1
|
Kim S, Jang G, Kim H, Lim D, Han KA, Um JW, Ko J. MDGAs perform activity-dependent synapse type-specific suppression via distinct extracellular mechanisms. Proc Natl Acad Sci U S A 2024; 121:e2322978121. [PMID: 38900791 PMCID: PMC11214077 DOI: 10.1073/pnas.2322978121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
MDGA (MAM domain containing glycosylphosphatidylinositol anchor) family proteins were previously identified as synaptic suppressive factors. However, various genetic manipulations have yielded often irreconcilable results, precluding precise evaluation of MDGA functions. Here, we found that, in cultured hippocampal neurons, conditional deletion of MDGA1 and MDGA2 causes specific alterations in synapse numbers, basal synaptic transmission, and synaptic strength at GABAergic and glutamatergic synapses, respectively. Moreover, MDGA2 deletion enhanced both N-methyl-D-aspartate (NMDA) receptor- and α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor-mediated postsynaptic responses. Strikingly, ablation of both MDGA1 and MDGA2 abolished the effect of deleting individual MDGAs that is abrogated by chronic blockade of synaptic activity. Molecular replacement experiments further showed that MDGA1 requires the meprin/A5 protein/PTPmu (MAM) domain, whereas MDGA2 acts via neuroligin-dependent and/or MAM domain-dependent pathways to regulate distinct postsynaptic properties. Together, our data demonstrate that MDGA paralogs act as unique negative regulators of activity-dependent postsynaptic organization at distinct synapse types, and cooperatively contribute to adjustment of excitation-inhibition balance.
Collapse
Affiliation(s)
- Seungjoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Dongseok Lim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Kyung Ah Han
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
- Center for Synapse Diversity and Specificity, Daegu Gyeongbuk Institute of Science and Technology, Daegu42988, Korea
| |
Collapse
|
2
|
Marshall AH, Hanson MA, Boyle DJ, Nagarajan D, Bibi N, Fitzgerald J, Gaitten E, Kokiko-Cochran ON, Gu B, Wester JC. Arid1b haploinsufficiency in pyramidal neurons causes cellular and circuit changes in neocortex but is not sufficient to produce behavioral or seizure phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597344. [PMID: 38895205 PMCID: PMC11185765 DOI: 10.1101/2024.06.04.597344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Arid1b is a high confidence risk gene for autism spectrum disorder that encodes a subunit of a chromatin remodeling complex expressed in neuronal progenitors. Haploinsufficiency causes a broad range of social, behavioral, and intellectual disability phenotypes, including Coffin-Siris syndrome. Recent work using transgenic mouse models suggests pathology is due to deficits in proliferation, survival, and synaptic development of cortical neurons. However, there is conflicting evidence regarding the relative roles of excitatory projection neurons and inhibitory interneurons in generating abnormal cognitive and behavioral phenotypes. Here, we conditionally knocked out either one or both copies of Arid1b from excitatory projection neuron progenitors and systematically investigated the effects on intrinsic membrane properties, synaptic physiology, social behavior, and seizure susceptibility. We found that disrupting Arid1b expression in excitatory neurons alters their membrane properties, including hyperpolarizing action potential threshold; however, these changes depend on neuronal subtype. Using paired whole-cell recordings, we found increased synaptic connectivity rate between projection neurons. Furthermore, we found reduced strength of excitatory synapses to parvalbumin (PV)-expression inhibitory interneurons. These data suggest an increase in the ratio of excitation to inhibition. However, the strength of inhibitory synapses from PV interneurons to excitatory neurons was enhanced, which may rebalance this ratio. Indeed, Arid1b haploinsufficiency in projection neurons was insufficient to cause social deficits and seizure phenotypes observed in a preclinical germline haploinsufficient mouse model. Our data suggest that while excitatory projection neurons likely contribute to autistic phenotypes, pathology in these cells is not the primary cause.
Collapse
|
3
|
Yang SM, Ghoshal A, Hubbard JM, Gackière F, Teyssié R, Neale SA, Hopkins SC, Koblan KS, Bristow LJ, Dedic N. TAAR1 agonist ulotaront modulates striatal and hippocampal glutamate function in a state-dependent manner. Neuropsychopharmacology 2024; 49:1091-1103. [PMID: 38110609 PMCID: PMC11109157 DOI: 10.1038/s41386-023-01779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 12/20/2023]
Abstract
Aberrant dopaminergic and glutamatergic function, particularly within the striatum and hippocampus, has repeatedly been associated with the pathophysiology of schizophrenia. Supported by preclinical and recent clinical data, trace amine-associated receptor 1 (TAAR1) agonism has emerged as a potential new treatment approach for schizophrenia. While current evidence implicates TAAR1-mediated regulation of dopaminergic tone as the primary circuit mechanism, little is known about the effects of TAAR1 agonists on the glutamatergic system and excitation-inhibition balance. Here we assessed the impact of ulotaront (SEP-363856), a TAAR1 agonist in Phase III clinical development for schizophrenia, on glutamate function in the mouse striatum and hippocampus. Ulotaront reduced spontaneous glutamatergic synaptic transmission and neuronal firing in striatal and hippocampal brain slices, respectively. Interestingly, ulotaront potentiated electrically-evoked excitatory synaptic transmission in both brain regions, suggesting the ability to modulate glutamatergic signaling in a state-dependent manner. Similar striatal effects were also observed with the TAAR1 agonist, RO5166017. Furthermore, we show that ulotaront regulates excitation-inhibition balance in the striatum by specifically modulating glutamatergic, but not GABAergic, spontaneous synaptic events. These findings expand the mechanistic circuit hypothesis of ulotaront and TAAR1 agonists, which may be uniquely positioned to normalize both the excessive dopaminergic tone and regulate abnormal glutamatergic function associated with schizophrenia.
Collapse
Affiliation(s)
- Sung M Yang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | - Ayan Ghoshal
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | | | | | | | | | - Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| |
Collapse
|
4
|
Koppensteiner P, Bhandari P, Önal C, Borges-Merjane C, Le Monnier E, Roy U, Nakamura Y, Sadakata T, Sanbo M, Hirabayashi M, Rhee J, Brose N, Jonas P, Shigemoto R. GABA B receptors induce phasic release from medial habenula terminals through activity-dependent recruitment of release-ready vesicles. Proc Natl Acad Sci U S A 2024; 121:e2301449121. [PMID: 38346189 PMCID: PMC10895368 DOI: 10.1073/pnas.2301449121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
GABAB receptor (GBR) activation inhibits neurotransmitter release in axon terminals in the brain, except in medial habenula (MHb) terminals, which show robust potentiation. However, mechanisms underlying this enigmatic potentiation remain elusive. Here, we report that GBR activation on MHb terminals induces an activity-dependent transition from a facilitating, tonic to a depressing, phasic neurotransmitter release mode. This transition is accompanied by a 4.1-fold increase in readily releasable vesicle pool (RRP) size and a 3.5-fold increase of docked synaptic vesicles (SVs) at the presynaptic active zone (AZ). Strikingly, the depressing phasic release exhibits looser coupling distance than the tonic release. Furthermore, the tonic and phasic release are selectively affected by deletion of synaptoporin (SPO) and Ca2+-dependent activator protein for secretion 2 (CAPS2), respectively. SPO modulates augmentation, the short-term plasticity associated with tonic release, and CAPS2 retains the increased RRP for initial responses in phasic response trains. The cytosolic protein CAPS2 showed a SV-associated distribution similar to the vesicular transmembrane protein SPO, and they were colocalized in the same terminals. We developed the "Flash and Freeze-fracture" method, and revealed the release of SPO-associated vesicles in both tonic and phasic modes and activity-dependent recruitment of CAPS2 to the AZ during phasic release, which lasted several minutes. Overall, these results indicate that GBR activation translocates CAPS2 to the AZ along with the fusion of CAPS2-associated SVs, contributing to persistency of the RRP increase. Thus, we identified structural and molecular mechanisms underlying tonic and phasic neurotransmitter release and their transition by GBR activation in MHb terminals.
Collapse
Affiliation(s)
| | - Pradeep Bhandari
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Cihan Önal
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | | | - Elodie Le Monnier
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Utsa Roy
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Yukihiro Nakamura
- Department of Pharmacology, Jikei University School of Medicine, Nishishinbashi, Minato-ku, Tokyo105-8461, Japan
| | - Tetsushi Sadakata
- Advanced Scientific Research Leaders Development Unit, Gunma University Graduate School of Medicine, Maebashi, Gunma371-8511, Japan
| | - Makoto Sanbo
- Section of Mammalian Transgenesis, National Institute for Physiological Sciences, Okazaki444-8585, Japan
| | - Masumi Hirabayashi
- Section of Mammalian Transgenesis, National Institute for Physiological Sciences, Okazaki444-8585, Japan
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen37077, Germany
| | - Peter Jonas
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, Klosterneuburg3400, Austria
| |
Collapse
|
5
|
Zheng Y, Kang S, O'Neill J, Bojak I. Spontaneous slow wave oscillations in extracellular field potential recordings reflect the alternating dominance of excitation and inhibition. J Physiol 2024; 602:713-736. [PMID: 38294945 DOI: 10.1113/jp284587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
In the resting state, cortical neurons can fire action potentials spontaneously but synchronously (Up state), followed by a quiescent period (Down state) before the cycle repeats. Extracellular recordings in the infragranular layer of cortex with a micro-electrode display a negative deflection (depth-negative) during Up states and a positive deflection (depth-positive) during Down states. The resulting slow wave oscillation (SWO) has been studied extensively during sleep and under anaesthesia. However, recent research on the balanced nature of synaptic excitation and inhibition has highlighted our limited understanding of its genesis. Specifically, are excitation and inhibition balanced during SWOs? We analyse spontaneous local field potentials (LFPs) during SWOs recorded from anaesthetised rats via a multi-channel laminar micro-electrode and show that the Down state consists of two distinct synaptic states: a Dynamic Down state associated with depth-positive LFPs and a prominent dipole in the extracellular field, and a Static Down state with negligible (≈ 0 mV $ \approx 0{\mathrm{\;mV}}$ ) LFPs and a lack of dipoles extracellularly. We demonstrate that depth-negative and -positive LFPs are generated by a shift in the balance of synaptic excitation and inhibition from excitation dominance (depth-negative) to inhibition dominance (depth-positive) in the infragranular layer neurons. Thus, although excitation and inhibition co-tune overall, differences in their timing lead to an alternation of dominance, manifesting as SWOs. We further show that Up state initiation is significantly faster if the preceding Down state is dynamic rather than static. Our findings provide a coherent picture of the dependence of SWOs on synaptic activity. KEY POINTS: Cortical neurons can exhibit repeated cycles of spontaneous activity interleaved with periods of relative silence, a phenomenon known as 'slow wave oscillation' (SWO). During SWOs, recordings of local field potentials (LFPs) in the neocortex show depth-negative deflection during the active period (Up state) and depth-positive deflection during the silent period (Down state). Here we further classified the Down state into a dynamic phase and a static phase based on a novel method of classification and revealed non-random, stereotypical sequences of the three states occurring with significantly different transitional kinetics. Our results suggest that the positive and negative deflections in the LFP reflect the shift of the instantaneous balance between excitatory and inhibitory synaptic activity of the local cortical neurons. The differences in transitional kinetics may imply distinct synaptic mechanisms for Up state initiation. The study may provide a new approach for investigating spontaneous brain rhythms.
