon-deficient IHCs. Performing 2D-STED fluorescence lifetime analysis, we arrive at estimates of the Ca2+ concentrations at stimulated IHC AZs of on average 25 µM. We propose that IHCs form bassoon-dependent presynaptic Ca2+-channel clusters of similar density but scalable length, thereby varying the number of Ca2+ channels amongst individual AZs.
Collapse
Affiliation(s)
- Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany.,Bernstein Focus for Neurotechnology, University of Göttingen, 37075 Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Nicolai T Urban
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany. .,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany.
| | - Tzu-Lun Ohn
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099, Göttingen, Germany.,Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany.,Bernstein Focus for Neurotechnology, University of Göttingen, 37075 Göttingen, Germany
| | - Thomas Frank
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099, Göttingen, Germany.,Friedrich Miescher Institute for Biomedical Research, 4058, Basel, Switzerland
| | - Philippe Jean
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Stefan W Hell
- Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Katrin I Willig
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany. .,Department of Nanobiophotonics, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany. .,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany. .,Optical Nanoscopy in Neuroscience, University Medical Center Göttingen, 37099, Göttingen, Germany.
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37099, Göttingen, Germany. .,Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany. .,Bernstein Focus for Neurotechnology, University of Göttingen, 37075 Göttingen, Germany. .,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany. .,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany. .,Bernstein Center for Computational Neuroscience, University of Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
170
|
Nakano Y, Karube F, Hirai Y, Kobayashi K, Hioki H, Okamoto S, Kameda H, Fujiyama F. Parvalbumin-producing striatal interneurons receive excitatory inputs onto proximal dendrites from the motor thalamus in male mice. J Neurosci Res 2018; 96:1186-1207. [PMID: 29314192 DOI: 10.1002/jnr.24214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 01/09/2023]
Abstract
In rodents, the dorsolateral striatum regulates voluntary movement by integrating excitatory inputs from the motor-related cerebral cortex and thalamus to produce contingent inhibitory output to other basal ganglia nuclei. Striatal parvalbumin (PV)-producing interneurons receiving this excitatory input then inhibit medium spiny neurons (MSNs) and modify their outputs. To understand basal ganglia function in motor control, it is important to reveal the precise synaptic organization of motor-related cortical and thalamic inputs to striatal PV interneurons. To examine which domains of the PV neurons receive these excitatory inputs, we used male bacterial artificial chromosome transgenic mice expressing somatodendritic membrane-targeted green fluorescent protein in PV neurons. An anterograde tracing study with the adeno-associated virus vector combined with immunodetection of pre- and postsynaptic markers visualized the distribution of the excitatory appositions on PV dendrites. Statistical analysis revealed that the density of thalamostriatal appositions along the dendrites was significantly higher on the proximal than distal dendrites. In contrast, there was no positional preference in the density of appositions from axons of the dorsofrontal cortex. Population observations of thalamostriatal and corticostriatal appositions by immunohistochemistry for pathway-specific vesicular glutamate transporters confirmed that thalamic inputs preferentially, and cortical ones less preferentially, made apposition on proximal dendrites of PV neurons. This axodendritic organization suggests that PV neurons produce fast and reliable inhibition of MSNs in response to thalamic inputs and process excitatory inputs from motor cortices locally and plastically, possibly together with other GABAergic and dopaminergic dendritic inputs, to modulate MSN inhibition.
Collapse
Affiliation(s)
- Yasutake Nakano
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Fuyuki Karube
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Yasuharu Hirai
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Hiroyuki Hioki
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichiro Okamoto
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kameda
- Department of Physiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Fumino Fujiyama
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| |
Collapse
|
171
|
Laghaei R, Ma J, Tarr TB, Homan AE, Kelly L, Tilvawala MS, Vuocolo BS, Rajasekaran HP, Meriney SD, Dittrich M. Transmitter release site organization can predict synaptic function at the neuromuscular junction. J Neurophysiol 2017; 119:1340-1355. [PMID: 29357458 DOI: 10.1152/jn.00168.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We have investigated the impact of transmitter release site (active zone; AZ) structure on synaptic function by physically rearranging the individual AZ elements in a previously published frog neuromuscular junction (NMJ) AZ model into the organization observed in a mouse NMJ AZ. We have used this strategy, purposefully without changing the properties of AZ elements between frog and mouse models (even though there are undoubtedly differences between frog and mouse AZ elements in vivo), to directly test how structure influences function at the level of an AZ. Despite a similarly ordered ion channel array substructure within both frog and mouse AZs, frog AZs are much longer and position docked vesicles in a different location relative to AZ ion channels. Physiologically, frog AZs have a lower probability of transmitter release compared with mouse AZs, and frog NMJs facilitate strongly during short stimulus trains in contrast with mouse NMJs that depress slightly. Using our computer modeling approach, we found that a simple rearrangement of the AZ building blocks of the frog model into a mouse AZ organization could recapitulate the physiological differences between these two synapses. These results highlight the importance of simple AZ protein organization to synaptic function. NEW & NOTEWORTHY A simple rearrangement of the basic building blocks in the frog neuromuscular junction model into a mouse transmitter release site configuration predicted the major physiological differences between these two synapses, suggesting that transmitter release site structure and organization is a strong predictor of function.
Collapse
Affiliation(s)
- Rozita Laghaei
- Biomedical Applications Group, Pittsburgh Supercomputing Center, Carnegie Mellon University , Pittsburgh, Pennsylvania
| | - Jun Ma
- Biomedical Applications Group, Pittsburgh Supercomputing Center, Carnegie Mellon University , Pittsburgh, Pennsylvania
| | - Tyler B Tarr
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Anne E Homan
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Lauren Kelly
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Megha S Tilvawala
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Blake S Vuocolo
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Harini P Rajasekaran
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Stephen D Meriney
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Markus Dittrich
- Biomedical Applications Group, Pittsburgh Supercomputing Center, Carnegie Mellon University , Pittsburgh, Pennsylvania.,Department of Neuroscience, Center for Neuroscience, University of Pittsburgh , Pittsburgh, Pennsylvania.,BioTeam Inc., Middleton , Massachusetts
| |
Collapse
|
172
|
Walter AM, Böhme MA, Sigrist SJ. Vesicle release site organization at synaptic active zones. Neurosci Res 2017; 127:3-13. [PMID: 29275162 DOI: 10.1016/j.neures.2017.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 11/30/2022]
Abstract
Information transfer between nerve cells (neurons) forms the basis of behavior, emotion, and survival. Signal transduction from one neuron to another occurs at synapses, and relies on both electrical and chemical signal propagation. At chemical synapses, incoming electrical action potentials trigger the release of chemical neurotransmitters that are sensed by the connected cell and here reconverted to an electrical signal. The presynaptic conversion of an electrical to a chemical signal is an energy demanding, highly regulated process that relies on a complex, evolutionarily conserved molecular machinery. Here, we review the biophysical characteristics of this process, the current knowledge of the molecules operating in this reaction and genetic specializations that may have evolved to shape inter-neuronal signaling.
