1
|
Kabeiseman E, Paulsen RT, Burrell BD. Characterization of a Fatty Acid Amide Hydrolase (FAAH) in Hirudo Verbana. Neurochem Res 2024; 49:3015-3029. [PMID: 39093361 PMCID: PMC11450075 DOI: 10.1007/s11064-024-04216-7] [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: 04/15/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
The endocannabinoid system plays a critical role in modulating both peripheral and central nervous system function. Despite being present throughout the animal kingdom, there has been relatively little investigation of the endocannabinoid system beyond traditional animal models. In this study, we report on the identification and characterization of a putative fatty acid amide hydrolase (FAAH) in the medicinal leech, Hirudo verbana. FAAH is the primary enzyme responsible for metabolizing the endocannabinoid signaling molecule arachidonoyl ethanolamide (anandamide or AEA) and therefore plays a critical role in regulating AEA levels in the nervous system. mRNA encoding Hirudo FAAH (HirFAAH) is expressed in the leech central nervous system (CNS) and sequence analysis suggests that this is an orthologue of FAAH-2 observed in vertebrates. Functionally, HirFAAH has serine hydrolase activity based on activity-based protein profiling (ABPP) studies using the fluorophosphonate probe TAMRA-FP. HirFAAH also hydrolyzes arachidonyl 7-amino, 4-methyl coumarin amide (AAMCA), a substrate specific to FAAH. Hydrolase activity during both the ABPP and AAMCA assays was eliminated by a mutation at a conserved catalytic serine. Activity was also blocked by the known FAAH inhibitor, URB597. Treatment of Hirudo ganglia with URB597 potentiated synapses made by the pressure-sensitive mechanosensory neuron (P cell), mimicking the effects of exogenously applied AEA. The Hirudo CNS has been a useful system in which to study properties of endocannabinoid modulation of nociception relevant to vertebrates. Therefore, this characterization of HirFAAH is an important contribution to comparative studies of the endocannabinoid system.
Collapse
Affiliation(s)
- Emily Kabeiseman
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research (CBBRe), Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Riley T Paulsen
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research (CBBRe), Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA
| | - Brian D Burrell
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research (CBBRe), Sanford School of Medicine, University of South Dakota, Vermillion, SD, 57069, USA.
| |
Collapse
|
2
|
Kim JW, Kleinfelter B, Kavalali ET, Monteggia LM. Distinct synaptic mechanisms drive the behavioral response to acute stress and rapid correction by ketamine. Neuropsychopharmacology 2024; 49:1916-1924. [PMID: 38956176 PMCID: PMC11473657 DOI: 10.1038/s41386-024-01908-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/09/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
Prevailing hypotheses on the mechanisms of antidepressant action posit that antidepressants directly counteract deficiencies in major neurotransmitter signaling systems that underlie depression. The rapidly acting antidepressant ketamine has been postulated to correct excess glutamatergic signaling via glutamatergic antagonism leading to the rescue of neuronal structural deficits and reversal of behavioral symptoms. We studied this premise using systemic administration of the acetylcholinesterase inhibitor physostigmine, which has been shown to rapidly elicit a shorter-term period of depressed mood in humans via cholinergic mechanisms. We observed that physostigmine induces acute stress in tandem with long term depression of glutamate release in the hippocampus of mice. However, ketamine rapidly acts to re-establish glutamatergic synaptic efficacy via postsynaptic signaling and behaviorally masks the reduction in passive coping induced by physostigmine. These results underscore the divergence of synaptic signaling mechanisms underlying mood changes and antidepressant action and highlight how distinct synaptic mechanisms may underlie neuropsychiatric disorders versus their treatment.
Collapse
Affiliation(s)
- Ji-Woon Kim
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240, USA
- College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Institute of Regulatory innovation through Science, Kyung Hee University, Seoul, Republic of Korea
| | - Benjamin Kleinfelter
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240, USA
| | - Ege T Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240, USA
| | - Lisa M Monteggia
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240, USA.