Collapse
Affiliation(s)
- Ying Zheng
- School of Biological Sciences, Whiteknights, University of Reading, Reading, UK
- Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading, UK
| | - Sungmin Kang
- School of Psychology, Cardiff University, Cardiff, UK
| | | | - Ingo Bojak
- Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading, UK
- School of Psychology and Clinical Language Science, Whiteknights, University of Reading, Reading, UK
| |
Collapse
|
6
|
Souza JR, Lima-Silveira L, Accorsi-Mendonça D, Machado BH. Enhancement of the Evoked Excitatory Transmission in the Nucleus Tractus Solitarius Neurons after Sustained Hypoxia in Mice Depends on A 2A Receptors. Neuroscience 2024; 536:57-71. [PMID: 37979842 DOI: 10.1016/j.neuroscience.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/06/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
The first synapses of the afferents of peripheral chemoreceptors are located in the Nucleus Tractus Solitarius (NTS) and there is evidence that short-term sustained hypoxia (SH - 24 h, FiO2 0.1) facilitates glutamatergic transmission in NTS neurons of rats. Adenosine is an important neuromodulator of synaptic transmission and hypoxia contributes to increase its extracellular concentration. The A2A receptors mediate the excitatory actions of adenosine and are active players in the modulation of neuronal networks in the NTS. Herein, we used knockout mice for A2A receptors (A2AKO) and electrophysiological recordings of NTS neurons were performed to evaluate the contribution of these receptors in the changes in synaptic transmission in NTS neurons of mice submitted to SH. The membrane passive properties and excitability of NTS neurons were not affected by SH and were similar between A2AKO and wild-type mice. The overall amplitude of spontaneous glutamatergic currents in NTS neurons of A2AKO mice was lower than in Balb/c WT mice. SH increased the amplitude of evoked glutamatergic currents of NTS neurons from WT mice by a non-presynaptic mechanism, but this enhancement was not observed in NTS neurons of A2AKO mice. Under normoxia, the amplitude of evoked glutamatergic currents was similar between WT and A2AKO mice. The data indicate that A2A receptors (a) modulate spontaneous glutamatergic currents, (b) do not modulate the evoked glutamatergic transmission in the NTS neurons under control conditions, and (c) are required for the enhancement of glutamatergic transmission observed in the NTS neurons of mice submitted to SH.
Collapse
Affiliation(s)
- Juliana R Souza
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Ludmila Lima-Silveira
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Daniela Accorsi-Mendonça
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil.
| |
Collapse
|
7
|
Kater MSJ, Baumgart KF, Badia-Soteras A, Heistek TS, Carney KE, Timmerman AJ, van Weering JRT, Smit AB, van der Knaap MS, Mansvelder HD, Verheijen MHG, Min R. A novel role for MLC1 in regulating astrocyte-synapse interactions. Glia 2023; 71:1770-1785. [PMID: 37002718 DOI: 10.1002/glia.24368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023]
Abstract
Loss of function of the astrocyte membrane protein MLC1 is the primary genetic cause of the rare white matter disease Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC), which is characterized by disrupted brain ion and water homeostasis. MLC1 is prominently present around fluid barriers in the brain, such as in astrocyte endfeet contacting blood vessels and in processes contacting the meninges. Whether the protein plays a role in other astrocyte domains is unknown. Here, we show that MLC1 is present in distal astrocyte processes, also known as perisynaptic astrocyte processes (PAPs) or astrocyte leaflets, which closely interact with excitatory synapses in the CA1 region of the hippocampus. We find that the PAP tip extending toward excitatory synapses is shortened in Mlc1-null mice. This affects glutamatergic synaptic transmission, resulting in a reduced rate of spontaneous release events and slower glutamate re-uptake under challenging conditions. Moreover, while PAPs in wildtype mice retract from the synapse upon fear conditioning, we reveal that this structural plasticity is disturbed in Mlc1-null mice, where PAPs are already shorter. Finally, Mlc1-null mice show reduced contextual fear memory. In conclusion, our study uncovers an unexpected role for the astrocyte protein MLC1 in regulating the structure of PAPs. Loss of MLC1 alters excitatory synaptic transmission, prevents normal PAP remodeling induced by fear conditioning and disrupts contextual fear memory expression. Thus, MLC1 is a new player in the regulation of astrocyte-synapse interactions.
Collapse
Affiliation(s)
- Mandy S J Kater
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Katharina F Baumgart
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Aina Badia-Soteras
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Tim S Heistek
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Karen E Carney
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - A Jacob Timmerman
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jan R T van Weering
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mark H G Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, The Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Feldthouse MG, Vyleta NP, Smith SM. PLC regulates spontaneous glutamate release triggered by extracellular calcium and readily releasable pool size in neocortical neurons. Front Cell Neurosci 2023; 17:1193485. [PMID: 37260580 PMCID: PMC10228687 DOI: 10.3389/fncel.2023.1193485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/26/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Dynamic physiological changes in brain extracellular calcium ([Ca2+]o) occur when high levels of neuronal activity lead to substantial Ca2+ entry via ion channels reducing local [Ca2+]o. Perturbations of the extracellular microenvironment that increase [Ca2+]o are commonly used to study how [Ca2+] regulates neuronal activity. At excitatory synapses, the Ca2+-sensing receptor (CaSR) and other G-protein coupled receptors link [Ca2+]o and spontaneous glutamate release. Phospholipase C (PLC) is activated by G-proteins and is hypothesized to mediate this process. Methods Patch-clamping cultured neocortical neurons, we tested how spontaneous glutamate release was affected by [Ca2+]o and inhibition of PLC activity. We used hypertonic sucrose (HS) to evaluate the readily releasable pool (RRP) and test if it was affected by inhibition of PLC activity. Results Spontaneous glutamate release substantially increased with [Ca2+]o, and inhibition of PLC activity, with U73122, abolished this effect. PLC-β1 is an abundant isoform in the neocortex, however, [Ca2+]o-dependent spontaneous release was unchanged in PLC-β1 null mutants (PLC-β1-/-). U73122 completely suppressed this response in PLC-β1-/- neurons, indicating that this residual [Ca2+]o-sensitivity may be mediated by other PLC isoforms. The RRP size was substantially reduced after incubation in U73122, but not U73343. Phorbol esters increased RRP size after PLC inhibition. Discussion Together these data point to a strong role for PLC in mediating changes in spontaneous release elicited by [Ca2+]o and other extracellular cues, possibly by modifying the size of the RRP.
Collapse
Affiliation(s)
- Maya G. Feldthouse
- Section of Pulmonary and Critical Care Medicine and Research and Development, VA Portland Health Care System, Portland, OR, United States
| | - Nicholas P. Vyleta
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Stephen M. Smith
- Section of Pulmonary and Critical Care Medicine and Research and Development, VA Portland Health Care System, Portland, OR, United States
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
9
|
Tousley AR, Yeh PWL, Yeh HH. Precocious emergence of cognitive and synaptic dysfunction in 3xTg-AD mice exposed prenatally to ethanol. Alcohol 2023; 107:56-72. [PMID: 36038084 PMCID: PMC10183974 DOI: 10.1016/j.alcohol.2022.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting approximately 50 million people worldwide. Early life risk factors for AD, including prenatal exposures, remain underexplored. Exposure of the fetus to alcohol (ethanol) is not uncommon during pregnancy, and may result in physical, behavioral, and cognitive changes that are first detected during childhood but result in lifelong challenges. Whether or not prenatal ethanol exposure may contribute to Alzheimer's disease risk is not yet known. Here we exposed a mouse model of Alzheimer's disease (3xTg-AD), bearing three dementia-associated transgenes, presenilin1 (PS1M146V), human amyloid precursor protein (APPSwe), and human tau (TauP301S), to ethanol on gestational days 13.5-16.5 using an established binge-type maternal ethanol exposure paradigm. We sought to investigate whether prenatal ethanol exposure resulted in a precocious onset or increased severity of AD progression, or both. We found that a brief binge-type gestational exposure to ethanol during a period of peak neuronal migration to the developing cortex resulted in an earlier onset of spatial memory deficits and behavioral inflexibility in the progeny, as assessed by performance on the modified Barnes maze task. The observed cognitive changes coincided with alterations to both GABAergic and glutamatergic synaptic transmission in layer V/VI neurons, diminished GABAergic interneurons, and increased β-amyloid accumulation in the medial prefrontal cortex. These findings provide the first preclinical evidence for prenatal ethanol exposure as a potential factor for modifying the onset of AD-like behavioral dysfunction and set the groundwork for more comprehensive investigations into the underpinnings of AD-like cognitive changes in individuals with fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Adelaide R Tousley
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States; MD-PhD Program, Geisel School of Medicine at Dartmouth; Integrative Neuroscience at Dartmouth Graduate Program, Hanover, NH, United States
| | - Pamela W L Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Hermes H Yeh
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.
| |
Collapse
|
10
|
Droogers WJ, MacGillavry HD. Plasticity of postsynaptic nanostructure. Mol Cell Neurosci 2023; 124:103819. [PMID: 36720293 DOI: 10.1016/j.mcn.2023.103819] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
The postsynaptic density (PSD) of excitatory synapses is built from a wide variety of scaffolding proteins, receptors, and signaling molecules that collectively orchestrate synaptic transmission. Seminal work over the past decades has led to the identification and functional characterization of many PSD components. In contrast, we know far less about how these constituents are assembled within synapses, and how this organization contributes to synapse function. Notably, recent evidence from high-resolution microscopy studies and in silico models, highlights the importance of the precise subsynaptic structure of the PSD for controlling the strength of synaptic transmission. Even further, activity-driven changes in the distribution of glutamate receptors are acknowledged to contribute to long-term changes in synaptic efficacy. Thus, defining the mechanisms that drive structural changes within the PSD are important for a molecular understanding of synaptic transmission and plasticity. Here, we review the current literature on how the PSD is organized to mediate basal synaptic transmission and how synaptic activity alters the nanoscale organization of synapses to sustain changes in synaptic strength.
Collapse
Affiliation(s)
- W J Droogers
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands
| | - H D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands.
| |
Collapse
|
11
|
Joshi SN, Joshi AN, Joshi ND. Interplay between biochemical processes and network properties generates neuronal up and down states at the tripartite synapse. Phys Rev E 2023; 107:024415. [PMID: 36932559 DOI: 10.1103/physreve.107.024415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 01/03/2023] [Indexed: 06/18/2023]
Abstract
Neuronal up and down states have long been known to exist both in vitro and in vivo. A variety of functions and mechanisms have been proposed for their generation, but there has not been a clear connection between the functions and mechanisms. We explore the potential contribution of cellular-level biochemistry to the network-level mechanisms thought to underlie the generation of up and down states. We develop a neurochemical model of a single tripartite synapse, assumed to be within a network of similar tripartite synapses, to investigate possible function-mechanism links for the appearance of up and down states. We characterize the behavior of our model in different regions of parameter space and show that resource limitation at the tripartite synapse affects its ability to faithfully transmit input signals, leading to extinction-down states. Recovery of resources allows for "reignition" into up states. The tripartite synapse exhibits distinctive "regimes" of operation depending on whether ATP, neurotransmitter (glutamate), both, or neither, is limiting. Our model qualitatively matches the behavior of six disparate experimental systems, including both in vitro and in vivo models, without changing any model parameters except those related to the experimental conditions. We also explore the effects of varying different critical parameters within the model. Here we show that availability of energy, represented by ATP, and glutamate for neurotransmission at the cellular level are intimately related, and are capable of promoting state transitions at the network level as ignition and extinction phenomena. Our model is complementary to existing models of neuronal up and down states in that it focuses on cellular-level dynamics while still retaining essential network-level processes. Our model predicts the existence of a "final common pathway" of behavior at the tripartite synapse arising from scarcity of resources and may explain use dependence in the phenomenon of "local sleep." Ultimately, sleeplike behavior may be a fundamental property of networks of tripartite synapses.