Collapse
Affiliation(s)
- Alexander M Walter
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, Berlin 13125, Germany.
| | - Mathias A Böhme
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Roessle-Straße 10, Berlin 13125, Germany
| | - Stephan J Sigrist
- Freie Universität Berlin, Institute for Biology/Genetics, Takustraße 6, 14195 Berlin, Germany; NeuroCure, Cluster of Excellence, Charité Universitätsmedizin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
173
|
How to maintain active zone integrity during high-frequency transmission. Neurosci Res 2017; 127:61-69. [PMID: 29221908 DOI: 10.1016/j.neures.2017.10.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/10/2017] [Accepted: 10/26/2017] [Indexed: 11/20/2022]
Abstract
In the central nervous system, the frequency at which reliable synaptic transmission can be maintained varies strongly between different types of synapses. Several pre- and postsynaptic processes must interact to enable high-frequency synaptic transmission. One of the mechanistically most challenging issues arises during repetitive neurotransmitter release, when synaptic vesicles fuse in rapid sequence with the presynaptic plasma membrane within the active zone (AZ), potentially interfering with the structural integrity of the AZ itself. Here we summarize potential mechanisms that help to maintain AZ integrity, including arrangement and mobility of release sites, calcium channel mobility, as well as release site clearance via lateral diffusion of vesicular proteins and via endocytotic membrane retrieval. We discuss how different types of synapses use these strategies to maintain high-frequency synaptic transmission.
Collapse
|
174
|
Glebov OO, Jackson RE, Winterflood CM, Owen DM, Barker EA, Doherty P, Ewers H, Burrone J. Nanoscale Structural Plasticity of the Active Zone Matrix Modulates Presynaptic Function. Cell Rep 2017; 18:2715-2728. [PMID: 28297674 PMCID: PMC5368346 DOI: 10.1016/j.celrep.2017.02.064] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 12/10/2016] [Accepted: 02/20/2017] [Indexed: 12/21/2022] Open
Abstract
The active zone (AZ) matrix of presynaptic terminals coordinates the recruitment of voltage-gated calcium channels (VGCCs) and synaptic vesicles to orchestrate neurotransmitter release. However, the spatial organization of the AZ and how it controls vesicle fusion remain poorly understood. Here, we employ super-resolution microscopy and ratiometric imaging to visualize the AZ structure on the nanoscale, revealing segregation between the AZ matrix, VGCCs, and putative release sites. Long-term blockade of neuronal activity leads to reversible AZ matrix unclustering and presynaptic actin depolymerization, allowing for enrichment of AZ machinery. Conversely, patterned optogenetic stimulation of postsynaptic neurons retrogradely enhanced AZ clustering. In individual synapses, AZ clustering was inversely correlated with local VGCC recruitment and vesicle cycling. Acute actin depolymerization led to rapid (5 min) nanoscale AZ matrix unclustering. We propose a model whereby neuronal activity modulates presynaptic function in a homeostatic manner by altering the clustering state of the AZ matrix.
Collapse
Affiliation(s)
- Oleg O Glebov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre For Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| | - Rachel E Jackson
- Centre For Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Christian M Winterflood
- Randall Division of Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London SE1 1UL, UK
| | - Dylan M Owen
- Randall Division of Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London SE1 1UL, UK; Department of Physics, Faculty of Natural and Mathematical Sciences, King's College London, London WC2R 2LS, UK
| | - Ellen A Barker
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Patrick Doherty
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Helge Ewers
- Randall Division of Molecular Biophysics, Faculty of Life Sciences and Medicine, King's College London, London SE1 1UL, UK; Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Juan Burrone
- Centre For Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| |
Collapse
|
175
|
Satnav for cells: Destination membrane fusion. Cell Calcium 2017; 68:14-23. [PMID: 29129204 DOI: 10.1016/j.ceca.2017.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/20/2017] [Accepted: 10/07/2017] [Indexed: 11/23/2022]
|
176
|
Szoboszlay M, Kirizs T, Nusser Z. Objective quantification of nanoscale protein distributions. Sci Rep 2017; 7:15240. [PMID: 29127366 PMCID: PMC5681686 DOI: 10.1038/s41598-017-15695-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/01/2017] [Indexed: 01/13/2023] Open
Abstract
Nanoscale distribution of molecules within small subcellular compartments of neurons critically influences their functional roles. Although, numerous ways of analyzing the spatial arrangement of proteins have been described, a thorough comparison of their effectiveness is missing. Here we present an open source software, GoldExt, with a plethora of measures for quantification of the nanoscale distribution of proteins in subcellular compartments (e.g. synapses) of nerve cells. First, we compared the ability of five different measures to distinguish artificial uniform and clustered patterns from random point patterns. Then, the performance of a set of clustering algorithms was evaluated on simulated datasets with predefined number of clusters. Finally, we applied the best performing methods to experimental data, and analyzed the nanoscale distribution of different pre- and postsynaptic proteins, revealing random, uniform and clustered sub-synaptic distribution patterns. Our results reveal that application of a single measure is sufficient to distinguish between different distributions.
Collapse
Affiliation(s)
- Miklos Szoboszlay
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Tekla Kirizs
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
177
|
Abstract
At each of the brain's vast number of synapses, the presynaptic nerve terminal, synaptic cleft, and postsynaptic specialization form a transcellular unit to enable efficient transmission of information between neurons. While we know much about the molecular machinery within each compartment, we are only beginning to understand how these compartments are structurally registered and functionally integrated with one another. This review will describe the organization of each compartment and then discuss their alignment across pre- and postsynaptic cells at a nanometer scale. We propose that this architecture may allow for precise synaptic information exchange and may be modulated to contribute to the remarkable plasticity of brain function.
Collapse
Affiliation(s)
- Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
178
|
Biederer T, Kaeser PS, Blanpied TA. Transcellular Nanoalignment of Synaptic Function. Neuron 2017; 96:680-696. [PMID: 29096080 PMCID: PMC5777221 DOI: 10.1016/j.neuron.2017.10.006] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/29/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
At each of the brain's vast number of synapses, the presynaptic nerve terminal, synaptic cleft, and postsynaptic specialization form a transcellular unit to enable efficient transmission of information between neurons. While we know much about the molecular machinery within each compartment, we are only beginning to understand how these compartments are structurally registered and functionally integrated with one another. This review will describe the organization of each compartment and then discuss their alignment across pre- and postsynaptic cells at a nanometer scale. We propose that this architecture may allow for precise synaptic information exchange and may be modulated to contribute to the remarkable plasticity of brain function.
Collapse
Affiliation(s)
- Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA.