| |
Collapse
|
3
|
Park PY, Bleakley LE, Saraya N, Al-Jawahiri R, Eck J, Aloi MA, Melland H, Baker K, Gordon SL. Correlation between evoked neurotransmitter release and adaptive functions in SYT1-associated neurodevelopmental disorder. EBioMedicine 2024; 109:105416. [PMID: 39481209 DOI: 10.1016/j.ebiom.2024.105416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Pathogenic missense variants in the essential synaptic vesicle protein synaptotagmin-1 (SYT1) cause a neurodevelopmental disorder characterised by motor delay and intellectual disability, hyperkinetic movement disorder, episodic agitation, and visual impairments. SYT1 is the presynaptic calcium sensor that triggers synchronous neurotransmitter release. We have previously shown that pathogenic variants around the calcium-sensing region of the critical C2B domain decrease synaptic vesicle exocytosis in neurons. METHODS Here, we have used cultured hippocampal neurons transfected with SYT1-pHluorin to examine how variants within the C2A and C2B domain of SYT1 impact evoked exocytosis. FINDINGS We show that recently identified variants within the facilitatory C2A domain of the protein (L159R, T196K, E209K, E219Q), as well as additional variants in the C2B domain (M303V, S309P, Y365C, G369D), share an underlying pathogenic mechanism, causing a graded and variant-dependent dominant-negative impairment in exocytosis. We establish that the extent of evoked exocytosis observed in vitro in the presence of SYT1 variants correlates with neurodevelopmental impacts of this disorder. Specifically, the severity of motor and communication impairments exhibited by individuals harbouring these variants correlates with multiple measures of exocytic efficiency. INTERPRETATION Together, this suggests that there is a genotype-function-phenotype relationship in SYT1-associated neurodevelopmental disorder, centring impaired evoked neurotransmitter release as a common pathogenic driver. Moreover, this points toward a direct link between control of neurotransmitter release and development of adaptive functions, providing a tractable target for therapeutic amelioration. FUNDING Australian National Health and Medical Research Council, UK Medical Research Council, Great Ormond Street Hospital Children's Charity, University of Melbourne.
Collapse
Affiliation(s)
- Paul Yangho Park
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Lauren Elizabeth Bleakley
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Nadia Saraya
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Reem Al-Jawahiri
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Josefine Eck
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Marc Anthony Aloi
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia
| | - Holly Melland
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia; Neuromedicines Discovery Centre, Monash University, Parkville, 3052, VIC, Australia
| | - Kate Baker
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK; Department of Medical Genetics, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Sarah Louise Gordon
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, 3052, VIC, Australia.
| |
Collapse
|
4
|
Shao C, Zhou WQ, Jia HY, Li G, Ma Y, Zhao M, Wu H, Yang K, Qian X, Zhang MM. Reassessment of capsaicin desensitization in the rodent spinal dorsal horn. Neurochem Int 2024; 181:105887. [PMID: 39437894 DOI: 10.1016/j.neuint.2024.105887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/06/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Capsaicin activates primary afferent transient receptor potential vanilloid 1 (TRPV1) in the spinal dorsal horn and induces exaggerated glutamate release. This capsaicin action is followed by a lasting refractory state referred to as "capsaicin desensitization", which is considered a presynaptic event. In this study, using whole-cell recordings and holographic photostimulation, we reassessed this notion by investigating presynaptic glutamate release and the postsynaptic glutamate response during capsaicin administration. We found that both presynaptic synchronous glutamate release and the postsynaptic glutamate response were largely attenuated in this refractory state; in contrast, asynchronous release was exaggerated. Further behavioral studies revealed a quick increase in the mechanical pain threshold with intrathecal capsaicin administration. Taken together, both presynaptic synchronous glutamate release and the postsynaptic response are downregulated during capsaicin desensitization, and this desensitization may transiently increase the pain threshold. Since both presynaptic synchronous release and postsynaptic glutamate responses are attenuated, the traditional electrophysiological evidence supporting capsaicin desensitization as a presynaptic event should be reassessed.
Collapse
Affiliation(s)
- Caifeng Shao
- Department of Anesthesiology, Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, 226001, China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Wen-Qian Zhou
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Hong-Yi Jia
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Guangying Li
- Department of Psychology, Encephalopathy Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, 130021, China
| | - Yifei Ma
- School of Public Health Class 2025, Capital Medical University, Beijing 100069, China
| | - Mingwei Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Hongjie Wu
- School of Life and Health Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China
| | - Kun Yang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; School of Life and Health Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266113, China; Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Xin Qian
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Ming-Ming Zhang
- Department of Anatomy, Histology and Embryology, K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
5
|
Nascimento F, Özyurt MG, Halablab K, Bhumbra GS, Caron G, Bączyk M, Zytnicki D, Manuel M, Roselli F, Brownstone R, Beato M. Spinal microcircuits go through multiphasic homeostatic compensations in a mouse model of motoneuron degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.10.588918. [PMID: 38645210 PMCID: PMC11030447 DOI: 10.1101/2024.04.10.588918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
In many neurological conditions, early-stage neural circuit adaption can preserve relatively normal behaviour. In some diseases, spinal motoneurons progressively degenerate yet movement is initially preserved. We therefore investigated whether these neurons and associated microcircuits adapt in a mouse model of progressive motoneuron degeneration. Using a combination of in vitro and in vivo electrophysiology and super-resolution microscopy, we found that, early in the disease, neurotransmission in a key pre-motor circuit, the recurrent inhibition mediated by Renshaw cells, is reduced by half due to impaired quantal size associated with decreased glycine receptor density. This impairment is specific, and not a widespread feature of spinal inhibitory circuits. Furthermore, it recovers at later stages of disease. Additionally, an increased probability of release from proprioceptive afferents leads to increased monosynaptic excitation of motoneurons. We reveal that in motoneuron degenerative conditions, spinal microcircuits undergo specific multiphasic homeostatic compensations that may contribute to preservation of force output.