Collapse
Affiliation(s)
- Shubhada N Joshi
- National Center for Adaptive Neurotechnologies (NCAN), David Axelrod Institute, Wadsworth Center, New York State Department of Health, 120 New Scotland Ave., Albany, New York 12208, USA
| | - Aditya N Joshi
- Stanford University School of Medicine, 300 Pasteur Dr., Stanford, California 94305, USA
| | - Narendra D Joshi
- General Electric Global Research, 1 Research Circle, Niskayuna, New York 12309, USA
| |
Collapse
|
12
|
Di Castro MA, Garofalo S, De Felice E, Meneghetti N, Di Pietro E, Mormino A, Mazzoni A, Caleo M, Maggi L, Limatola C. Environmental enrichment counteracts the effects of glioma in primary visual cortex. Neurobiol Dis 2022; 174:105894. [DOI: 10.1016/j.nbd.2022.105894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
|
13
|
Formisano R, Rosikon KD, Singh A, Dhillon HS. The dopamine membrane transporter plays an active modulatory role in synaptic dopamine homeostasis. J Neurosci Res 2022; 100:1551-1559. [PMID: 34747520 PMCID: PMC9079189 DOI: 10.1002/jnr.24965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/17/2021] [Accepted: 08/29/2021] [Indexed: 11/11/2022]
Abstract
Modulatory mechanisms of neurotransmitter release and clearance are highly controlled processes whose finely tuned regulation is critical for functioning of the nervous system. Dysregulation of the monoamine neurotransmitter dopamine can lead to several neuropathies. Synaptic modulation of dopamine is known to involve pre-synaptic D2 auto-receptors and acid sensing ion channels. In addition, the dopamine membrane transporter (DAT), which is responsible for clearance of dopamine from the synaptic cleft, is suspected to play an active role in modulating release of dopamine. Using functional imaging on the Caenorhabditis elegans model system, we show that DAT-1 acts as a negative feedback modulator to neurotransmitter vesicle fusion. Results from our fluorescence recovery after photo-bleaching (FRAP) based experiments were followed up with and reaffirmed using swimming-induced paralysis behavioral assays. Utilizing our numerical FRAP data we have developed a mechanistic model to dissect the dynamics of synaptic vesicle fusion, and compare the feedback effects of DAT-1 with the dopamine auto-receptor. Our experimental results and the mechanistic model are of potential broader significance, as similar dynamics are likely to be used by other synaptic modulators including membrane transporters for other neurotransmitters across species.
Collapse
Affiliation(s)
- Rosaria Formisano
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Katarzyna D. Rosikon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, Biomedical Engineering, Data Sciences Institute, University of Delaware, Newark, DE, USA
| | - Harbinder S. Dhillon
- Department of Biological Sciences, Delaware Center for Neuroscience Research, Delaware State University, Dover, DE, USA
| |
Collapse
|
14
|
Heuser JE. The Structural Basis of Long-Term Potentiation in Hippocampal Synapses, Revealed by Electron Microscopy Imaging of Lanthanum-Induced Synaptic Vesicle Recycling. Front Cell Neurosci 2022; 16:920360. [PMID: 35978856 PMCID: PMC9376242 DOI: 10.3389/fncel.2022.920360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022] Open
Abstract
Hippocampal neurons in dissociated cell cultures were exposed to the trivalent cation lanthanum for short periods (15–30 min) and prepared for electron microscopy (EM), to evaluate the stimulatory effects of this cation on synaptic ultrastructure. Not only were characteristic ultrastructural changes of exaggerated synaptic vesicle turnover seen within the presynapses of these cultures—including synaptic vesicle depletion and proliferation of vesicle-recycling structures—but the overall architecture of a large proportion of the synapses in the cultures was dramatically altered, due to large postsynaptic “bulges” or herniations into the presynapses. Moreover, in most cases, these postsynaptic herniations or protrusions produced by lanthanum were seen by EM to distort or break or “perforate” the so-called postsynaptic densities (PSDs) that harbor receptors and recognition molecules essential for synaptic function. These dramatic EM observations lead us to postulate that such PSD breakages or “perforations” could very possibly create essential substrates or “tags” for synaptic growth, simply by creating fragmented free edges around the PSDs, into which new receptors and recognition molecules could be recruited more easily, and thus, they could represent the physical substrate for the important synaptic growth process known as “long-term potentiation” (LTP). All of this was created simply in hippocampal dissociated cell cultures, and simply by pushing synaptic vesicle recycling way beyond its normal limits with the trivalent cation lanthanum, but we argued in this report that such fundamental changes in synaptic architecture—given that they can occur at all—could also occur at the extremes of normal neuronal activity, which are presumed to lead to learning and memory.
Collapse
|
15
|
Han Y, Cao R, Qin L, Chen LY, Tang AH, Südhof TC, Zhang B. Neuroligin-3 confines AMPA receptors into nanoclusters, thereby controlling synaptic strength at the calyx of Held synapses. SCIENCE ADVANCES 2022; 8:eabo4173. [PMID: 35704570 PMCID: PMC9200272 DOI: 10.1126/sciadv.abo4173] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/02/2022] [Indexed: 05/30/2023]
Abstract
The subsynaptic organization of postsynaptic neurotransmitter receptors into nanoclusters that are aligned with presynaptic release sites is essential for the high fidelity of synaptic transmission. However, the mechanisms controlling the nanoscale organization of neurotransmitter receptors in vivo remain incompletely understood. Here, we deconstructed the role of neuroligin-3 (Nlgn3), a postsynaptic adhesion molecule linked to autism, in organizing AMPA-type glutamate receptors in the calyx of Held synapse. Deletion of Nlgn3 lowered the amplitude and slowed the kinetics of AMPA receptor-mediated synaptic responses. Super-resolution microscopy revealed that, unexpectedly, these impairments in synaptic transmission were associated with an increase in the size of postsynaptic PSD-95 and AMPA receptor nanoclusters but a decrease of the densities in these clusters. Modeling showed that a dilution of AMPA receptors into larger nanocluster volumes decreases synaptic strength. Nlgn3, likely by binding to presynaptic neurexins, thus is a key organizer of AMPA receptor nanoclusters that likely acts via PSD-95 adaptors to optimize the fidelity of synaptic transmission.
Collapse
Affiliation(s)
- Ying Han
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Ran Cao
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China
- CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Liming Qin
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lulu Y. Chen
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94043, USA
| | - Ai-Hui Tang
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China
- CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94043, USA
| | - Bo Zhang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
16
|
Lazarevic V, Yang Y, Paslawski W, Svenningsson P. α-Synuclein induced cholesterol lowering increases tonic and reduces depolarization-evoked synaptic vesicle recycling and glutamate release. NPJ Parkinsons Dis 2022; 8:71. [PMID: 35672421 PMCID: PMC9174203 DOI: 10.1038/s41531-022-00334-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
α-Synuclein (α-syn) is a key molecule linked to Parkinson's disease pathology. Physiologically, the monomeric α-syn in the presynaptic termini is involved in regulation of neurotransmission, but the pathophysiology of extracellular monomeric α-syn is still unknown. Utilizing both in vivo and in vitro approaches, we investigated how extracellular α-syn impact presynaptic structure and function. Our data revealed that treatment with exogenous α-syn leads to increased tonic and decreased depolarization-evoked synaptic vesicle (SV) recycling and glutamate release. This was associated with mobilization of molecularly distinct SV pools and reorganization of active zone components. Our study also showed that exogenous α-syn impaired neuronal cholesterol level and that the cholesterol binding domain of α-syn was sufficient to exert the same presynaptic phenotype as the full-length protein. The present study sheds new light on physiological functions of extracellular α-syn in overall maintenance of presynaptic activity that involves the reorganization of both presynaptic compartment and cholesterol-rich plasma membrane domains.
Collapse
Affiliation(s)
- Vesna Lazarevic
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Yunting Yang
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
17
|
Alipio JB, Riggs LM, Plank M, Keller A. Environmental Enrichment Mitigates the Long-Lasting Sequelae of Perinatal Fentanyl Exposure in Mice. J Neurosci 2022; 42:3557-3569. [PMID: 35332082 PMCID: PMC9053848 DOI: 10.1523/jneurosci.2083-21.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
The opioid epidemic is a rapidly evolving societal issue driven, in part, by a surge in synthetic opioid use. A rise in fentanyl use among pregnant women has led to a 40-fold increase in the number of perinatally-exposed infants in the past decade. These children are more likely to develop mood-related and somatosensory-related conditions later in life, suggesting that fentanyl may permanently alter neural development. Here, we examined the behavioral and synaptic consequences of perinatal fentanyl exposure in adolescent male and female C57BL/6J mice and assessed the therapeutic potential of environmental enrichment to mitigate these effects. Dams were given ad libitum access to fentanyl (10 µg/ml, per os) across pregnancy and until weaning [postnatal day (PD)21]. Perinatally-exposed adolescent mice displayed hyperactivity (PD45), enhanced sensitivity to anxiogenic environments (PD46), and sensory maladaptation (PD47), sustained behavioral effects that were completely normalized by environmental enrichment (PD21-PD45). Additionally, environmental enrichment normalized the fentanyl-induced changes in the frequency of miniature EPSCs (mEPSCs) of layer 2/3 neurons in the primary somatosensory cortex (S1). We also demonstrate that fentanyl impairs short-term potentiation (STP) and long-term potentiation (LTP) in S1 layer 2/3 neurons, which, instead, exhibit a sustained depression of synaptic transmission that is restored by environmental enrichment. On its own, environmental enrichment suppressed long-term depression (LTD) of control S1 neurons from vehicle-treated mice subjected to standard housing conditions. These results demonstrate that the lasting effects of fentanyl can be ameliorated with a noninvasive intervention introduced during early development.SIGNIFICANCE STATEMENT Illicit use of fentanyl accounts for a large proportion of opioid-related overdose deaths. Children exposed to opioids during development have a higher risk of developing neuropsychiatric disorders later in life. Here, we employ a preclinical model of perinatal fentanyl exposure that recapitulates these long-term impairments and show, for the first time, that environmental enrichment can reverse deficits in somatosensory circuit function and behavior. These findings have the potential to directly inform and guide ongoing efforts to mitigate the consequences of perinatal opioid exposure.
Collapse
Affiliation(s)
- Jason Bondoc Alipio
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Lace Marie Riggs
- Department of Psychiatry, Division of Translational and Basic Science, Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Madeline Plank
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Asaf Keller
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
18
|
Tsentsevitsky AN, Khaziev EF, Kovyazina IV, Petrov AM. GIRK channel as a versatile regulator of neurotransmitter release via L-type Ca 2+ channel-dependent mechanism in the neuromuscular junction. Neuropharmacology 2022; 209:109021. [PMID: 35245509 DOI: 10.1016/j.neuropharm.2022.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 01/04/2023]
Abstract
G protein-gated inwardly rectifying potassium (GIRK) channels are one of the main regulators of neuronal excitability. Activation of GIRK channels in the CNS usually leads to postsynaptic inhibition. However, the function of GIRK channels in the presynaptic processes, notably neurotransmitter release form motor nerve terminals, is yet to be comprehensively understood. Here, using electrophysiological and fluorescent approaches, the role of GIRK channels in neurotransmitter release from frog motor nerve terminals was studied. We found that the inhibition of GIRK channels with nanomolar tertiapin-Q synchronized exocytosis events with action potential but suppressed spontaneous and evoked neurotransmitter release, as well as Ca2+ transient and membrane permeability for K+. The action of GIRK channel inhibition on evoked neurotransmission was prevented by selective antagonist of voltage-gated Ca2+ channels of L-type. Furthermore, the effects of muscarinic acetylcholine receptor activation on neurotransmitter release, Ca2+ transient and K+ channel activity were markedly modulated by inhibition of GIRK channels. Thus, at the motor nerve terminals GIRK channels can regulate timing of neurotransmitter release and be a positive modulator of synaptic vesicle exocytosis acting partially via L-type Ca2+ channels. In addition, GIRK channels are key players in a feedback control of neurotransmitter release by muscarinic acetylcholine receptors.