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
179
|
Pulido C, Marty A. Quantal Fluctuations in Central Mammalian Synapses: Functional Role of Vesicular Docking Sites. Physiol Rev 2017; 97:1403-1430. [DOI: 10.1152/physrev.00032.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 12/23/2022] Open
Abstract
Quantal fluctuations are an integral part of synaptic signaling. At the frog neuromuscular junction, Bernard Katz proposed that quantal fluctuations originate at “reactive sites” where specific structures of the presynaptic membrane interact with synaptic vesicles. However, the physical nature of reactive sites has remained unclear, both at the frog neuromuscular junction and at central synapses. Many central synapses, called simple synapses, are small structures containing a single presynaptic active zone and a single postsynaptic density of receptors. Several lines of evidence indicate that simple synapses may release several synaptic vesicles in response to a single action potential. However, in some synapses at least, each release event activates a significant fraction of the postsynaptic receptors, giving rise to a sublinear relation between vesicular release and postsynaptic current. Partial receptor saturation as well as synaptic jitter gives to simple synapse signaling the appearance of a binary process. Recent investigations of simple synapses indicate that the number of released vesicles follows binomial statistics, with a maximum reflecting the number of docking sites present in the active zone. These results suggest that at central synapses, vesicular docking sites represent the reactive sites proposed by Katz. The macromolecular architecture and molecular composition of docking sites are presently investigated with novel combinations of techniques. It is proposed that variations in docking site numbers are central in defining intersynaptic variability and that docking site occupancy is a key parameter regulating short-term synaptic plasticity.
Collapse
Affiliation(s)
- Camila Pulido
- Laboratory of Brain Physiology, CNRS UMR 8118, Paris Descartes University, Paris, France
| | - Alain Marty
- Laboratory of Brain Physiology, CNRS UMR 8118, Paris Descartes University, Paris, France
| |
Collapse
|
180
|
Reddy-Alla S, Böhme MA, Reynolds E, Beis C, Grasskamp AT, Mampell MM, Maglione M, Jusyte M, Rey U, Babikir H, McCarthy AW, Quentin C, Matkovic T, Bergeron DD, Mushtaq Z, Göttfert F, Owald D, Mielke T, Hell SW, Sigrist SJ, Walter AM. Stable Positioning of Unc13 Restricts Synaptic Vesicle Fusion to Defined Release Sites to Promote Synchronous Neurotransmission. Neuron 2017; 95:1350-1364.e12. [DOI: 10.1016/j.neuron.2017.08.016] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 05/10/2017] [Accepted: 08/09/2017] [Indexed: 12/24/2022]
|
181
|
Lübbert M, Goral RO, Satterfield R, Putzke T, van den Maagdenberg AM, Kamasawa N, Young SM. A novel region in the Ca V2.1 α 1 subunit C-terminus regulates fast synaptic vesicle fusion and vesicle docking at the mammalian presynaptic active zone. eLife 2017; 6. [PMID: 28786379 PMCID: PMC5548488 DOI: 10.7554/elife.28412] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 07/06/2017] [Indexed: 01/23/2023] Open
Abstract
In central nervous system (CNS) synapses, action potential-evoked neurotransmitter release is principally mediated by CaV2.1 calcium channels (CaV2.1) and is highly dependent on the physical distance between CaV2.1 and synaptic vesicles (coupling). Although various active zone proteins are proposed to control coupling and abundance of CaV2.1 through direct interactions with the CaV2.1 α1 subunit C-terminus at the active zone, the role of these interaction partners is controversial. To define the intrinsic motifs that regulate coupling, we expressed mutant CaV2.1 α1 subunits on a CaV2.1 null background at the calyx of Held presynaptic terminal. Our results identified a region that directly controlled fast synaptic vesicle release and vesicle docking at the active zone independent of CaV2.1 abundance. In addition, proposed individual direct interactions with active zone proteins are insufficient for CaV2.1 abundance and coupling. Therefore, our work advances our molecular understanding of CaV2.1 regulation of neurotransmitter release in mammalian CNS synapses.
Collapse
Affiliation(s)
- Matthias Lübbert
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | - R Oliver Goral
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Department of Anatomy and Cell Biology, University of Iowa, Iowa City, United States
| | - Rachel Satterfield
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | - Travis Putzke
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | | | - Naomi Kamasawa
- Max Planck Florida Electron Microscopy Core, Max Planck Florida Institute for Neuroscience, Jupiter, United States
| | - Samuel M Young
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, United States.,Department of Anatomy and Cell Biology, University of Iowa, Iowa City, United States.,Department of Otolaryngology, University of Iowa, Iowa City, United States.,Iowa Neuroscience Institute, University of Iowa, Iowa City, United States.,Aging Mind Brain Initiative, University of Iowa, Iowa City, United States
| |
Collapse
|
182
|
Cooperative stochastic binding and unbinding explain synaptic size dynamics and statistics. PLoS Comput Biol 2017; 13:e1005668. [PMID: 28704399 PMCID: PMC5546711 DOI: 10.1371/journal.pcbi.1005668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 08/07/2017] [Accepted: 06/30/2017] [Indexed: 11/28/2022] Open
Abstract
Synapses are dynamic molecular assemblies whose sizes fluctuate significantly over time-scales of hours and days. In the current study, we examined the possibility that the spontaneous microscopic dynamics exhibited by synaptic molecules can explain the macroscopic size fluctuations of individual synapses and the statistical properties of synaptic populations. We present a mesoscopic model, which ties the two levels. Its basic premise is that synaptic size fluctuations reflect cooperative assimilation and removal of molecules at a patch of postsynaptic membrane. The introduction of cooperativity to both assimilation and removal in a stochastic biophysical model of these processes, gives rise to features qualitatively similar to those measured experimentally: nanoclusters of synaptic scaffolds, fluctuations in synaptic sizes, skewed, stable size distributions and their scaling in response to perturbations. Our model thus points to the potentially fundamental role of cooperativity in dictating synaptic remodeling dynamics and offers a conceptual understanding of these dynamics in terms of central microscopic features and processes. Neurons communicate through specialized sites of cell–cell contact known as synapses. This vast set of connections is believed to be crucial for sensory processing, motor function, learning and memory. Experimental data from recent years suggest that synapses are not static structures, but rather dynamic assemblies of molecules that move in, out and between nearby synapses, with these dynamics driving changes in synaptic properties over time. Thus, in addition to changes directed by activity or other physiological signals, synapses also exhibit spontaneous changes that have particular dynamical and statistical signatures. Given the immense complexity of synapses at the molecular scale, how can one hope to understand the principles that govern these spontaneous changes and statistical signatures? Here we offer a mesoscopic modelling approach—situated between detailed microscopic and abstract macroscopic approaches—to advance this understanding. Our model, based on simplified biophysical assumptions, shows that spontaneous cooperative binding and unbinding of proteins at synaptic sites can give rise to dynamic and statistical signatures similar to those measured in experiments. Importantly, we find cooperativity to be indispensable in this regard. Our model thus offers a conceptual understanding of synaptic dynamics and statistical features in terms of a fundamental biological principle, namely cooperativity.