Collapse
Affiliation(s)
- Filipe Nascimento
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - M. Görkem Özyurt
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Kareen Halablab
- Department of Neurology, Ulm University, Ulm, Germany
- German Centre for Neurodegenerative Diseases-Ulm (DZNE-Ulm), Ulm, Germany
| | - Gardave Singh Bhumbra
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
| | - Guillaume Caron
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Marcin Bączyk
- Department of Neurobiology, Poznań University of Physical Education, Poznań, Poland
| | - Daniel Zytnicki
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Université Paris Cité, Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Marin Manuel
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- German Centre for Neurodegenerative Diseases-Ulm (DZNE-Ulm), Ulm, Germany
| | - Rob Brownstone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marco Beato
- Department of Neuroscience Physiology and Pharmacology (NPP), Gower Street, University College London, WC1E 6BT, UK
| |
Collapse
|
6
|
Ramos-Moreno T, Cifra A, Litsa NL, Melin E, Ahl M, Christiansen SH, Gøtzsche CR, Cescon M, Bonaldo P, van Loo K, Borger V, Jasper JA, Becker A, van Vliet EA, Aronica E, Woldbye DP, Kokaia M. Collagen VI: Role in synaptic transmission and seizure-related excitability. Exp Neurol 2024; 380:114911. [PMID: 39094767 DOI: 10.1016/j.expneurol.2024.114911] [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: 03/20/2024] [Revised: 07/05/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Collagen VI (Col-VI) is an extracellular matrix protein primarily known for its bridging role in connective tissues that has been suggested to play a neuroprotective role. In the present study we report increased mRNA and protein expression of Col-VI in the hippocampus and cortex at a late stage of epileptogenesis in a post-status epilepticus (SE) model of epilepsy and in brain tissue from patients with epilepsy. We further present a novel finding that exposure of mouse hippocampal slices to Col-VI augments paired-pulse facilitation in Schaffer collateral-CA1 excitatory synapses indicating decreased release probability of glutamate. In line with this finding, lack of Col-VI expression in the knock-out mice show paired-pulse depression in these synapses, suggesting increased release probability of glutamate. In addition, we observed dynamic changes in Col-VI blood plasma levels in rats after Kainate-induced SE, and increased levels of Col-VI mRNA and protein in autopsy or postmortem brain of humans suffering from epilepsy. Thus, our data indicate that elevated levels of ColVI following seizures leads to attenuated glutamatergic transmission, ultimately resulting in less overall network excitability. Presumably, increased Col-VI may act as part of endogenous compensatory mechanism against enhanced excitability during epileptogenic processes in the hippocampus, and could be further investigated as a potential functional biomarker of epileptogenesis, and/or a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Tania Ramos-Moreno
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Alexandra Cifra
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Nikitidou Ledri Litsa
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Esbjörn Melin
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Matilda Ahl
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden
| | - Sören H Christiansen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Casper R Gøtzsche
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, I-35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, I-35131 Padova, Italy
| | - Karen van Loo
- Institut für Neuropathologie, Universitätsklinikum Bonn, Bonn, Germany
| | - Valeri Borger
- Institut für Neuropathologie, Universitätsklinikum Bonn, Bonn, Germany
| | - J Anink Jasper
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Albert Becker
- Institut für Neuropathologie, Universitätsklinikum Bonn, Bonn, Germany
| | - Erwin A van Vliet
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - David P Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Merab Kokaia
- Epilepsy Centre, Department of Clinical Sciences, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|
7
|
Boddu AV, Brinkerhoff S, Bashir AE, Crowder CM, Awad M, Gonzalez CL, Walker HC. Directional Stimulus-Evoked Pallidal Electrophysiology in Primary Dystonia. Tremor Other Hyperkinet Mov (N Y) 2024; 14:46. [PMID: 39308989 PMCID: PMC11414461 DOI: 10.5334/tohm.916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Background Deep brain stimulation for dystonia improves motor symptoms but variable and delayed responses challenge patient selection, targeting, and device programming. Case Report Here we studied intracranial electrophysiology in a patient with primary dystonia and observed evoked resonant neural activity (ERNA) in the globus pallidus interna. These local stimulus-evoked potentials displayed refractory periods and paired-pulse facilitation at clinically relevant interstimulus intervals. Sensing from directional DBS contacts localized ERNA to an effective stimulation site in the ventral posterolateral portion of the pallidum. Discussion To the best of our knowledge, this is the first observation of ERNA in the globus pallidus interna in a patient with primary dystonia. Stimulus-evoked activity could eventually guide both directional and adaptive stimulation for dystonia and other complex neuropsychiatric disorders.