Collapse
Affiliation(s)
- Andrei N Tsentsevitsky
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC "Kazan Scientific Center of RAS", P.o. Box 30, 420111, Kazan, Russia
| | - Eduard F Khaziev
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC "Kazan Scientific Center of RAS", P.o. Box 30, 420111, Kazan, Russia
| | - Irina V Kovyazina
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC "Kazan Scientific Center of RAS", P.o. Box 30, 420111, Kazan, Russia; Kazan State Medical University, Butlerov St., 49, 420008, Kazan, Russia.
| | - Alexey M Petrov
- Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, FRC "Kazan Scientific Center of RAS", P.o. Box 30, 420111, Kazan, Russia; Kazan State Medical University, Butlerov St., 49, 420008, Kazan, Russia
| |
Collapse
|
19
|
Zbili M, Rama S, Benitez MJ, Fronzaroli-Molinieres L, Bialowas A, Boumedine-Guignon N, Garrido JJ, Debanne D. Homeostatic regulation of axonal Kv1.1 channels accounts for both synaptic and intrinsic modifications in the hippocampal CA3 circuit. Proc Natl Acad Sci U S A 2021; 118:e2110601118. [PMID: 34799447 PMCID: PMC8617510 DOI: 10.1073/pnas.2110601118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Homeostatic plasticity of intrinsic excitability goes hand in hand with homeostatic plasticity of synaptic transmission. However, the mechanisms linking the two forms of homeostatic regulation have not been identified so far. Using electrophysiological, imaging, and immunohistochemical techniques, we show here that blockade of excitatory synaptic receptors for 2 to 3 d induces an up-regulation of both synaptic transmission at CA3-CA3 connections and intrinsic excitability of CA3 pyramidal neurons. Intrinsic plasticity was found to be mediated by a reduction of Kv1.1 channel density at the axon initial segment. In activity-deprived circuits, CA3-CA3 synapses were found to express a high release probability, an insensitivity to dendrotoxin, and a lack of depolarization-induced presynaptic facilitation, indicating a reduction in presynaptic Kv1.1 function. Further support for the down-regulation of axonal Kv1.1 channels in activity-deprived neurons was the broadening of action potentials measured in the axon. We conclude that regulation of the axonal Kv1.1 channel constitutes a major mechanism linking intrinsic excitability and synaptic strength that accounts for the functional synergy existing between homeostatic regulation of intrinsic excitability and synaptic transmission.
Collapse
Affiliation(s)
- Mickaël Zbili
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Sylvain Rama
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Maria-José Benitez
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
- Departamento de Química Física Aplicada, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Laure Fronzaroli-Molinieres
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Andrzej Bialowas
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Norah Boumedine-Guignon
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France
| | - Juan José Garrido
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas (CSIC), Madrid 28002, Spain
| | - Dominique Debanne
- Unité de Neurobiologie des canaux Ioniques et de la Synapse (UNIS), UMR_S 1072, INSERM, Aix-Marseille Université, Marseille 13015, France;
| |
Collapse
|
20
|
Ramsey AM, Tang AH, LeGates TA, Gou XZ, Carbone BE, Thompson SM, Biederer T, Blanpied TA. Subsynaptic positioning of AMPARs by LRRTM2 controls synaptic strength. SCIENCE ADVANCES 2021; 7:7/34/eabf3126. [PMID: 34417170 PMCID: PMC8378824 DOI: 10.1126/sciadv.abf3126] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 06/30/2021] [Indexed: 05/07/2023]
Abstract
Recent evidence suggests that nano-organization of proteins within synapses may control the strength of communication between neurons in the brain. The unique subsynaptic distribution of glutamate receptors, which cluster in nanoalignment with presynaptic sites of glutamate release, supports this hypothesis. However, testing it has been difficult because mechanisms controlling subsynaptic organization remain unknown. Reasoning that transcellular interactions could position AMPA receptors (AMPARs), we targeted a key transsynaptic adhesion molecule implicated in controlling AMPAR number, LRRTM2, using engineered, rapid proteolysis. Severing the LRRTM2 extracellular domain led quickly to nanoscale declustering of AMPARs away from release sites, not prompting their escape from synapses until much later. This rapid remodeling of AMPAR position produced significant deficits in evoked, but not spontaneous, postsynaptic receptor activation. These results dissociate receptor numbers from their nanopositioning in determination of synaptic function and support the novel concept that adhesion molecules acutely position receptors to dynamically control synaptic strength.
Collapse
Affiliation(s)
- Austin M Ramsey
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tara A LeGates
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Beatrice E Carbone
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Scott M Thompson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Thomas A Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
21
|
Function of Drosophila Synaptotagmins in membrane trafficking at synapses. Cell Mol Life Sci 2021; 78:4335-4364. [PMID: 33619613 PMCID: PMC8164606 DOI: 10.1007/s00018-021-03788-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
The Synaptotagmin (SYT) family of proteins play key roles in regulating membrane trafficking at neuronal synapses. Using both Ca2+-dependent and Ca2+-independent interactions, several SYT isoforms participate in synchronous and asynchronous fusion of synaptic vesicles (SVs) while preventing spontaneous release that occurs in the absence of stimulation. Changes in the function or abundance of the SYT1 and SYT7 isoforms alter the number and route by which SVs fuse at nerve terminals. Several SYT family members also regulate trafficking of other subcellular organelles at synapses, including dense core vesicles (DCV), exosomes, and postsynaptic vesicles. Although SYTs are linked to trafficking of multiple classes of synaptic membrane compartments, how and when they interact with lipids, the SNARE machinery and other release effectors are still being elucidated. Given mutations in the SYT family cause disorders in both the central and peripheral nervous system in humans, ongoing efforts are defining how these proteins regulate vesicle trafficking within distinct neuronal compartments. Here, we review the Drosophila SYT family and examine their role in synaptic communication. Studies in this invertebrate model have revealed key similarities and several differences with the predicted activity of their mammalian counterparts. In addition, we highlight the remaining areas of uncertainty in the field and describe outstanding questions on how the SYT family regulates membrane trafficking at nerve terminals.
Collapse
|
22
|
Kuhlmann N, Wagner Valladolid M, Quesada-Ramírez L, Farrer MJ, Milnerwood AJ. Chronic and Acute Manipulation of Cortical Glutamate Transmission Induces Structural and Synaptic Changes in Co-cultured Striatal Neurons. Front Cell Neurosci 2021; 15:569031. [PMID: 33679324 PMCID: PMC7930618 DOI: 10.3389/fncel.2021.569031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
In contrast to the prenatal topographic development of sensory cortices, striatal circuit organization is slow and requires the functional maturation of cortical and thalamic excitatory inputs throughout the first postnatal month. While mechanisms regulating synapse development and plasticity are quite well described at excitatory synapses of glutamatergic neurons in the neocortex, comparatively little is known of how this translates to glutamate synapses onto GABAergic neurons in the striatum. Here we investigate excitatory striatal synapse plasticity in an in vitro system, where glutamate can be studied in isolation from dopamine and other neuromodulators. We examined pre-and post-synaptic structural and functional plasticity in GABAergic striatal spiny projection neurons (SPNs), co-cultured with glutamatergic cortical neurons. After synapse formation, medium-term (24 h) TTX silencing increased the density of filopodia, and modestly decreased dendritic spine density, when assayed at 21 days in vitro (DIV). Spine reductions appeared to require residual spontaneous activation of ionotropic glutamate receptors. Conversely, chronic (14 days) TTX silencing markedly reduced spine density without any observed increase in filopodia density. Time-dependent, biphasic changes to the presynaptic marker Synapsin-1 were also observed, independent of residual spontaneous activity. Acute silencing (3 h) did not affect presynaptic markers or postsynaptic structures. To induce rapid, activity-dependent plasticity in striatal neurons, a chemical NMDA receptor-dependent “long-term potentiation (LTP)” paradigm was employed. Within 30 min, this increased spine and GluA1 cluster densities, and the percentage of spines containing GluA1 clusters, without altering the presynaptic signal. The results demonstrate that the growth and pruning of dendritic protrusions is an active process, requiring glutamate receptor activity in striatal projection neurons. Furthermore, NMDA receptor activation is sufficient to drive glutamatergic structural plasticity in SPNs, in the absence of dopamine or other neuromodulators.
Collapse
Affiliation(s)
- Naila Kuhlmann
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Lucía Quesada-Ramírez
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada
| | - Matthew J Farrer
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Austen J Milnerwood
- Centre for Applied Neurogenetics (CAN), University of British Columbia, Vancouver, BC, Canada.,Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
朱 佳. Effects of Tetrodotoxin on Different Types of Synaptic Secretion. Biophysics (Nagoya-shi) 2021. [DOI: 10.12677/biphy.2021.93007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
24
|
Activation of tyrosine phosphatase PTP1B in pyramidal neurons impairs endocannabinoid signaling by tyrosine receptor kinase trkB and causes schizophrenia-like behaviors in mice. Neuropsychopharmacology 2020; 45:1884-1895. [PMID: 32610340 PMCID: PMC7608138 DOI: 10.1038/s41386-020-0755-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Schizophrenia is a debilitating disorder affecting young adults displaying symptoms of cognitive impairment, anxiety, and early social isolation prior to episodes of auditory hallucinations. Cannabis use has been tied to schizophrenia-like symptoms, indicating that dysregulated endogenous cannabinoid signaling may be causally linked to schizophrenia. Previously, we reported that glutamatergic neuron-selective ablation of Lmo4, an endogenous inhibitor of the tyrosine phosphatase PTP1B, impairs endocannabinoid (eCB) production from the metabotropic glutamate receptor mGluR5. These Lmo4-deficient mice display anxiety-like behaviors that are alleviated by local shRNA knockdown or pharmacological inhibition of PTP1B that restores mGluR5-dependent eCB production in the amygdala. Here, we report that these Lmo4-deficient mice also display schizophrenia-like behaviors: impaired working memory assessed in the Y maze and defective sensory gating by prepulse inhibition of the acoustic startle response. Modulation of inhibitory inputs onto layer 2/3 pyramidal neurons of the prefrontal cortex relies on eCB signaling from the brain-derived neurotrophic factor receptor trkB, rather than mGluR5, and this mechanism was defective in Lmo4-deficient mice. Genetic ablation of PTP1B in the glutamatergic neurons lacking Lmo4 restored tyrosine phosphorylation of trkB, trkB-mediated eCB signaling, and ameliorated schizophrenia-like behaviors. Pharmacological inhibition of PTP1B with trodusquemine also restored trkB phosphorylation and improved schizophrenia-like behaviors by restoring eCB signaling, since the CB1 receptor antagonist 1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide blocked this effect. Thus, activation of PTP1B in pyramidal neurons contributes to schizophrenia-like behaviors in Lmo4-deficient mice and genetic or pharmacological intervention targeting PTP1B ameliorates schizophrenia-related deficits.