Collapse
|
183
|
Wang SSH, Held RG, Wong MY, Liu C, Karakhanyan A, Kaeser PS. Fusion Competent Synaptic Vesicles Persist upon Active Zone Disruption and Loss of Vesicle Docking. Neuron 2017; 91:777-791. [PMID: 27537483 DOI: 10.1016/j.neuron.2016.07.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/23/2016] [Accepted: 06/23/2016] [Indexed: 11/19/2022]
Abstract
In a nerve terminal, synaptic vesicle docking and release are restricted to an active zone. The active zone is a protein scaffold that is attached to the presynaptic plasma membrane and opposed to postsynaptic receptors. Here, we generated conditional knockout mice removing the active zone proteins RIM and ELKS, which additionally led to loss of Munc13, Bassoon, Piccolo, and RIM-BP, indicating disassembly of the active zone. We observed a near-complete lack of synaptic vesicle docking and a strong reduction in vesicular release probability and the speed of exocytosis, but total vesicle numbers, SNARE protein levels, and postsynaptic densities remained unaffected. Despite loss of the priming proteins Munc13 and RIM and of docked vesicles, a pool of releasable vesicles remained. Thus, the active zone is necessary for synaptic vesicle docking and to enhance release probability, but releasable vesicles can be localized distant from the presynaptic plasma membrane.
Collapse
Affiliation(s)
- Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Richard G Held
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Man Yan Wong
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Aziz Karakhanyan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
184
|
Numbers of presynaptic Ca 2+ channel clusters match those of functionally defined vesicular docking sites in single central synapses. Proc Natl Acad Sci U S A 2017; 114:E5246-E5255. [PMID: 28607047 DOI: 10.1073/pnas.1704470114] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Many central synapses contain a single presynaptic active zone and a single postsynaptic density. Vesicular release statistics at such "simple synapses" indicate that they contain a small complement of docking sites where vesicles repetitively dock and fuse. In this work, we investigate functional and morphological aspects of docking sites at simple synapses made between cerebellar parallel fibers and molecular layer interneurons. Using immunogold labeling of SDS-treated freeze-fracture replicas, we find that Cav2.1 channels form several clusters per active zone with about nine channels per cluster. The mean value and range of intersynaptic variation are similar for Cav2.1 cluster numbers and for functional estimates of docking-site numbers obtained from the maximum numbers of released vesicles per action potential. Both numbers grow in relation with synaptic size and decrease by a similar extent with age between 2 wk and 4 wk postnatal. Thus, the mean docking-site numbers were 3.15 at 2 wk (range: 1-10) and 2.03 at 4 wk (range: 1-4), whereas the mean numbers of Cav2.1 clusters were 2.84 at 2 wk (range: 1-8) and 2.37 at 4 wk (range: 1-5). These changes were accompanied by decreases of miniature current amplitude (from 93 pA to 56 pA), active-zone surface area (from 0.0427 μm2 to 0.0234 μm2), and initial success rate (from 0.609 to 0.353), indicating a tightening of synaptic transmission with development. Altogether, these results suggest a close correspondence between the number of functionally defined vesicular docking sites and that of clusters of voltage-gated calcium channels.
Collapse
|
185
|
Alonso B, Bartolomé-Martín D, Ferrero JJ, Ramírez-Franco J, Torres M, Sánchez-Prieto J. CB1 receptors down-regulate a cAMP/Epac2/PLC pathway to silence the nerve terminals of cerebellar granule cells. J Neurochem 2017; 142:350-364. [PMID: 28445587 DOI: 10.1111/jnc.14059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 01/06/2023]
Abstract
Cannabinoid receptors mediate short-term retrograde inhibition of neurotransmitter release, as well as long-term depression of synaptic transmission at excitatory synapses. The responses of individual nerve terminals in VGLUT1-pHluorin transfected cerebellar granule cells to cannabinoids have shown that prolonged activation of cannabinoid type 1 receptors (CB1Rs) silences a subpopulation of previously active synaptic boutons. Adopting a combined pharmacological and genetic approach to study the molecular mechanisms of CB1R-induced silencing, we found that adenylyl cyclase inhibition decreases cAMP levels while it increases the number of silent synaptic boutons and occludes the induction of further silencing by the cannabinoid agonist HU-210. Guanine nucleotide exchange proteins directly activated by cAMP (Epac proteins) mediate some of the presynaptic effects of cAMP in the potentiation of synaptic transmission. ESI05, a selective Epac2 inhibitor, and U-73122, the specific inhibitor of phospholipase C (PLC), both augment the number of silent synaptic boutons. Moreover, they abolish the capacity of the Epac activator, 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate monosodium hydrate, to prevent HU-210-induced silencing consistent with PLC signaling lying downstream of Epac2 proteins. Furthermore, Rab3-interacting molecule (RIM)1α KO cells have many more basally silent synaptic boutons (12.9 ± 3.5%) than wild-type cells (1.1 ± 0.5%). HU-210 induced further silencing in these mutant cells, although 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate monosodium hydrate only awoke the HU-210-induced silence and not the basally silent synaptic boutons. This behavior can be rescued by expressing RIM1α in RIM1α KO cells, these cells behaving very much like wild-type cells. These findings support the hypothesis that a cAMP/Epac/PLC signaling pathway targeting the release machinery appears to mediate cannabinoid-induced presynaptic silencing.
Collapse
Affiliation(s)
- Beatris Alonso
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - David Bartolomé-Martín
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - José Javier Ferrero
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Jorge Ramírez-Franco
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Magdalena Torres
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
186
|
Van Vactor D, Sigrist SJ. Presynaptic morphogenesis, active zone organization and structural plasticity in Drosophila. Curr Opin Neurobiol 2017; 43:119-129. [PMID: 28388491 DOI: 10.1016/j.conb.2017.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/31/2022]
Abstract
Effective adaptation of neural circuit function to a changing environment requires many forms of plasticity. Among these, structural plasticity is one of the most durable, and is also an intrinsic part of the developmental logic for the formation and refinement of synaptic connectivity. Structural plasticity of presynaptic sites can involve the addition, remodeling, or removal of pre- and post-synaptic elements. However, this requires coordination of morphogenesis and assembly of the subcellular machinery for neurotransmitter release within the presynaptic neuron, as well as coordination of these events with the postsynaptic cell. While much progress has been made in revealing the cell biological mechanisms of postsynaptic structural plasticity, our understanding of presynaptic mechanisms is less complete.