Collapse
Affiliation(s)
- Aditya V Boddu
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sarah Brinkerhoff
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adam E Bashir
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Camerron M Crowder
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mohammed Awad
- Department of Electrical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Harrison C Walker
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
8
|
Aghi K, Schultz R, Newman ZL, Mendonça P, Li R, Bakshinska D, Isacoff EY. Synapse-to-synapse plasticity variability balanced to generate input-wide constancy of transmitter release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612562. [PMID: 39314438 PMCID: PMC11419063 DOI: 10.1101/2024.09.11.612562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Basal synaptic strength can vary greatly between synapses formed by an individual neuron because of diverse probabilities of action potential (AP) evoked transmitter release ( Pr ). Optical quantal analysis on large numbers of identified Drosophila larval glutamatergic synapses shows that short-term plasticity (STP) also varies greatly between synapses made by an individual type I motor neuron (MN) onto a single body wall muscle. Synapses with high and low P r and different forms and level of STP have a random spatial distribution in the MN nerve terminal, and ones with very different properties can be located within 200 nm of one other. While synapses start off with widely diverse basal P r at low MN AP firing frequency and change P r differentially when MN firing frequency increases, the overall distribution of P r remains remarkably constant due to a balance between the numbers of synapses that facilitate and depress as well as their degree of change and basal synaptic weights. This constancy in transmitter release can ensure robustness across changing behavioral conditions.
Collapse
|
9
|
Malkin J, O'Donnell C, Houghton CJ, Aitchison L. Signatures of Bayesian inference emerge from energy-efficient synapses. eLife 2024; 12:RP92595. [PMID: 39106188 PMCID: PMC11302983 DOI: 10.7554/elife.92595] [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] [Indexed: 08/09/2024] Open
Abstract
Biological synaptic transmission is unreliable, and this unreliability likely degrades neural circuit performance. While there are biophysical mechanisms that can increase reliability, for instance by increasing vesicle release probability, these mechanisms cost energy. We examined four such mechanisms along with the associated scaling of the energetic costs. We then embedded these energetic costs for reliability in artificial neural networks (ANNs) with trainable stochastic synapses, and trained these networks on standard image classification tasks. The resulting networks revealed a tradeoff between circuit performance and the energetic cost of synaptic reliability. Additionally, the optimised networks exhibited two testable predictions consistent with pre-existing experimental data. Specifically, synapses with lower variability tended to have (1) higher input firing rates and (2) lower learning rates. Surprisingly, these predictions also arise when synapse statistics are inferred through Bayesian inference. Indeed, we were able to find a formal, theoretical link between the performance-reliability cost tradeoff and Bayesian inference. This connection suggests two incompatible possibilities: evolution may have chanced upon a scheme for implementing Bayesian inference by optimising energy efficiency, or alternatively, energy-efficient synapses may display signatures of Bayesian inference without actually using Bayes to reason about uncertainty.
Collapse
Affiliation(s)
- James Malkin
- Faculty of Engineering, University of BristolBristolUnited Kingdom
| | - Cian O'Donnell
- Faculty of Engineering, University of BristolBristolUnited Kingdom
- Intelligent Systems Research Centre, School of Computing, Engineering, and Intelligent Systems, Ulster UniversityDerry/LondonderryUnited Kingdom
| | - Conor J Houghton
- Faculty of Engineering, University of BristolBristolUnited Kingdom
| | | |
Collapse
|
10
|
Savtchenko LP, Rusakov DA. Equal levels of pre- and postsynaptic potentiation produce unequal outcomes. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230235. [PMID: 38853561 PMCID: PMC11343314 DOI: 10.1098/rstb.2023.0235] [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: 10/10/2023] [Revised: 12/06/2023] [Accepted: 01/05/2024] [Indexed: 06/11/2024] Open
Abstract
Which proportion of the long-term potentiation (LTP) expressed in the bulk of excitatory synapses is postsynaptic and which presynaptic remains debatable. To understand better the possible impact of either LTP form, we explored a realistic model of a CA1 pyramidal cell equipped with known membrane mechanisms and multiple, stochastic excitatory axo-spinous synapses. Our simulations were designed to establish an input-output transfer function, the dependence between the frequency of presynaptic action potentials triggering probabilistic synaptic discharges and the average frequency of postsynaptic spiking. We found that, within the typical physiological range, potentiation of the postsynaptic current results in a greater overall output than an equivalent increase in presynaptic release probability. This difference grows stronger at lower input frequencies and lower release probabilities. Simulations with a non-hierarchical circular network of principal neurons indicated that equal increases in either synaptic fidelity or synaptic strength of individual connections also produce distinct changes in network activity, although the network phenomenology is likely to be complex. These observations should help to interpret the machinery of LTP phenomena documented in situ. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Leonid P. Savtchenko
- UCL Queen Square Institute of Neurology, University College London, LondonWC1N 3BG, UK
| | - Dmitri A. Rusakov
- UCL Queen Square Institute of Neurology, University College London, LondonWC1N 3BG, UK
| |
Collapse
|
11
|