Collapse
|
25
|
Joffe ME, Winder DG, Conn PJ. Contrasting sex-dependent adaptations to synaptic physiology and membrane properties of prefrontal cortex interneuron subtypes in a mouse model of binge drinking. Neuropharmacology 2020; 178:108126. [PMID: 32781000 DOI: 10.1016/j.neuropharm.2020.108126] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/18/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Alcohol use disorder (AUD) affects all sexes, however women who develop AUD may be particularly susceptible to cravings and other components of the disease. While many brain regions are involved in AUD etiology, proper prefrontal cortex (PFC) function is particularly important for top-down craving management and the moderation of drinking behaviors. Essential regulation of PFC output is provided by local inhibitory interneurons, yet how drinking affects interneuron physiology remains poorly understood, particularly in female individuals. To address this gap, we generated fluorescent reporter transgenic mice to label the two major classes of interneuron in deep layer prelimbic PFC, based on expression of parvalbumin (PV-IN) or somatostatin (SST-IN). We then interrogated PV-IN and SST-IN membrane and synaptic physiology in a rodent model of binge drinking. Beginning in late adolescence, mice received 3-4 weeks of intermittent access (IA) ethanol. We prepared acute brain slices one day after the last drinking session. PV-INs but not SST-INs from IA ethanol mice displayed increased excitability relative to controls, regardless of sex. On the contrary, synaptic adaptations to PV-INs differed based on sex. While drinking decreased excitatory synaptic strength onto PV-INs from female mice, PV-INs from IA ethanol male mice exhibited potentiated excitatory transmission relative to controls. In contrast, decreased synaptic strength onto SST-INs was observed following IA ethanol in all groups of mice. Together, these findings illustrate novel sex differences in drinking-related PFC pathophysiology. Discovering means to restore PV-IN and SST-IN dysfunction following extended drinking provides opportunities for developing new treatments for all AUD patients.
Collapse
Affiliation(s)
- Max E Joffe
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA.
| | - Danny G Winder
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Warren Center for Neuroscience Drug Discovery, Nashville, TN, 37232, USA; Vanderbilt Center for Addiction Research, Nashville, TN, 37232, USA
| |
Collapse
|
26
|
Taylor CP, Harris EW. Analgesia with Gabapentin and Pregabalin May Involve N-Methyl-d-Aspartate Receptors, Neurexins, and Thrombospondins. J Pharmacol Exp Ther 2020; 374:161-174. [DOI: 10.1124/jpet.120.266056] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/17/2020] [Indexed: 11/22/2022] Open
|
27
|
McCabe MP, Cullen ER, Barrows CM, Shore AN, Tooke KI, Laprade KA, Stafford JM, Weston MC. Genetic inactivation of mTORC1 or mTORC2 in neurons reveals distinct functions in glutamatergic synaptic transmission. eLife 2020; 9:e51440. [PMID: 32125271 PMCID: PMC7080408 DOI: 10.7554/elife.51440] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Although mTOR signaling is known as a broad regulator of cell growth and proliferation, in neurons it regulates synaptic transmission, which is thought to be a major mechanism through which altered mTOR signaling leads to neurological disease. Although previous studies have delineated postsynaptic roles for mTOR, whether it regulates presynaptic function is largely unknown. Moreover, the mTOR kinase operates in two complexes, mTORC1 and mTORC2, suggesting that mTOR's role in synaptic transmission may be complex-specific. To better understand their roles in synaptic transmission, we genetically inactivated mTORC1 or mTORC2 in cultured mouse glutamatergic hippocampal neurons. Inactivation of either complex reduced neuron growth and evoked EPSCs (eEPSCs), however, the effects of mTORC1 on eEPSCs were postsynaptic and the effects of mTORC2 were presynaptic. Despite postsynaptic inhibition of evoked release, mTORC1 inactivation enhanced spontaneous vesicle fusion and replenishment, suggesting that mTORC1 and mTORC2 differentially modulate postsynaptic responsiveness and presynaptic release to optimize glutamatergic synaptic transmission.
Collapse
Affiliation(s)
- Matthew P McCabe
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Erin R Cullen
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Caitlynn M Barrows
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Amy N Shore
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Katherine I Tooke
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Kathryn A Laprade
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - James M Stafford
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| | - Matthew C Weston
- University of Vermont, Department of Neurological SciencesBurlingtonUnited States
| |
Collapse
|
28
|
Ge D, Noakes PG, Lavidis NA. What are Neurotransmitter Release Sites and Do They Interact? Neuroscience 2020; 425:157-168. [DOI: 10.1016/j.neuroscience.2019.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022]
|
29
|
(2R,6R)-hydroxynorketamine rapidly potentiates hippocampal glutamatergic transmission through a synapse-specific presynaptic mechanism. Neuropsychopharmacology 2020; 45:426-436. [PMID: 31216563 PMCID: PMC6901515 DOI: 10.1038/s41386-019-0443-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/17/2019] [Accepted: 06/11/2019] [Indexed: 12/18/2022]
Abstract
Preclinical studies indicate that (2R,6R)-hydroxynorketamine (HNK) retains the rapid and sustained antidepressant-like actions of ketamine, but is spared its dissociative-like properties and abuse potential. While (2R,6R)-HNK is thought to exert its antidepressant-like effects by potentiating α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission, it is unknown how it exerts this effect. The acute synaptic effects of (2R,6R)-HNK were examined by recording field excitatory postsynaptic potentials (fEPSPs) and miniature excitatory postsynaptic currents (mEPSCs) in rat hippocampal slices. (2R,6R)-HNK bath application caused a rapid and persistent potentiation of AMPAR-mediated Schaffer collateral (SC)-CA1 fEPSPs in slices derived from male and female rats. The (2R,6R)-HNK-induced potentiation occurred independent of N-methyl-D-aspartate receptor (NMDAR) activity, was accompanied by a concentration-dependent decrease in paired pulse ratios, and was occluded by raising glutamate release probability. In additon, in the presence of tetrodotoxin, (2R,6R)-HNK increased the frequency, but not amplitude, of mEPSC events, confirming a presynaptic site of action that is independent of glutamatergic network disinhibition. A dual extracellular recording configuration revealed that the presynaptic effects of (2R,6R)-HNK were synapse-selective, occurring in CA1-projecting SC terminals, but not in CA1-projecting temporoammonic terminals. Overall, we found that (2R,6R)-HNK enhances excitatory synaptic transmission in the hippocampus through a concentration-dependent, NMDAR-independent, and synapse-selective increase in glutamate release probability with no direct actions on AMPAR function. These findings provide novel insight regarding (2R,6R)-HNK's acute mechanism of action, and may inform novel antidepressant drug mechanisms that could yield superior efficacy, safety, and tolerability.
Collapse
|
30
|
Bencsik N, Pusztai S, Borbély S, Fekete A, Dülk M, Kis V, Pesti S, Vas V, Szűcs A, Buday L, Schlett K. Dendritic spine morphology and memory formation depend on postsynaptic Caskin proteins. Sci Rep 2019; 9:16843. [PMID: 31727973 PMCID: PMC6856520 DOI: 10.1038/s41598-019-53317-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
CASK-interactive proteins, Caskin1 and Caskin2, are multidomain neuronal scaffold proteins. Recent data from Caskin1 knockout animals indicated only a mild role of Caskin1 in anxiety and pain perception. In this work, we show that deletion of both Caskins leads to severe deficits in novelty recognition and spatial memory. Ultrastructural analyses revealed a reduction in synaptic profiles and dendritic spine areas of CA1 hippocampal pyramidal neurons of double knockout mice. Loss of Caskin proteins impaired LTP induction in hippocampal slices, while miniature EPSCs in dissociated hippocampal cultures appeared to be unaffected. In cultured Caskin knockout hippocampal neurons, overexpressed Caskin1 was enriched in dendritic spine heads and increased the amount of mushroom-shaped dendritic spines. Chemically induced LTP (cLTP) mediated enlargement of spine heads was augmented in the knockout mice and was not influenced by Caskin1. Immunocytochemistry and immunoprecipitation confirmed that Shank2, a master scaffold of the postsynaptic density, and Caskin1 co-localized within the same complex. Phosphorylation of AMPA receptors was specifically altered by Caskin deficiency and was not elevated by cLTP treatment further. Taken together, our results prove a previously unnoticed postsynaptic role of Caskin scaffold proteins and indicate that Caskins influence learning abilities via regulating spine morphology and AMPA receptor localisation.
Collapse
Affiliation(s)
- Norbert Bencsik
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Szilvia Pusztai
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Sándor Borbély
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.,Institute of Cognitive Neuroscience and Psychology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Fekete
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Metta Dülk
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Viktor Kis
- Department Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Szabolcs Pesti
- Department Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Virág Vas
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Attila Szűcs
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - László Buday
- Institute of Enzymology, Research Centre of Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Department Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
31
|
Modeling a Neurexin-3α Human Mutation in Mouse Neurons Identifies a Novel Role in the Regulation of Transsynaptic Signaling and Neurotransmitter Release at Excitatory Synapses. J Neurosci 2019; 39:9065-9082. [PMID: 31578233 DOI: 10.1523/jneurosci.1261-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/13/2019] [Accepted: 09/22/2019] [Indexed: 12/27/2022] Open
Abstract
Presynaptic α-neurexins are highly expressed and more frequently linked to neuropsychiatric and neurodevelopmental disorders than β-neurexins. However, how extracellular sequences specific to α-neurexins enable synaptic transmission is poorly understood. We identified a mutation in an extracellular region of neurexin-3α (A687T), located in a region conserved among α-neurexins and throughout vertebrate evolution, in a patient diagnosed with profound intellectual disability and epilepsy. We systematically interrogated this mutation using a knockdown-replacement approach, and discovered that the A687T mutation enhanced presynaptic morphology and increased two critical presynaptic parameters: (1) presynaptic release probability, and (2) the size of the readily releasable pool exclusively at excitatory synapses in mixed sex primary mouse hippocampal cultures. Introduction of the mutation in vivo and subsequent analysis in ex vivo brain slices made from male and female mice revealed a significant increase in excitatory presynaptic neurotransmission that occluded presynaptic but not postsynaptic LTP. Mechanistically, neurexin-3αA687T enhanced binding to LRRTM2 without altering binding to postsynaptic neuroligin-1. Thus, neurexin-3αA687T unexpectedly produced the first neurexin presynaptic gain-of-function phenotype and revealed unanticipated novel insights into how α-neurexin extracellular sequences govern both transsynaptic adhesion and presynaptic neurotransmitter release.SIGNIFICANCE STATEMENT Despite decades of scientific scrutiny, how precise α-neurexin extracellular sequences control synapse function remains enigmatic. One largely unpursued avenue to identify the role of precise extracellular sequences is the interrogation of naturally occurring missense mutations. Here, we identified a neurexin-3α missense mutation in a compound heterozygous patient diagnosed with profound intellectual disability and epilepsy and systematically interrogated this mutation. Using in vitro and in vivo molecular replacement, electrophysiology, electron microscopy, and structure-function analyses, we reveal a novel role for neurexin-3α, unanticipated based on α-neurexin knock-out models, in controlling presynaptic morphology and neurotransmitter release at excitatory synapses. Our findings represent the first neurexin gain-of-function phenotype and provide new fundamentally important insight into the synaptic biology of α-neurexins.