Collapse
Affiliation(s)
- David Van Vactor
- Department of Cell Biology and Program in Neuroscience, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA; Okinawa Institute of Science and Technology, Graduate University, Tancha 1919-1, Onna-son, Okinawa, Japan.
| | - Stephan J Sigrist
- Institut für Biologie/Genetik and NeuroCure, Freie Universität Berlin, Takustrasse 6, D-14195 Berlin, Germany.
| |
Collapse
|
187
|
Hirano M, Takada Y, Wong CF, Yamaguchi K, Kotani H, Kurokawa T, Mori MX, Snutch TP, Ronjat M, De Waard M, Mori Y. C-terminal splice variants of P/Q-type Ca 2+ channel Ca V2.1 α 1 subunits are differentially regulated by Rab3-interacting molecule proteins. J Biol Chem 2017; 292:9365-9381. [PMID: 28377503 DOI: 10.1074/jbc.m117.778829] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/26/2017] [Indexed: 11/06/2022] Open
Abstract
Voltage-dependent Ca2+ channels (VDCCs) mediate neurotransmitter release controlled by presynaptic proteins such as the scaffolding proteins Rab3-interacting molecules (RIMs). RIMs confer sustained activity and anchoring of synaptic vesicles to the VDCCs. Multiple sites on the VDCC α1 and β subunits have been reported to mediate the RIMs-VDCC interaction, but their significance is unclear. Because alternative splicing of exons 44 and 47 in the P/Q-type VDCC α1 subunit CaV2.1 gene generates major variants of the CaV2.1 C-terminal region, known for associating with presynaptic proteins, we focused here on the protein regions encoded by these two exons. Co-immunoprecipitation experiments indicated that the C-terminal domain (CTD) encoded by CaV2.1 exons 40-47 interacts with the α-RIMs, RIM1α and RIM2α, and this interaction was abolished by alternative splicing that deletes the protein regions encoded by exons 44 and 47. Electrophysiological characterization of VDCC currents revealed that the suppressive effect of RIM2α on voltage-dependent inactivation (VDI) was stronger than that of RIM1α for the CaV2.1 variant containing the region encoded by exons 44 and 47. Importantly, in the CaV2.1 variant in which exons 44 and 47 were deleted, strong RIM2α-mediated VDI suppression was attenuated to a level comparable with that of RIM1α-mediated VDI suppression, which was unaffected by the exclusion of exons 44 and 47. Studies of deletion mutants of the exon 47 region identified 17 amino acid residues on the C-terminal side of a polyglutamine stretch as being essential for the potentiated VDI suppression characteristic of RIM2α. These results suggest that the interactions of the CaV2.1 CTD with RIMs enable CaV2.1 proteins to distinguish α-RIM isoforms in VDI suppression of P/Q-type VDCC currents.
Collapse
Affiliation(s)
- Mitsuru Hirano
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Yoshinori Takada
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Chee Fah Wong
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and.,the Department of Biology, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, 35900 Tanjung Malim, Perak, Malaysia
| | - Kazuma Yamaguchi
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Hiroshi Kotani
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Tatsuki Kurokawa
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Masayuki X Mori
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and
| | - Terrance P Snutch
- the Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada, and
| | - Michel Ronjat
- the LabEx Ion Channels, Science and Therapeutics, INSERM UMR1087/CNRS UMR6291, Institut du Thorax, Université de Nantes, Nantes F-44000, France
| | - Michel De Waard
- the LabEx Ion Channels, Science and Therapeutics, INSERM UMR1087/CNRS UMR6291, Institut du Thorax, Université de Nantes, Nantes F-44000, France
| | - Yasuo Mori
- From the Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, and .,the Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyoto 615-8510, Japan
| |
Collapse
|
188
|
Maschi D, Klyachko VA. Spatiotemporal Regulation of Synaptic Vesicle Fusion Sites in Central Synapses. Neuron 2017; 94:65-73.e3. [DOI: 10.1016/j.neuron.2017.03.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 02/10/2017] [Accepted: 03/02/2017] [Indexed: 12/22/2022]
|
189
|
Sweeney AM, Fleming KE, McCauley JP, Rodriguez MF, Martin ET, Sousa AA, Leapman RD, Scimemi A. PAR1 activation induces rapid changes in glutamate uptake and astrocyte morphology. Sci Rep 2017; 7:43606. [PMID: 28256580 PMCID: PMC5335386 DOI: 10.1038/srep43606] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 01/26/2017] [Indexed: 01/24/2023] Open
Abstract
The G-protein coupled, protease-activated receptor 1 (PAR1) is a membrane protein expressed in astrocytes. Fine astrocytic processes are in tight contact with neurons and blood vessels and shape excitatory synaptic transmission due to their abundant expression of glutamate transporters. PAR1 is proteolytically-activated by bloodstream serine proteases also involved in the formation of blood clots. PAR1 activation has been suggested to play a key role in pathological states like thrombosis, hemostasis and inflammation. What remains unclear is whether PAR1 activation also regulates glutamate uptake in astrocytes and how this shapes excitatory synaptic transmission among neurons. Here we show that, in the mouse hippocampus, PAR1 activation induces a rapid structural re-organization of the neuropil surrounding glutamatergic synapses, which is associated with faster clearance of synaptically-released glutamate from the extracellular space. This effect can be recapitulated using realistic 3D Monte Carlo reaction-diffusion simulations, based on axial scanning transmission electron microscopy (STEM) tomography reconstructions of excitatory synapses. The faster glutamate clearance induced by PAR1 activation leads to short- and long-term changes in excitatory synaptic transmission. Together, these findings identify PAR1 as an important regulator of glutamatergic signaling in the hippocampus and a possible target molecule to limit brain damage during hemorrhagic stroke.
Collapse
Affiliation(s)
- Amanda M Sweeney
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA
| | - Kelsey E Fleming
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA
| | - John P McCauley
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA
| | - Marvin F Rodriguez
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA.,SUNY Oneonta, Dept. Computer Science, 108 Ravine Parkway, Oneonta NY 13820, USA
| | - Elliot T Martin
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA
| | - Alioscka A Sousa
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 9000 Rockville Pike, Bethesda MD 20852, USA
| | - Richard D Leapman
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, 9000 Rockville Pike, Bethesda MD 20852, USA
| | - Annalisa Scimemi
- SUNY Albany, Dept. Biology, 1400 Washington Avenue, Albany NY 12222, USA
| |
Collapse
|
190
|
Gundersen CB. The Structure of the Synaptic Vesicle-Plasma Membrane Interface Constrains SNARE Models of Rapid, Synchronous Exocytosis at Nerve Terminals. Front Mol Neurosci 2017; 10:48. [PMID: 28280457 PMCID: PMC5321675 DOI: 10.3389/fnmol.2017.00048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 02/13/2017] [Indexed: 12/27/2022] Open
Abstract
Contemporary models of neurotransmitter release invoke direct or indirect interactions between the Ca2+ sensor, synaptotagmin and the incompletely zippered soluble, N-ethyl-maleimide-sensitive factor attachment protein receptor (SNARE) complex. However, recent electron microscopic (EM) investigations have raised pragmatic issues concerning the mechanism by which SNAREs trigger membrane fusion at nerve terminals. The first issue is related to the finding that the area of contact between a “fully primed” synaptic vesicle and the plasma membrane can exceed 600 nm2. Approximately four-thousands lipid molecules can inhabit this contact zone. Thus, renewed efforts will be needed to explain how the zippering of as few as two SNARE complexes mobilizes these lipids to achieve membrane fusion. The second issue emerges from the finding that “docking filaments” are sandwiched within the area of vesicle-plasma membrane contact. It is challenging to reconcile the location of these filaments with SNARE models of exocytosis. Instead, this commentary outlines how these data are more compatible with a model in which a cluster of synaptotagmins catalyzes exocytotic membrane fusion.