Früh S, Boudkkazi S, Koppensteiner P, Sereikaite V, Chen LY, Fernandez-Fernandez D, Rem PD, Ulrich D, Schwenk J, Chen Z, Le Monnier E, Fritzius T, Innocenti SM, Besseyrias V, Trovò L, Stawarski M, Argilli E, Sherr EH, van Bon B, Kamsteeg EJ, Iascone M, Pilotta A, Cutrì MR, Azamian MS, Hernández-García A, Lalani SR, Rosenfeld JA, Zhao X, Vogel TP, Ona H, Scott DA, Scheiffele P, Strømgaard K, Tafti M, Gassmann M, Fakler B, Shigemoto R, Bettler B. Monoallelic de novo AJAP1 loss-of-function variants disrupt trans-synaptic control of neurotransmitter release. SCIENCE ADVANCES 2024; 10:eadk5462. [PMID: 38985877 PMCID: PMC11235169 DOI: 10.1126/sciadv.adk5462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 06/04/2024] [Indexed: 07/12/2024]
Abstract
Adherens junction-associated protein 1 (AJAP1) has been implicated in brain diseases; however, a pathogenic mechanism has not been identified. AJAP1 is widely expressed in neurons and binds to γ-aminobutyric acid type B receptors (GBRs), which inhibit neurotransmitter release at most synapses in the brain. Here, we show that AJAP1 is selectively expressed in dendrites and trans-synaptically recruits GBRs to presynaptic sites of neurons expressing AJAP1. We have identified several monoallelic AJAP1 variants in individuals with epilepsy and/or neurodevelopmental disorders. Specifically, we show that the variant p.(W183C) lacks binding to GBRs, resulting in the inability to recruit them. Ultrastructural analysis revealed significantly decreased presynaptic GBR levels in Ajap1-/- and Ajap1W183C/+ mice. Consequently, these mice exhibited reduced GBR-mediated presynaptic inhibition at excitatory and inhibitory synapses, along with impaired synaptic plasticity. Our study reveals that AJAP1 enables the postsynaptic neuron to regulate the level of presynaptic GBR-mediated inhibition, supporting the clinical relevance of loss-of-function AJAP1 variants.
Collapse
Affiliation(s)
- Simon Früh
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Sami Boudkkazi
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Peter Koppensteiner
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Vita Sereikaite
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Li-Yuan Chen
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Diego Fernandez-Fernandez
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Pascal D. Rem
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Daniel Ulrich
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Jochen Schwenk
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Ziyang Chen
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Elodie Le Monnier
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Thorsten Fritzius
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | | | - Valérie Besseyrias
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Luca Trovò
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Emanuela Argilli
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
- Institute of Human Genetics and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Elliott H. Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
- Institute of Human Genetics and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bregje van Bon
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, Netherlands
| | - Erik-Jan Kamsteeg
- Department of Human Genetics, Radboud University Medical Center, Nijmegen 6525, Netherlands
| | - Maria Iascone
- Laboratorio Genetica Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Mahshid S. Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrés Hernández-García
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Seema R. Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaonan Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor Genetics, Houston, TX 77021, USA
| | - Tiphanie P. Vogel
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Herda Ona
- Division of Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Daryl A. Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter Scheiffele
- Biocenter, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Mehdi Tafti
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Bernd Fakler
- Institute of Physiology II, University of Freiburg, Hermann-Herderstrasse 7, 79104 Freiburg, Germany
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| |
Collapse
|
12
|
Moore JJ, Genkin A, Tournoy M, Pughe-Sanford JL, de Ruyter van Steveninck RR, Chklovskii DB. The neuron as a direct data-driven controller. Proc Natl Acad Sci U S A 2024; 121:e2311893121. [PMID: 38913890 PMCID: PMC11228465 DOI: 10.1073/pnas.2311893121] [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: 08/22/2023] [Accepted: 04/12/2024] [Indexed: 06/26/2024] Open
Abstract
In the quest to model neuronal function amid gaps in physiological data, a promising strategy is to develop a normative theory that interprets neuronal physiology as optimizing a computational objective. This study extends current normative models, which primarily optimize prediction, by conceptualizing neurons as optimal feedback controllers. We posit that neurons, especially those beyond early sensory areas, steer their environment toward a specific desired state through their output. This environment comprises both synaptically interlinked neurons and external motor sensory feedback loops, enabling neurons to evaluate the effectiveness of their control via synaptic feedback. To model neurons as biologically feasible controllers which implicitly identify loop dynamics, infer latent states, and optimize control we utilize the contemporary direct data-driven control (DD-DC) framework. Our DD-DC neuron model explains various neurophysiological phenomena: the shift from potentiation to depression in spike-timing-dependent plasticity with its asymmetry, the duration and adaptive nature of feedforward and feedback neuronal filters, the imprecision in spike generation under constant stimulation, and the characteristic operational variability and noise in the brain. Our model presents a significant departure from the traditional, feedforward, instant-response McCulloch-Pitts-Rosenblatt neuron, offering a modern, biologically informed fundamental unit for constructing neural networks.