Collapse
|
32
|
Formisano R, Mersha MD, Caplan J, Singh A, Rankin CH, Tavernarakis N, Dhillon HS. Synaptic vesicle fusion is modulated through feedback inhibition by dopamine auto-receptors. Synapse 2019; 74:e22131. [PMID: 31494966 DOI: 10.1002/syn.22131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 01/11/2023]
Abstract
Mechanisms of synaptic vesicular fusion and neurotransmitter clearance are highly controlled processes whose finely-tuned regulation is critical for neural function. This modulation has been suggested to involve pre-synaptic auto-receptors; however, their underlying mechanisms of action remain unclear. Previous studies with the well-defined C. elegans nervous system have used functional imaging to implicate acid sensing ion channels (ASIC-1) to describe synaptic vesicle fusion dynamics within its eight dopaminergic neurons. Implementing a similar imaging approach with a pH-sensitive fluorescent reporter and fluorescence resonance after photobleaching (FRAP), we analyzed dynamic imaging data collected from individual synaptic termini in live animals. We present evidence that constitutive fusion of neurotransmitter vesicles on dopaminergic synaptic termini is modulated through DOP-2 auto-receptors via a negative feedback loop. Integrating our previous results showing the role of ASIC-1 in a positive feedback loop, we also put forth an updated model for synaptic vesicle fusion in which, along with DAT-1 and ASIC-1, the dopamine auto-receptor DOP-2 lies at a modulatory hub at dopaminergic synapses. Our findings are of potential broader significance as similar mechanisms are likely to be used by auto-receptors for other small molecule neurotransmitters across species.
Collapse
Affiliation(s)
- Rosaria Formisano
- Department of Biological Sciences, Delaware State University, Dover, Delaware
| | - Mahlet D Mersha
- Department of Biological Sciences, Delaware State University, Dover, Delaware
| | - Jeff Caplan
- Delaware Biotechnology Institute, University of Delaware, Newark, Delaware
| | - Abhyudai Singh
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, Delaware
| | - Catharine H Rankin
- Department of Psychology and DM Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Heraklion, Greece
| | - Harbinder S Dhillon
- Department of Biological Sciences, Delaware State University, Dover, Delaware
| |
Collapse
|
33
|
Metzbower SR, Joo Y, Benavides DR, Blanpied TA. Properties of Individual Hippocampal Synapses Influencing NMDA-Receptor Activation by Spontaneous Neurotransmission. eNeuro 2019; 6:ENEURO.0419-18.2019. [PMID: 31110134 PMCID: PMC6541874 DOI: 10.1523/eneuro.0419-18.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/05/2019] [Accepted: 05/12/2019] [Indexed: 12/14/2022] Open
Abstract
NMDA receptor (NMDAR) activation is critical for maintenance and modification of synapse strength. Specifically, NMDAR activation by spontaneous glutamate release has been shown to mediate some forms of synaptic plasticity as well as synaptic development. Interestingly, there is evidence that within individual synapses each release mode may be segregated such that postsynaptically there are distinct pools of responsive receptors. To examine potential regulators of NMDAR activation because of spontaneous glutamate release in cultured hippocampal neurons, we used GCaMP6f imaging at single synapses in concert with confocal and super-resolution imaging. Using these single-spine approaches, we found that Ca2+ entry activated by spontaneous release tends to be carried by GluN2B-NMDARs. Additionally, the amount of NMDAR activation varies greatly both between synapses and within synapses, and is unrelated to spine and synapse size, but does correlate loosely with synapse distance from the soma. Despite the critical role of spontaneous activation of NMDARs in maintaining synaptic function, their activation seems to be controlled factors other than synapse size or synapse distance from the soma. It is most likely that NMDAR activation by spontaneous release influenced variability in subsynaptic receptor position, release site position, vesicle content, and channel properties. Therefore, spontaneous activation of NMDARs appears to be regulated distinctly from other receptor types, notably AMPARs, within individual synapses.
Collapse
Affiliation(s)
| | - Yuyoung Joo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | | |
Collapse
|
34
|
Silm K, Yang J, Marcott PF, Asensio CS, Eriksen J, Guthrie DA, Newman AH, Ford CP, Edwards RH. Synaptic Vesicle Recycling Pathway Determines Neurotransmitter Content and Release Properties. Neuron 2019; 102:786-800.e5. [PMID: 31003725 PMCID: PMC6541489 DOI: 10.1016/j.neuron.2019.03.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 01/28/2019] [Accepted: 03/19/2019] [Indexed: 01/03/2023]
Abstract
In contrast to temporal coding by synaptically acting neurotransmitters such as glutamate, neuromodulators such as monoamines signal changes in firing rate. The two modes of signaling have been thought to reflect differences in release by different cells. We now find that midbrain dopamine neurons release glutamate and dopamine with different properties that reflect storage in different synaptic vesicles. The vesicles differ in release probability, coupling to presynaptic Ca2+ channels and frequency dependence. Although previous work has attributed variation in these properties to differences in location or cytoskeletal association of synaptic vesicles, the release of different transmitters shows that intrinsic differences in vesicle identity drive different modes of release. Indeed, dopamine but not glutamate vesicles depend on the adaptor protein AP-3, revealing an unrecognized linkage between the pathway of synaptic vesicle recycling and the properties of exocytosis. Storage of the two transmitters in different vesicles enables the transmission of distinct signals.
Collapse
Affiliation(s)
- Kätlin Silm
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Jing Yang
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Pamela F Marcott
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Cedric S Asensio
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Jacob Eriksen
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA
| | - Daryl A Guthrie
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes of Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Amy H Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institutes of Drug Abuse - Intramural Research Program, Baltimore, MD 21224, USA
| | - Christopher P Ford
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Robert H Edwards
- Departments of Neurology and Physiology, Graduate Programs in Neuroscience and Cell Biology, Kavli Institute for Fundamental Neuroscience, Weill Institute for the Neurosciences, UCSF School of Medicine, San Francisco, CA 94143, USA.
| |
Collapse
|
35
|
Conditional Deletion of CC2D1A Reduces Hippocampal Synaptic Plasticity and Impairs Cognitive Function through Rac1 Hyperactivation. J Neurosci 2019; 39:4959-4975. [PMID: 30992372 DOI: 10.1523/jneurosci.2395-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 11/21/2022] Open
Abstract
Coiled-coil and C2 domain containing 1A (CC2D1A) is an evolutionarily conserved protein, originally identified as a nuclear factor-κB activator through a large-scale screen of human genes. Mutations in the human Cc2d1a gene result in autosomal recessive nonsyndromic intellectual disability. It remains unclear, however, how Cc2d1a mutation leads to alterations in brain function. Here, we have taken advantage of Cre/loxP recombinase-based strategy to conditionally delete Cc2d1a exclusively from excitatory neurons of male mouse forebrain to examine its role in hippocampal synaptic plasticity and cognitive function. We confirmed the expression of CC2D1A protein and mRNA in the mouse hippocampus. Double immunofluorescence staining showed that CC2D1A is expressed in both excitatory and inhibitory neurons of the adult hippocampus. Conditional deletion of Cc2d1a (cKO) from excitatory neurons leads to impaired performance in object location memory test and altered anxiety-like behavior. Consistently, cKO mice displayed a deficit in the maintenance of LTP in the CA1 region of hippocampal slices. Cc2d1a deletion also resulted in decreased complexity of apical and basal dendritic arbors of CA1 pyramidal neurons. An enhanced basal Rac1 activity was observed following Cc2d1a deletion, and this enhancement was mediated by reduced SUMO-specific protease 1 (SENP1) and SENP3 expression, thus increasing the amount of Rac1 SUMOylation. Furthermore, partial blockade of Rac1 activity rescued impairments in LTP and object location memory performance in cKO mice. Together, our results implicate Rac1 hyperactivity in synaptic plasticity and cognitive deficits observed in Cc2d1a cKO mice and reveal a novel role for CC2D1A in regulating hippocampal synaptic function.SIGNIFICANCE STATEMENT CC2D1A is abundantly expressed in the brain, but there is little known about its physiological function. Taking advantage of Cc2d1a cKO mice, the present study highlights the importance of CC2D1A in the maintenance of LTP at Schaffer collateral-CA1 synapses and the formation of hippocampus-dependent long-term object location memory. Our findings establish a critical link between elevated Rac1 activity, structural and synaptic plasticity alterations, and cognitive impairment caused by Cc2d1a deletion. Moreover, partial blockade of Rac1 activity rescues synaptic plasticity and memory deficits in Cc2d1a cKO mice. Such insights may have implications for the utility of Rac1 inhibitors in the treatment of intellectual disability caused by Cc2d1a mutations in human patients.
Collapse
|
36
|
A Poly-Glutamine Region in the Drosophila VAChT Dictates Fill-Level of Cholinergic Synaptic Vesicles. eNeuro 2019; 6:eN-NWR-0477-18. [PMID: 30847389 PMCID: PMC6402538 DOI: 10.1523/eneuro.0477-18.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 01/08/2023] Open
Abstract
While the primary role of vesicular transporters is to load neurotransmitters into synaptic vesicles (SVs), accumulating evidence suggests that these proteins also contribute to additional aspects of synaptic function, including vesicle release. In this study, we extend the role of the VAChT to include regulating the transmitter content of SVs. We report that manipulation of a C-terminal poly-glutamine (polyQ) region in the Drosophila VAChT is sufficient to influence transmitter content, and release frequency, of cholinergic vesicles from the terminals of premotor interneurons. Specifically, we find that reduction of the polyQ region, by one glutamine residue (13Q to 12Q), results in a significant increase in both amplitude and frequency of spontaneous cholinergic miniature EPSCs (mEPSCs) recorded in the aCC and RP2 motoneurons. Moreover, this truncation also results in evoked synaptic currents that show increased duration: consistent with increased ACh release. By contrast, extension of the polyQ region by one glutamine (13Q to 14Q) is sufficient to reduce mEPSC amplitude and frequency and, moreover, prevents evoked SV release. Finally, a complete deletion of the polyQ region (13Q to 0Q) has no obvious effects to mEPSCs, but again evoked synaptic currents show increased duration. The mechanisms that ensure SVs are filled to physiologically-appropriate levels remain unknown. Our study identifies the polyQ region of the insect VAChT to be required for correct vesicle transmitter loading and, thus, provides opportunity to increase understanding of this critical aspect of neurotransmission.
Collapse
|
37
|
Postnikova TY, Trofimova AM, Ergina JL, Zubareva OE, Kalemenev SV, Zaitsev AV. Transient Switching of NMDA-Dependent Long-Term Synaptic Potentiation in CA3-CA1 Hippocampal Synapses to mGluR 1-Dependent Potentiation After Pentylenetetrazole-Induced Acute Seizures in Young Rats. Cell Mol Neurobiol 2019; 39:287-300. [PMID: 30607810 DOI: 10.1007/s10571-018-00647-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/29/2018] [Indexed: 01/18/2023]
Abstract
The mechanisms of impairment in long-term potentiation after status epilepticus (SE) remain unclear. We investigated the properties of LTP induced by theta-burst stimulation in hippocampal slices of rats 3 h and 1, 3, and 7 days after SE. Seizures were induced in 3-week old rats by a single injection of pentylenetetrazole (PTZ). Only animals with generalized seizures lasting more than 30 min were included in the experiments. The results revealed that LTP was strongly attenuated in the CA1 hippocampal area after PTZ-induced SE as compared with that in control animals. Saturation of synaptic responses following epileptic activity does not explain weakening of LTP because neither the quantal size of the excitatory responses nor the slopes of the input-output curves for field excitatory postsynaptic potentials changed in the post-SE rats. After PTZ-induced SE, NMDA-dependent LTP was suppressed, and LTP transiently switched to the mGluR1-dependent form. This finding does not appear to have been reported previously in the literature. An antagonist of NMDA receptors, D-2-amino-5-phosphonovalerate, did not block LTP induction in 3-h and 1-day post-SE slices. An antagonist of mGluR1, FTIDS, completely prevented LTP in 1-day post-SE slices; whereas it did not affect LTP induction in control and post-SE slices at the other studied times. mGluR1-dependent LTP was postsynaptically expressed and did not require NMDA receptor activation. Recovery of NMDA-dependent LTP occurred 7 day after SE. Transient switching between NMDA-dependent LTP and mGluR1-dependent LTP could play a role in the pathogenesis of acquired epilepsy.