Collapse
Affiliation(s)
- Cameron B Gundersen
- Department of Molecular and Medical Pharmacology, David Geffen UCLA School of Medicine Los Angeles, CA, USA
| |
Collapse
|
191
|
Pan-neurexin perturbation results in compromised synapse stability and a reduction in readily releasable synaptic vesicle pool size. Sci Rep 2017; 7:42920. [PMID: 28220838 PMCID: PMC5318902 DOI: 10.1038/srep42920] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/16/2017] [Indexed: 11/08/2022] Open
Abstract
Neurexins are a diverse family of cell adhesion molecules that localize to presynaptic specializations of CNS neurons. Heterologous expression of neurexins in non-neuronal cells leads to the recruitment of postsynaptic proteins in contacting dendrites of co-cultured neurons, implicating neurexins in synapse formation. However, isoform-specific knockouts of either all α- or all β-neurexins show defects in synaptic transmission but an unaltered density of glutamatergic synapses, a finding that argues against an essential function of neurexins in synaptogenesis. To address the role of neurexin in synapse formation and function, we disrupted the function of all α- and β-neurexins in cultured hippocampal neurons by shRNA knockdown or by overexpressing a neurexin mutant that is unable to bind to postsynaptic neurexin ligands. We show that neurexin perturbation results in an attenuation of neurotransmitter release that is in large part due to a reduction in the number of readily releasable synaptic vesicles. We also find that neurexin perturbation fails to alter the ability of neurons to form synapses, but rather leads to more frequent synapse elimination. These experiments suggest that neurexins are dispensable for the formation of initial synaptic contacts, but play an essential role in the stabilization and functional maturation of synapses.
Collapse
|
192
|
Ubiquitin Ligase RNF138 Promotes Episodic Ataxia Type 2-Associated Aberrant Degradation of Human Ca v2.1 (P/Q-Type) Calcium Channels. J Neurosci 2017; 37:2485-2503. [PMID: 28167673 DOI: 10.1523/jneurosci.3070-16.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/09/2017] [Accepted: 01/31/2017] [Indexed: 11/21/2022] Open
Abstract
Voltage-gated CaV2.1 channels comprise a pore-forming α1A subunit with auxiliary α2δ and β subunits. CaV2.1 channels play an essential role in regulating synaptic signaling. Mutations in the human gene encoding the CaV2.1 subunit are associated with the cerebellar disease episodic ataxia type 2 (EA2). Several EA2-causing mutants exhibit impaired protein stability and exert dominant-negative suppression of CaV2.1 wild-type (WT) protein expression via aberrant proteasomal degradation. Here, we set out to delineate the protein degradation mechanism of human CaV2.1 subunit by identifying RNF138, an E3 ubiquitin ligase, as a novel CaV2.1-binding partner. In neurons, RNF138 and CaV2.1 coexist in the same protein complex and display notable subcellular colocalization at presynaptic and postsynaptic regions. Overexpression of RNF138 promotes polyubiquitination and accelerates protein turnover of CaV2.1. Disrupting endogenous RNF138 function with a mutant (RNF138-H36E) or shRNA infection significantly upregulates the CaV2.1 protein level and enhances CaV2.1 protein stability. Disrupting endogenous RNF138 function also effectively rescues the defective protein expression of EA2 mutants, as well as fully reversing EA2 mutant-induced excessive proteasomal degradation of CaV2.1 WT subunits. RNF138-H36E coexpression only partially restores the dominant-negative effect of EA2 mutants on CaV2.1 WT functional expression, which can be attributed to defective membrane trafficking of CaV2.1 WT in the presence of EA2 mutants. We propose that RNF138 plays a critical role in the homeostatic regulation of CaV2.1 protein level and functional expression and that RNF138 serves as the primary E3 ubiquitin ligase promoting EA2-associated aberrant degradation of human CaV2.1 subunits.SIGNIFICANCE STATEMENT Loss-of-function mutations in the human CaV2.1 subunit are linked to episodic ataxia type 2 (EA2), a dominantly inherited disease characterized by paroxysmal attacks of ataxia and nystagmus. EA2-causing mutants may exert dominant-negative effects on the CaV2.1 wild-type subunit via aberrant proteasomal degradation. The molecular nature of the CaV2.1 ubiquitin-proteasome degradation pathway is currently unknown. The present study reports the first identification of an E3 ubiquitin ligase for CaV2.1, RNF138. CaV2.1 protein stability is dynamically regulated by RNF138 and auxiliary α2δ and β subunits. We provide a proof of concept that protecting the human CaV2.1 subunit from excessive proteasomal degradation with specific interruption of endogenous RNF138 function may partially contribute to the future development of a novel therapeutic strategy for EA2 patients.
Collapse
|
193
|
Mukunda CL, Narayanan R. Degeneracy in the regulation of short-term plasticity and synaptic filtering by presynaptic mechanisms. J Physiol 2017; 595:2611-2637. [PMID: 28026868 DOI: 10.1113/jp273482] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS We develop a new biophysically rooted, physiologically constrained conductance-based synaptic model to mechanistically account for short-term facilitation and depression, respectively through residual calcium and transmitter depletion kinetics. We address the specific question of how presynaptic components (including voltage-gated ion channels, pumps, buffers and release-handling mechanisms) and interactions among them define synaptic filtering and short-term plasticity profiles. Employing global sensitivity analyses (GSAs), we show that near-identical synaptic filters and short-term plasticity profiles could emerge from disparate presynaptic parametric combinations with weak pairwise correlations. Using virtual knockout models, a technique to address the question of channel-specific contributions within the GSA framework, we unveil the differential and variable impact of each ion channel on synaptic physiology. Our conclusions strengthen the argument that parametric and interactional complexity in biological systems should not be viewed from the limited curse-of-dimensionality standpoint, but from the evolutionarily advantageous perspective of providing functional robustness through degeneracy. ABSTRACT Information processing in neurons is known to emerge as a gestalt of pre- and post-synaptic filtering. However, the impact of presynaptic mechanisms on synaptic filters has not been quantitatively assessed. Here, we developed a biophysically rooted, conductance-based model synapse that was endowed with six different voltage-gated ion channels, calcium pumps, calcium buffer and neurotransmitter-replenishment mechanisms in the presynaptic terminal. We tuned our model to match the short-term plasticity profile and band-pass structure of Schaffer collateral synapses, and performed sensitivity analyses to demonstrate that presynaptic voltage-gated ion channels regulated synaptic filters through changes in excitability and associated calcium influx. These sensitivity analyses also revealed that calcium- and release-control mechanisms were effective regulators of synaptic filters, but accomplished this without changes in terminal excitability or calcium influx. Next, to perform global sensitivity analysis, we generated 7000 randomized models spanning 15 presynaptic parameters, and computed eight different physiological measurements in each of these models. We validated these models by applying experimentally obtained bounds on their measurements, and found 104 (∼1.5%) models to match the validation criteria for all eight measurements. Analysing these valid models, we demonstrate that analogous synaptic filters emerge from disparate combinations of presynaptic parameters exhibiting weak pairwise correlations. Finally, using virtual knockout models, we establish the variable and differential impact of different presynaptic channels on synaptic filters, underlining the critical importance of interactions among different presynaptic components in defining synaptic physiology. Our results have significant implications for protein-localization strategies required for physiological robustness and for degeneracy in long-term synaptic plasticity profiles.