Collapse
Affiliation(s)
- Jason J Moore
- Neuroscience Institute, New York University Grossman School of Medicine, New York City, NY 10016
- Center for Computational Neuroscience, Flatiron Institute, New York City, NY 10010
| | - Alexander Genkin
- Center for Computational Neuroscience, Flatiron Institute, New York City, NY 10010
| | - Magnus Tournoy
- Center for Computational Neuroscience, Flatiron Institute, New York City, NY 10010
| | | | | | - Dmitri B Chklovskii
- Neuroscience Institute, New York University Grossman School of Medicine, New York City, NY 10016
- Center for Computational Neuroscience, Flatiron Institute, New York City, NY 10010
| |
Collapse
|
13
|
Bruentgens F, Moreno Velasquez L, Stumpf A, Parthier D, Breustedt J, Benfenati F, Milovanovic D, Schmitz D, Orlando M. The Lack of Synapsin Alters Presynaptic Plasticity at Hippocampal Mossy Fibers in Male Mice. eNeuro 2024; 11:ENEURO.0330-23.2024. [PMID: 38866497 PMCID: PMC11223178 DOI: 10.1523/eneuro.0330-23.2024] [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: 08/29/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Synapsins are highly abundant presynaptic proteins that play a crucial role in neurotransmission and plasticity via the clustering of synaptic vesicles. The synapsin III isoform is usually downregulated after development, but in hippocampal mossy fiber boutons, it persists in adulthood. Mossy fiber boutons express presynaptic forms of short- and long-term plasticity, which are thought to underlie different forms of learning. Previous research on synapsins at this synapse focused on synapsin isoforms I and II. Thus, a complete picture regarding the role of synapsins in mossy fiber plasticity is still missing. Here, we investigated presynaptic plasticity at hippocampal mossy fiber boutons by combining electrophysiological field recordings and transmission electron microscopy in a mouse model lacking all synapsin isoforms. We found decreased short-term plasticity, i.e., decreased facilitation and post-tetanic potentiation, but increased long-term potentiation in male synapsin triple knock-out (KO) mice. At the ultrastructural level, we observed more dispersed vesicles and a higher density of active zones in mossy fiber boutons from KO animals. Our results indicate that all synapsin isoforms are required for fine regulation of short- and long-term presynaptic plasticity at the mossy fiber synapse.
Collapse
Affiliation(s)
- Felicitas Bruentgens
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Laura Moreno Velasquez
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Alexander Stumpf
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Daniel Parthier
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
| | - Jörg Breustedt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa 16163, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Dragomir Milovanovic
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
| | - Dietmar Schmitz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin 10117, Germany
- Einstein Center for Neurosciences, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Marta Orlando
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| |
Collapse
|
14
|
Dodt S, Widdershooven NV, Dreisow ML, Weiher L, Steuernagel L, Wunderlich FT, Brüning JC, Fenselau H. NPY-mediated synaptic plasticity in the extended amygdala prioritizes feeding during starvation. Nat Commun 2024; 15:5439. [PMID: 38937485 PMCID: PMC11211344 DOI: 10.1038/s41467-024-49766-0] [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: 08/30/2023] [Accepted: 06/18/2024] [Indexed: 06/29/2024] Open
Abstract
Efficient control of feeding behavior requires the coordinated adjustment of complex motivational and affective neurocircuits. Neuropeptides from energy-sensing hypothalamic neurons are potent feeding modulators, but how these endogenous signals shape relevant circuits remains unclear. Here, we examine how the orexigenic neuropeptide Y (NPY) adapts GABAergic inputs to the bed nucleus of the stria terminalis (BNST). We find that fasting increases synaptic connectivity between agouti-related peptide (AgRP)-expressing 'hunger' and BNST neurons, a circuit that promotes feeding. In contrast, GABAergic input from the central amygdala (CeA), an extended amygdala circuit that decreases feeding, is reduced. Activating NPY-expressing AgRP neurons evokes these synaptic adaptations, which are absent in NPY-deficient mice. Moreover, fasting diminishes the ability of CeA projections in the BNST to suppress food intake, and NPY-deficient mice fail to decrease anxiety in order to promote feeding. Thus, AgRP neurons drive input-specific synaptic plasticity, enabling a selective shift in hunger and anxiety signaling during starvation through NPY.