Collapse
Affiliation(s)
- Tatyana Y Postnikova
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia
- Peter the Great St.Petersburg Polytechnic University (SPbPU), Saint Petersburg, Russia
| | - Alina M Trofimova
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia
| | - Julia L Ergina
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia
| | - Olga E Zubareva
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia
| | - Sergey V Kalemenev
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia
| | - Aleksey V Zaitsev
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry of RAS (IEPhB), Saint Petersburg, Russia.
- Peter the Great St.Petersburg Polytechnic University (SPbPU), Saint Petersburg, Russia.
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia.
| |
Collapse
|
38
|
Liu H, Li L, Nedelcu D, Hall Q, Zhou L, Wang W, Yu Y, Kaplan JM, Hu Z. Heterodimerization of UNC-13/RIM regulates synaptic vesicle release probability but not priming in C. elegans. eLife 2019; 8:40585. [PMID: 30802206 PMCID: PMC6389284 DOI: 10.7554/elife.40585] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/03/2019] [Indexed: 12/17/2022] Open
Abstract
UNC-13 proteins play an essential role in synaptic transmission by recruiting synaptic vesicles (SVs) to become available for release, which is termed SV priming. Here we show that the C2A domain of UNC-13L, like the corresponding domain in mammalian Munc13-1, displays two conserved binding modes: forming C2A/C2A homodimers, or forming a heterodimer with the zinc finger domain of UNC-10/RIM (C2A/RIM). Functional analysis revealed that UNC-13L’s C2A promotes synaptic transmission by regulating a post-priming process. Stimulus-evoked release but not SV priming, was impaired in unc-10 mutants deficient for C2A/RIM heterodimerization, leading to decreased release probability. Disrupting C2A/C2A homodimerization in UNC-13L-rescued animals had no effect on synaptic transmission, but fully restored the evoked release and the release probability of unc-10/RIM mutants deficient for C2A/RIM heterodimerization. Thus, our results support the model that RIM binding C2A releases UNC-13L from an autoinhibitory homodimeric complex to become fusion-competent by functioning as a switch only.
Collapse
Affiliation(s)
- Haowen Liu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Lei Li
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Daniel Nedelcu
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Qi Hall
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States.,Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Lijun Zhou
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Howard Hughes Medical Institute, Boston, United States
| | - Wei Wang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Yi Yu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Joshua M Kaplan
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Zhitao Hu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
39
|
A Photoactivatable Botulinum Neurotoxin for Inducible Control of Neurotransmission. Neuron 2019; 101:863-875.e6. [PMID: 30704911 DOI: 10.1016/j.neuron.2019.01.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/14/2018] [Accepted: 12/28/2018] [Indexed: 12/18/2022]
Abstract
Regulated secretion is critical for diverse biological processes ranging from immune and endocrine signaling to synaptic transmission. Botulinum and tetanus neurotoxins, which specifically proteolyze vesicle fusion proteins involved in regulated secretion, have been widely used as experimental tools to block these processes. Genetic expression of these toxins in the nervous system has been a powerful approach for disrupting neurotransmitter release within defined circuitry, but their current utility in the brain and elsewhere remains limited by lack of spatial and temporal control. Here we engineered botulinum neurotoxin B so that it can be activated with blue light. We demonstrate the utility of this approach for inducibly disrupting excitatory neurotransmission, providing a first-in-class optogenetic tool for persistent, light-triggered synaptic inhibition. In addition to blocking neurotransmitter release, this approach will have broad utility for conditionally disrupting regulated secretion of diverse bioactive molecules, including neuropeptides, neuromodulators, hormones, and immune molecules. VIDEO ABSTRACT.
Collapse
|
40
|
Yee AG, Forbes B, Cheung PY, Martini A, Burrell MH, Freestone PS, Lipski J. Action potential and calcium dependence of tonic somatodendritic dopamine release in the Substantia Nigra pars compacta. J Neurochem 2018; 148:462-479. [PMID: 30203851 DOI: 10.1111/jnc.14587] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/09/2018] [Accepted: 09/04/2018] [Indexed: 12/31/2022]
Abstract
Despite the importance of somatodendritic dopamine (DA) release in the Substantia Nigra pars compacta (SNc), its mechanism remains poorly understood. Using a novel approach combining fast-scan controlled-adsorption voltammetry (FSCAV) and single-unit electrophysiology, we have investigated the mechanism of somatodendritic release by directly correlating basal (non-stimulated) extracellular DA concentration ([DA]out ), with pharmacologically-induced changes of firing of nigral dopaminergic neurons in rat brain slices. FSCAV measurements indicated that basal [DA]out in the SNc was 40.7 ± 2.0 nM (at 34 ± 0.5°C), which was enhanced by amphetamine, cocaine, and L-DOPA, and reduced by VMAT2 inhibitor, Ro4-1284. Complete inhibition of firing by TTX decreased basal [DA]out , but this reduction was smaller than the effect of D2 receptor agonist, quinpirole. Despite similar effects on neuronal firing, the larger decrease in [DA]out evoked by quinpirole was attributed to cell membrane hyperpolarization and greater reduction in cytosolic free Ca2+ ([Ca2+ ]in ). Decreasing extracellular Ca2+ also reduced basal [DA]out , despite increasing firing frequency. Furthermore, inhibiting L-type Ca2+ channels decreased basal [DA]out , although specific Cav 1.3 channel inhibition did not affect firing rate. Inhibition of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) also decreased [DA]out , demonstrating the importance of intracellular Ca2+ stores for somatodendritic release. Finally, in vivo FSCAV measurements showed that basal [DA]out in the SNc was 79.8 ± 10.9 nM in urethane-anesthetized rats, which was enhanced by amphetamine. Overall, our findings indicate that although tonic somatodendritic DA release is largely independent of action potentials, basal [DA]out is strongly regulated by voltage-dependent Ca2+ influx and release of intracellular Ca2+ . OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Andrew G Yee
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Blaze Forbes
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Pang-Ying Cheung
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | | | - Mark H Burrell
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Peter S Freestone
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Janusz Lipski
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
41
|
Estrada-Sánchez AM, Castro D, Portillo-Ortiz K, Jang K, Nedjat-Haiem M, Levine MS, Cepeda C. Complete but not partial inhibition of glutamate transporters exacerbates cortical excitability in the R6/2 mouse model of Huntington's disease. CNS Neurosci Ther 2018; 25:509-518. [PMID: 30311425 DOI: 10.1111/cns.13070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 01/05/2023] Open
Abstract
AIM Deficient glutamate reuptake occurs in the cerebral cortex of Huntington's disease (HD) patients and murine models. Here, we examine the effects of partial or complete blockade of glutamate transporters on excitatory postsynaptic currents (EPSCs) of cortical pyramidal neurons (CPNs). METHODS Whole-cell patch clamp recordings of CPNs in slices from symptomatic R6/2 mice and wild-type (WT) littermates were used to examine the effects of selective or concurrent inhibition of glutamate reuptake transporters. RESULTS Selective inhibition of the glial glutamate transporter 1 (GLT-1) or the glutamate aspartate transporter (GLAST) produced slight decreases in decay time of evoked EPSCs in CPNs from WT and R6/2 mice with no significant differences between genotypes. In contrast, concurrent inhibition of both transporters with DL-TBOA induced a significant increase in area and decay time and this effect was significantly greater in R6/2 CPNs. Furthermore, full blockade also reduced spontaneous EPSC frequency and exacerbated epileptiform activity in CPNs from symptomatic R6/2 mice. CONCLUSIONS R6/2 CPNs are more sensitive to glutamate accumulation during full inhibition of both glutamate transporters, and these neurons have homeostatic mechanisms to cope with inhibition of GLT-1 or GLAST by a mechanism that involves upregulation of either transporter when the other is deficient.
Collapse
Affiliation(s)
- Ana María Estrada-Sánchez
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Daniel Castro
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Kenia Portillo-Ortiz
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Katrina Jang
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Michael Nedjat-Haiem
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Michael S Levine
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
42
|
Sculpting neurotransmission during synaptic development by 2D nanostructured interfaces. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2521-2532. [DOI: 10.1016/j.nano.2017.01.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/08/2016] [Accepted: 01/04/2017] [Indexed: 11/23/2022]
|
43
|
He T, Nitabach MN, Lnenicka GA. Parvalbumin expression affects synaptic development and physiology at the Drosophila larval NMJ. J Neurogenet 2018; 32:209-220. [PMID: 30175644 DOI: 10.1080/01677063.2018.1498496] [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] [Indexed: 10/28/2022]
Abstract
Presynaptic Ca2+ appears to play multiple roles in synaptic development and physiology. We examined the effect of buffering presynaptic Ca2+ by expressing parvalbumin (PV) in Drosophila neurons, which do not normally express PV. The studies were performed on the identified Ib terminal that innervates muscle fiber 5. The volume-averaged, residual Ca2+ resulting from single action potentials (APs) and AP trains was measured using the fluorescent Ca2+ indicator, OGB-1. PV reduced the amplitude and decay time constant (τ) for single-AP Ca2+ transients. For AP trains, there was a reduction in the rate of rise and decay of [Ca2+]i but the plateau [Ca2+]i was not affected. Electrophysiological recordings from muscle fiber 5 showed a reduction in paired-pulse facilitation, particularly the F1 component; this was likely due to the reduction in residual Ca2+. These synapses also showed reduced synaptic enhancement during AP trains, presumably due to less buildup of synaptic facilitation. The transmitter release for single APs was increased for the PV-expressing terminals and this may have been a homeostatic response to the decrease in facilitation. Confocal microscopy was used to examine the structure of the motor terminals and PV expression resulted in smaller motor terminals with fewer synaptic boutons and active zones. This result supports earlier proposals that increased AP activity promotes motor terminal growth through increases in presynaptic [Ca2+]i.
Collapse
Affiliation(s)
- Tao He
- a Division of Pulmonary and Critical Care Medicine , David Geffen School of Medicine at UCLA , Los Angeles , CA , USA
| | - Michael N Nitabach
- b Department of Cellular and Molecular Physiology , Yale School of Medicine , New Haven , CT , USA
| | - Gregory A Lnenicka
- c Department of Biological Sciences , University at Albany , Albany , NY , USA
| |
Collapse
|
44
|
Courtney NA, Briguglio JS, Bradberry MM, Greer C, Chapman ER. Excitatory and Inhibitory Neurons Utilize Different Ca 2+ Sensors and Sources to Regulate Spontaneous Release. Neuron 2018; 98:977-991.e5. [PMID: 29754754 DOI: 10.1016/j.neuron.2018.04.022] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 03/19/2018] [Accepted: 04/17/2018] [Indexed: 01/16/2023]
Abstract
Spontaneous neurotransmitter release (mini) is an important form of Ca2+-dependent synaptic transmission that occurs in the absence of action potentials. A molecular understanding of this process requires an identification of the underlying Ca2+ sensors. Here, we address the roles of the relatively low- and high-affinity Ca2+ sensors, synapotagmin-1 (syt1) and Doc2α/β, respectively. We found that both syt1 and Doc2 regulate minis, but, surprisingly, their relative contributions depend on whether release was from excitatory or inhibitory neurons. Doc2α promoted glutamatergic minis, while Doc2β and syt1 both regulated GABAergic minis. We identified Ca2+ ligand mutations in Doc2 that either disrupted or constitutively activated the regulation of minis. Finally, Ca2+ entry via voltage-gated Ca2+ channels triggered miniature GABA release by activating syt1, but had no effect on Doc2-driven minis. This work reveals an unexpected divergence in the regulation of spontaneous excitatory and inhibitory transmission in terms of both Ca2+ sensors and sources.