Collapse
Affiliation(s)
- Chinmayee L Mukunda
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
194
|
Target Cell Type-Dependent Differences in Ca 2+ Channel Function Underlie Distinct Release Probabilities at Hippocampal Glutamatergic Terminals. J Neurosci 2017; 37:1910-1924. [PMID: 28115484 DOI: 10.1523/jneurosci.2024-16.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 01/04/2017] [Accepted: 01/10/2017] [Indexed: 12/24/2022] Open
Abstract
Target cell type-dependent differences in presynaptic release probability (Pr ) and short-term plasticity are intriguing features of cortical microcircuits that increase the computational power of neuronal networks. Here, we tested the hypothesis that different voltage-gated Ca2+ channel densities in presynaptic active zones (AZs) underlie different Pr values. Two-photon Ca2+ imaging, triple immunofluorescent labeling, and 3D electron microscopic (EM) reconstruction of rat CA3 pyramidal cell axon terminals revealed ∼1.7-1.9 times higher Ca2+ inflow per AZ area in high Pr boutons synapsing onto parvalbumin-positive interneurons (INs) than in low Pr boutons synapsing onto mGluR1α-positive INs. EM replica immunogold labeling, however, demonstrated only 1.15 times larger Cav2.1 and Cav2.2 subunit densities in high Pr AZs. Our results indicate target cell type-specific modulation of voltage-gated Ca2+ channel function or different subunit composition as possible mechanisms underlying the functional differences. In addition, high Pr synapses are also characterized by a higher density of docked vesicles, suggesting that a concerted action of these mechanisms underlies the functional differences.SIGNIFICANCE STATEMENT Target cell type-dependent variability in presynaptic properties is an intriguing feature of cortical synapses. When a single cortical pyramidal cell establishes a synapse onto a somatostatin-expressing interneuron (IN), the synapse releases glutamate with low probability, whereas the next bouton of the same axon has high release probability when its postsynaptic target is a parvalbumin-expressing IN. Here, we used combined molecular, imaging, and anatomical approaches to investigate the mechanisms underlying these differences. Our functional experiments implied an approximately twofold larger Ca2+ channel density in high release probability boutons, whereas freeze-fracture immunolocalization demonstrated only a 15% difference in Ca2+ channel subunit densities. Our results point toward a postsynaptic target cell type-dependent regulation of Ca2+ channel function or different subunit composition as the underlying mechanism.
Collapse
|
195
|
Bruckner JJ, Zhan H, Gratz SJ, Rao M, Ukken F, Zilberg G, O'Connor-Giles KM. Fife organizes synaptic vesicles and calcium channels for high-probability neurotransmitter release. J Cell Biol 2016; 216:231-246. [PMID: 27998991 PMCID: PMC5223599 DOI: 10.1083/jcb.201601098] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 10/19/2016] [Accepted: 11/29/2016] [Indexed: 11/22/2022] Open
Abstract
Fife is a Piccolo-RIM–related protein that regulates neurotransmission and motor behavior through an unknown mechanism. Here, Bruckner et al. show that Fife organizes synaptic vesicle docking and coupling to calcium channels to establish and modulate synaptic strength. The strength of synaptic connections varies significantly and is a key determinant of communication within neural circuits. Mechanistic insight into presynaptic factors that establish and modulate neurotransmitter release properties is crucial to understanding synapse strength, circuit function, and neural plasticity. We previously identified Drosophila Piccolo-RIM-related Fife, which regulates neurotransmission and motor behavior through an unknown mechanism. Here, we demonstrate that Fife localizes and interacts with RIM at the active zone cytomatrix to promote neurotransmitter release. Loss of Fife results in the severe disruption of active zone cytomatrix architecture and molecular organization. Through electron tomographic and electrophysiological studies, we find a decrease in the accumulation of release-ready synaptic vesicles and their release probability caused by impaired coupling to Ca2+ channels. Finally, we find that Fife is essential for the homeostatic modulation of neurotransmission. We propose that Fife organizes active zones to create synaptic vesicle release sites within nanometer distance of Ca2+ channel clusters for reliable and modifiable neurotransmitter release.
Collapse
Affiliation(s)
- Joseph J Bruckner
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Hong Zhan
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Scott J Gratz
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Monica Rao
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Fiona Ukken
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Gregory Zilberg
- Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| | - Kate M O'Connor-Giles
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53706 .,Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706.,Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
196
|
Oh YM, Karube F, Takahashi S, Kobayashi K, Takada M, Uchigashima M, Watanabe M, Nishizawa K, Kobayashi K, Fujiyama F. Using a novel PV-Cre rat model to characterize pallidonigral cells and their terminations. Brain Struct Funct 2016; 222:2359-2378. [PMID: 27995326 DOI: 10.1007/s00429-016-1346-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/25/2016] [Indexed: 10/20/2022]
Abstract
In the present study, we generated a novel parvalbumin (PV)-Cre rat model and conducted detailed morphological and electrophysiological investigations of axons from PV neurons in globus pallidus (GP). The GP is considered as a relay nucleus in the indirect pathway of the basal ganglia (BG). Previous studies have used molecular profiling and projection patterns to demonstrate cellular heterogeneity in the GP; for example, PV-expressing neurons are known to comprise approximately 50% of GP neurons and represent majority of prototypic neurons that project to the subthalamic nucleus and/or output nuclei of BG, entopeduncular nucleus and substantia nigra (SN). The present study aimed to identify the characteristic projection patterns of PV neurons in the GP (PV-GP neurons) and determine whether these neurons target dopaminergic or GABAergic neurons in SN pars compacta (SNc) or reticulata (SNr), respectively. We initially found that (1) 57% of PV neurons co-expressed Lim-homeobox 6, (2) the PV-GP terminals were preferentially distributed in the ventral part of dorsal tier of SNc, (3) PV-GP neurons formed basket-like appositions with the somata of tyrosine hydroxylase, PV, calretinin and cholecystokinin immunoreactive neurons in the SN, and (4) in vitro whole-cell recording during optogenetic photo-stimulation of PV-GP terminals in SNc demonstrated that PV-GP neurons strongly inhibited dopamine neurons via GABAA receptors. These results suggest that dopamine neurons receive direct focal inputs from PV-GP prototypic neurons. The identification of high-contrast inhibitory systems on dopamine neurons might represent a key step toward understanding the BG function.