Collapse
Affiliation(s)
- Stephan Dodt
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Noah V Widdershooven
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Marie-Luise Dreisow
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
| | - Lisa Weiher
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany
- Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany.
- Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch-Straße 21, 50931, Cologne, Germany.
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931, Cologne, Germany.
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50924, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne, 50931, Germany.
| |
Collapse
|
15
|
Tsotsokou G, Miliou A, Trompoukis G, Leontiadis LJ, Papatheodoropoulos C. Region-Related Differences in Short-Term Synaptic Plasticity and Synaptotagmin-7 in the Male and Female Hippocampus of a Rat Model of Fragile X Syndrome. Int J Mol Sci 2024; 25:6975. [PMID: 39000085 PMCID: PMC11240911 DOI: 10.3390/ijms25136975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Fragile X syndrome (FXS) is an intellectual developmental disorder characterized, inter alia, by deficits in the short-term processing of neural information, such as sensory processing and working memory. The primary cause of FXS is the loss of fragile X messenger ribonucleoprotein (FMRP), which is profoundly involved in synaptic function and plasticity. Short-term synaptic plasticity (STSP) may play important roles in functions that are affected by FXS. Recent evidence points to the crucial involvement of the presynaptic calcium sensor synaptotagmin-7 (Syt-7) in STSP. However, how the loss of FMRP affects STSP and Syt-7 have been insufficiently studied. Furthermore, males and females are affected differently by FXS, but the underlying mechanisms remain elusive. The aim of the present study was to investigate possible changes in STSP and the expression of Syt-7 in the dorsal (DH) and ventral (VH) hippocampus of adult males and females in a Fmr1-knockout (KO) rat model of FXS. We found that the paired-pulse ratio (PPR) and frequency facilitation/depression (FF/D), two forms of STSP, as well as the expression of Syt-7, are normal in adult KO males, but the PPR is increased in the ventral hippocampus of KO females (6.4 ± 3.7 vs. 18.3 ± 4.2 at 25 ms in wild type (WT) and KO, respectively). Furthermore, we found no gender-related differences, but did find robust region-dependent difference in the STSP (e.g., the PPR at 50 ms: 50.0 ± 5.5 vs. 17.6 ± 2.9 in DH and VH of WT male rats; 53.1 ± 3.6 vs. 19.3 ± 4.6 in DH and VH of WT female rats; 48.1 ± 2.3 vs. 19.1 ± 3.3 in DH and VH of KO male rats; and 51.2 ± 3.3 vs. 24.7 ± 4.3 in DH and VH of KO female rats). AMPA receptors are similarly expressed in the two hippocampal segments of the two genotypes and in both genders. Also, basal excitatory synaptic transmission is higher in males compared to females. Interestingly, we found more than a twofold higher level of Syt-7, not synaptotagmin-1, in the dorsal compared to the ventral hippocampus in the males of both genotypes (0.43 ± 0.1 vs. 0.16 ± 0.02 in DH and VH of WT male rats, and 0.6 ± 0.13 vs. 0.23 ± 0.04 in DH and VH of KO male rats) and in the WT females (0.97 ± 0.23 vs. 0.31 ± 0.09 in DH and VH). These results point to the susceptibility of the female ventral hippocampus to FMRP loss. Importantly, the different levels of Syt-7, which parallel the higher score of the dorsal vs. ventral hippocampus on synaptic facilitation, suggest that Syt-7 may play a pivotal role in defining the striking differences in STSP along the long axis of the hippocampus.