Collapse
Affiliation(s)
- Nicholas A Courtney
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Joseph S Briguglio
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Mazdak M Bradberry
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Christina Greer
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Edwin R Chapman
- Department of Neuroscience and Howard Hughes Medical Institute, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI 53705, USA.
| |
Collapse
|
45
|
Ge D, Lavidis N. Climatic modulation of neurotransmitter release in amphibian neuromuscular junctions: role of dynorphin-A. Am J Physiol Regul Integr Comp Physiol 2018; 314:R716-R723. [DOI: 10.1152/ajpregu.00263.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amphibian neuromuscular junctions (NMJs) become relatively more silent during the dry winter season in Australia. During the dry, calcium sensitivity is reduced, whereas calcium dependence remains unchanged. Endogenous opioid peptides play an important role in the regulation of the physiological functions of active and dormant vertebrates. Previous findings suggest that dynorphin-A is more potent than other opiates in decreasing evoked neurotransmission in amphibian NMJs. Dynorphin-A has been shown not to alter the amplitude or the frequency of miniature quantal neurotransmitter release. In the present study, we report that dynorphin-A exerted a more pronounced inhibitory effect on evoked neurotransmitter release during the dry (hibernating period) when compared with the wet (active period) season. Dynorphin-A increased the frequency and decreased the amplitude of miniature neurotransmitter release only at relatively high concentration during the dry season. In the present study, we propose that dynorphin-A suppresses evoked neurotransmitter release and thus contraction of skeletal muscles, while allowing subthreshold activation of the NMJ by miniature neurotransmission, thus preventing any significant neuromuscular remodeling. The inhibitory effect of dynorphin-A on evoked transmitter release is reduced by increasing the extracellular calcium concentration.
Collapse
Affiliation(s)
- Dengyun Ge
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Nickolas Lavidis
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
46
|
Liu X, Liu S. Cholecystokinin selectively activates short axon cells to enhance inhibition of olfactory bulb output neurons. J Physiol 2018; 596:2185-2207. [PMID: 29572837 DOI: 10.1113/jp275511] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/15/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Cholecystokinin (CCK) via CCK-B receptors significantly enhances the GABAA receptor-mediated synaptic inhibition of principal olfactory bulb (OB) output neurons. This CCK action requires action potentials in presynaptic neurons. The enhanced inhibition of OB output neurons is a result of CCK-elevated inhibitory input from the glomerular circuit. CCK modulation of the glomerular circuit also leads to potentiated presynaptic inhibition of olfactory nerve terminals and postsynaptic inhibition of glomerular neurons. Selective excitation of short axon cells underlies the CCK-potentiated glomerular inhibition. ABSTRACT Neuropeptides such as cholecystokinin (CCK) are important for many brain functions, including sensory processing. CCK is predominantly present in a subpopulation of excitatory neurons and activation of CCK receptors is implicated in olfactory signal processing in the olfactory bulb (OB). However, the cellular and circuit mechanisms underlying the actions of CCK in the OB remain elusive. In the present study, we characterized the effects of CCK on synaptic inhibition of the principal OB output neurons mitral/tufted cells (MTCs) followed by mechanistic analyses at both circuit and cellular levels. First, we found that CCK via CCK-B receptors enhances the GABAA receptor-mediated spontaneous IPSCs in MTCs. Second, CCK does not affect the action potential independent miniature IPSCs in MTCs. Third, CCK potentiates glomerular inhibition resulting in increased GABAB receptor-mediated presynaptic inhibition of olfactory nerve terminals and enhanced spontaneous IPSCs in MTCs and glomerular neurons. Fourth, CCK enhances miniature IPSCs in the excitatory external tufted cells, although neither in the inhibitory short axon cells (SACs) nor in periglomerular cells (PGCs). Finally, CCK excites all tested SACs and a very small minority of GABAergic neurons in the granule cell layer or in periglomerular cells, but not in deep SACs. These results demonstrate that CCK selectively activates SACs to engage the SAC-formed interglomerular circuit and thus elevates inhibition broadly in the OB glomerular layer. This modulation may prevent the system from saturating in response to a high concentration of odourants or facilitate the detection of weak stimuli by increasing signal-to-noise ratio.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Anatomy & Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shaolin Liu
- Department of Anatomy & Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
47
|
Paeonol promotes hippocampal synaptic transmission: The role of the Kv2.1 potassium channel. Eur J Pharmacol 2018; 827:227-237. [PMID: 29550337 DOI: 10.1016/j.ejphar.2018.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 12/30/2022]
Abstract
Paeonol is a major constituent of the Chinese herb Moutan cortex radices. Recent studies report that paeonol has neuroprotective effects and improves impaired learning and memory. However, its underlying mechanisms by which paeonol contributes to synaptic transmission remain unclear. In this study, we found that paeonol increased the frequency of miniature excitatory postsynaptic currents (mEPSCs) and spontaneous excitatory postsynaptic currents (sEPSCs), but had no effect on the amplitude in rat hippocampal CA1 neurons. Similarly, the acetylcholinesterase (AChE) inhibitor rivastigmine increased the frequency of mEPSCs, but had no effect upon amplitude in rat hippocampal neurons. Rivastigmine also inhibited the delayed outward K+ currents in rat hippocampal CA1 neurons, but had no effect in nucleus ambiguus (NA) neurons. The Kv2 blocker guangxitoxin-1E increased the frequency of both mEPSCs and sEPSCs of rat hippocampal CA1 neurons, without affecting their amplitude. Our results suggest that paeonol and rivastigmine enhance spontaneous presynaptic transmitter release, which may be associated with the inhibition of the hippocampal Kv2 current and with therapeutic potential in neurotransmitter deficits found in Alzheimer's disease (AD). Moreover, our data also show that paeonol protects against Aβ25-35-induced impairment of long-term potentiation (LTP) in mouse hippocampal neurons. However, guangxitoxin-1E failed to potentiate the evoked field excitatory postsynaptic potentials (fEPSPs), LTP and Aβ25-35-induced impairment of LTP. These results indicate that paeonol may has the potential to improve learning and memory in AD. Interestingly, this effect is not involved in the inhibition of the hippocampal Kv2 current.
Collapse
|
48
|
Calahorro F, Izquierdo PG. The presynaptic machinery at the synapse of C. elegans. INVERTEBRATE NEUROSCIENCE : IN 2018; 18:4. [PMID: 29532181 PMCID: PMC5851683 DOI: 10.1007/s10158-018-0207-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/22/2018] [Indexed: 11/17/2022]
Abstract
Synapses are specialized contact sites that mediate information flow between neurons and their targets. Important physical interactions across the synapse are mediated by synaptic adhesion molecules. These adhesions regulate formation of synapses during development and play a role during mature synaptic function. Importantly, genes regulating synaptogenesis and axon regeneration are conserved across the animal phyla. Genetic screens in the nematode Caenorhabditis elegans have identified a number of molecules required for synapse patterning and assembly. C. elegans is able to survive even with its neuronal function severely compromised. This is in comparison with Drosophila and mice where increased complexity makes them less tolerant to impaired function. Although this fact may reflect differences in the function of the homologous proteins in the synapses between these organisms, the most likely interpretation is that many of these components are equally important, but not absolutely essential, for synaptic transmission to support the relatively undemanding life style of laboratory maintained C. elegans. Here, we review research on the major group of synaptic proteins, involved in the presynaptic machinery in C. elegans, showing a strong conservation between higher organisms and highlight how C. elegans can be used as an informative tool for dissecting synaptic components, based on a simple nervous system organization.
Collapse
Affiliation(s)
- Fernando Calahorro
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK.
| | - Patricia G Izquierdo
- Biological Sciences, University of Southampton, Life Sciences Building 85, Southampton, SO17 1BJ, UK
| |
Collapse
|
49
|
Williams CL, Smith SM. Calcium dependence of spontaneous neurotransmitter release. J Neurosci Res 2018; 96:335-347. [PMID: 28699241 PMCID: PMC5766384 DOI: 10.1002/jnr.24116] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 01/14/2023]
Abstract
Spontaneous release of neurotransmitters is regulated by extracellular [Ca2+ ] and intracellular [Ca2+ ]. Curiously, some of the mechanisms of Ca2+ signaling at central synapses are different at excitatory and inhibitory synapses. While the stochastic activity of voltage-activated Ca2+ channels triggers a majority of spontaneous release at inhibitory synapses, this is not the case at excitatory nerve terminals. Ca2+ release from intracellular stores regulates spontaneous release at excitatory and inhibitory terminals, as do agonists of the Ca2+ -sensing receptor. Molecular machinery triggering spontaneous vesicle fusion may differ from that underlying evoked release and may be one of the sources of heterogeneity in release mechanisms.
Collapse
Affiliation(s)
- Courtney L. Williams
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| | - Stephen M. Smith
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
50
|
Wollman LB, Levine RB, Fregosi RF. Developmental plasticity of GABAergic neurotransmission to brainstem motoneurons. J Physiol 2018; 596:5993-6008. [PMID: 29352468 DOI: 10.1113/jp274923] [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] [Received: 07/13/2017] [Accepted: 01/16/2018] [Indexed: 01/15/2023] Open
Abstract
KEY POINTS Critical homeostatic behaviours such as suckling, swallowing and breathing depend on the precise control of tongue muscle activity. Perinatal nicotine exposure has multiple effects on baseline inhibitory GABAergic neurotransmission to hypoglossal motoneurons (XIIMNs), consistent with homeostatic compensations directed at maintaining normal motoneuron output. Developmental nicotine exposure (DNE) alters how GABAergic neurotransmission is modulated by acute activation of nicotinic acetylcholine receptors, which may provide insight into mechanisms by which nicotine exposure alters motor function under conditions that result in increased release of GABA, such as hypoxia, or endogenous acetylcholine, as occurs in the transition from NREM to REM sleep, or in response to exogenous nicotine. ABSTRACT Nicotinic acetylcholine receptor (nAChR) signalling regulates neuronal differentiation and synaptogenesis. Here we test the hypothesis that developmental nicotine exposure (DNE) disrupts the development of GABAergic synaptic transmission to hypoglossal motoneurons (XIIMNs). GABAergic spontaneous and miniature inhibitory postsynaptic currents (sIPSCs/mIPSCs) were recorded from XIIMNs in brainstem slices from control and DNE rat pups of either sex, 1-5 days old, at baseline and following acute stimulation of nAChRs with nicotine. At baseline, sIPSCs were less frequent and smaller in DNE cells (consistent with decreased action potential-mediated GABA release), and mIPSCs were more frequent (consistent with increased vesicular GABA release from presynaptic terminals). Acute nicotine challenge increased sIPSC frequency in both groups, though the increase was greater in DNE cells. Acute nicotine challenge did not change the frequency of mIPSCs in either group, though mIPSC amplitude increased significantly in DNE cells, but not control cells. Stimulation of postsynaptic GABAA receptors with muscimol caused a significantly greater chloride current in DNE cells than in control cells. The increased quantal release of GABA, coupled with the rise in the strength of postsynaptic inhibition may be homeostatic adjustments to the decreased action-potential-mediated input from GABAergic interneurons. However, this will exaggerate synaptic inhibition under conditions where the release of GABA (e.g. hypoxia) or ACh (sleep-wake transitions) is increased. These findings reveal a mechanism that may explain why DNE is associated with deficits in the ability to respond appropriately to chemosensory stimuli or to changes in neuromodulation secondary to changes in central nervous system state.
Collapse
Affiliation(s)
- Lila Buls Wollman
- Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Richard B Levine
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Neuroscience, The University of Arizona, Tucson, AZ, USA
| | - Ralph F Fregosi
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Neuroscience, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|