Collapse
Affiliation(s)
- Yoon-Mi Oh
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan
| | - Fuyuki Karube
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan
| | - Susumu Takahashi
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, 484-8506, Japan
| | - Motokazu Uchigashima
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Kayo Nishizawa
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Fumino Fujiyama
- Laboratory of Neural Circuitry, Graduate School of Brain Science, Doshisha University, Kyotanabe, 610-0394, Japan.
| |
Collapse
|
197
|
Fu P, Wen Y, Xiong Y, Zhang Y, Zhang H, Xie Y, Shi Q. Abnormal Expression of FBXL20 in Refractory Epilepsy Patients and a Pilocarpine-Induced Rat Model. Neurochem Res 2016; 41:3020-3031. [PMID: 27502938 DOI: 10.1007/s11064-016-2021-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 07/08/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
E3 ubiquitin ligases are important protein-modifying enzymes involved in the pathogenesis of a variety of neurodegenerative diseases. F-box and leucine-rich repeat protein 20 (FBXL20), an E3 ubiquitin ligase widely expressed in the central nervous system, plays an important role in the ubiquitin-dependent degradation of regulating synaptic membrane exocytosis 1 (RIM1), which is an important factor in the release of synaptic vesicles. FBXL20 has been associated with a variety of neurodegenerative diseases; thus, we hypothesized that FBXL20 is involved in the development of epilepsy. Herein, we used immunofluorescence staining, immunohistochemistry and western blotting to determine the expression pattern of FBXL20 in temporal lobe epilepsy patients and pilocarpine-induced epilepsy animal models. We also injected SD rats with lentivirus-vector mediated overexpression of FBXL20. The results showed that FBXL20 is expressed in the membrane and the cytoplasm of cortical neurons, and overexpression of FBXL20 decreased the onset level of spontaneous seizure, the frequency and duration of seizures. Additionally, FBXL20 protein level was decreased but RIM1 protein level was increased in the epileptic group compared with the LV-FBXL20 and LV-GFP group. These findings in humans were consistent with the results from a pilocarpine-induced animal model of chronic epilepsy. Thus, abnormal expression of FBXL20 might play an important role in the development of epilepsy.
Collapse
Affiliation(s)
- Pengfei Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - YueTao Wen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yan Xiong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanke Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haiyang Zhang
- Department of Pediatric Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, 610000, China
| | - Yanfeng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Quanhong Shi
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
198
|
Delvendahl I, Hallermann S. The Cerebellar Mossy Fiber Synapse as a Model for High-Frequency Transmission in the Mammalian CNS. Trends Neurosci 2016; 39:722-737. [DOI: 10.1016/j.tins.2016.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/17/2016] [Accepted: 09/20/2016] [Indexed: 10/20/2022]
|
199
|
Voigt A, Freund R, Heck J, Missler M, Obermair GJ, Thomas U, Heine M. Dynamic association of calcium channel subunits at the cellular membrane. NEUROPHOTONICS 2016; 3:041809. [PMID: 27872869 PMCID: PMC5093230 DOI: 10.1117/1.nph.3.4.041809] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/10/2016] [Indexed: 05/25/2023]
Abstract
High voltage gated calcium channels (VGCCs) are composed of at least three subunits, one pore forming [Formula: see text]-subunit, an intracellular [Formula: see text]-variant, and a mostly extracellular [Formula: see text]-variant. Interactions between these subunits determine the kinetic properties of VGCCs. It is unclear whether these interactions are stable over time or rather transient. Here, we used single-molecule tracking to investigate the surface diffusion of [Formula: see text]- and [Formula: see text]-subunits at the cell surface. We found that [Formula: see text]-subunits show higher surface mobility than [Formula: see text]-subunits, and that they are only transiently confined together, suggesting a weak association between [Formula: see text]- and [Formula: see text]-subunits. Moreover, we observed that different [Formula: see text]-subunits engage in different degrees of association with the [Formula: see text]-subunit, revealing the tighter interaction of [Formula: see text] with [Formula: see text]. These data indicate a distinct regulation of the [Formula: see text] interaction in VGCC subtypes. We modeled their membrane dynamics in a Monte Carlo simulation using experimentally determined diffusion constants. Our modeling predicts that the ratio of associated [Formula: see text]- and [Formula: see text]-subunits mainly depends on their expression density and confinement in the membrane. Based on the different motilities of particular [Formula: see text]-subunit combinations, we propose that their dynamic assembly and disassembly represent an important mechanism to regulate the signaling properties of VGCC.
Collapse
Affiliation(s)
- Andreas Voigt
- Otto-von-Guericke-University of Magdeburg, Lehrstuhl Systemverfahrenstechnik, Universitätsplatz 2, Magdeburg D-39106, Germany
| | - Romy Freund
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Jennifer Heck
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Markus Missler
- Westfälische Wilhelms-University, Institute of Anatomy and Molecular Neurobiology, Vesaliusweg 2, Münster 48149, Germany
| | - Gerald J. Obermair
- Medical University Innsbruck, Division of Physiology, Department of Physiology and Medical Physics, Schöpfstrasse 41, Innsbruck 6020, Austria
| | - Ulrich Thomas
- Leibniz-Institute of Neurobiology, Department Neurochemistry, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Martin Heine
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
- Otto-von-Guericke-University Magdeburg, Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, Magdeburg D-39106, Germany
| |
Collapse
|
200
|
RIM-binding protein 2 regulates release probability by fine-tuning calcium channel localization at murine hippocampal synapses. Proc Natl Acad Sci U S A 2016; 113:11615-11620. [PMID: 27671655 DOI: 10.1073/pnas.1605256113] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The tight spatial coupling of synaptic vesicles and voltage-gated Ca2+ channels (CaVs) ensures efficient action potential-triggered neurotransmitter release from presynaptic active zones (AZs). Rab-interacting molecule-binding proteins (RIM-BPs) interact with Ca2+ channels and via RIM with other components of the release machinery. Although human RIM-BPs have been implicated in autism spectrum disorders, little is known about the role of mammalian RIM-BPs in synaptic transmission. We investigated RIM-BP2-deficient murine hippocampal neurons in cultures and slices. Short-term facilitation is significantly enhanced in both model systems. Detailed analysis in culture revealed a reduction in initial release probability, which presumably underlies the increased short-term facilitation. Superresolution microscopy revealed an impairment in CaV2.1 clustering at AZs, which likely alters Ca2+ nanodomains at release sites and thereby affects release probability. Additional deletion of RIM-BP1 does not exacerbate the phenotype, indicating that RIM-BP2 is the dominating RIM-BP isoform at these synapses.
Collapse
|