Collapse
Affiliation(s)
| | | | | | | | - Costas Papatheodoropoulos
- Lab of Physiology-Neurophysiology, Department of Medicine, University of Patras, 265 04 Patras, Greece; (G.T.); (A.M.); (G.T.); (L.J.L.)
| |
Collapse
|
16
|
Simões de Oliveira L, O'Leary HE, Nawaz S, Loureiro R, Davenport EC, Baxter P, Louros SR, Dando O, Perkins E, Peltier J, Trost M, Osterweil EK, Hardingham GE, Cousin MA, Chattarji S, Booker SA, Benke TA, Wyllie DJA, Kind PC. Enhanced hippocampal LTP but normal NMDA receptor and AMPA receptor function in a rat model of CDKL5 deficiency disorder. Mol Autism 2024; 15:28. [PMID: 38877552 PMCID: PMC11177379 DOI: 10.1186/s13229-024-00601-9] [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: 11/03/2023] [Accepted: 05/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Mutations in the X-linked gene cyclin-dependent kinase-like 5 (CDKL5) cause a severe neurological disorder characterised by early-onset epileptic seizures, autism and intellectual disability (ID). Impaired hippocampal function has been implicated in other models of monogenic forms of autism spectrum disorders and ID and is often linked to epilepsy and behavioural abnormalities. Many individuals with CDKL5 deficiency disorder (CDD) have null mutations and complete loss of CDKL5 protein, therefore in the current study we used a Cdkl5-/y rat model to elucidate the impact of CDKL5 loss on cellular excitability and synaptic function of CA1 pyramidal cells (PCs). We hypothesised abnormal pre and/or post synaptic function and plasticity would be observed in the hippocampus of Cdkl5-/y rats. METHODS To allow cross-species comparisons of phenotypes associated with the loss of CDKL5, we generated a loss of function mutation in exon 8 of the rat Cdkl5 gene and assessed the impact of the loss of CDLK5 using a combination of extracellular and whole-cell electrophysiological recordings, biochemistry, and histology. RESULTS Our results indicate that CA1 hippocampal long-term potentiation (LTP) is enhanced in slices prepared from juvenile, but not adult, Cdkl5-/y rats. Enhanced LTP does not result from changes in NMDA receptor function or subunit expression as these remain unaltered throughout development. Furthermore, Ca2+ permeable AMPA receptor mediated currents are unchanged in Cdkl5-/y rats. We observe reduced mEPSC frequency accompanied by increased spine density in basal dendrites of CA1 PCs, however we find no evidence supporting an increase in silent synapses when assessed using a minimal stimulation protocol in slices. Additionally, we found no change in paired-pulse ratio, consistent with normal release probability at Schaffer collateral to CA1 PC synapses. CONCLUSIONS Our data indicate a role for CDKL5 in hippocampal synaptic function and raise the possibility that altered intracellular signalling rather than synaptic deficits contribute to the altered plasticity. LIMITATIONS This study has focussed on the electrophysiological and anatomical properties of hippocampal CA1 PCs across early postnatal development. Studies involving other brain regions, older animals and behavioural phenotypes associated with the loss of CDKL5 are needed to understand the pathophysiology of CDD.
Collapse
MESH Headings
- Animals
- Male
- Rats
- CA1 Region, Hippocampal/metabolism
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- Disease Models, Animal
- Epileptic Syndromes/genetics
- Epileptic Syndromes/metabolism
- Excitatory Postsynaptic Potentials
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/metabolism
- Genetic Diseases, X-Linked/physiopathology
- Hippocampus/metabolism
- Long-Term Potentiation
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- Pyramidal Cells/metabolism
- Pyramidal Cells/pathology
- Receptors, AMPA/metabolism
- Receptors, AMPA/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, N-Methyl-D-Aspartate/genetics
- Spasms, Infantile/genetics
- Spasms, Infantile/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- Laura Simões de Oliveira
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Heather E O'Leary
- School of Medicine, University of Colorado, Denver, CO, USA
- Department of Pharmacology, University of Colorado Denver, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Sarfaraz Nawaz
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Rita Loureiro
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | | | - Paul Baxter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Susana R Louros
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Owen Dando
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Emma Perkins
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Julien Peltier
- Faculty of Medical Sciences, Newcastle University Biosciences Institute, Newcastle upon Tyne, NE2 4HH, UK
| | - Matthias Trost
- Faculty of Medical Sciences, Newcastle University Biosciences Institute, Newcastle upon Tyne, NE2 4HH, UK
| | - Emily K Osterweil
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Sumantra Chattarji
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bangalore, 560065, India
- Centre for Brain Development and Repair, Instem, Bangalore, India
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK
| | - Tim A Benke
- School of Medicine, University of Colorado, Denver, CO, USA.
- Department of Pharmacology, University of Colorado Denver, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK.
- Centre for Brain Development and Repair, Instem, Bangalore, India.
- Centre for Discovery Brain Sciences, Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh, EH8 9XD, UK.
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
- Simons Initiative for the Developing Brain, Patrick Wild Centre, University of Edinburgh, Edinburgh, UK.
- Centre for Brain Development and Repair, Instem, Bangalore, India.
- Centre for Discovery Brain Sciences, Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
17
|
Sasibhushana RB, Shankaranarayana Rao BS, Srikumar BN. Anxiety-, and depression-like behavior following short-term finasteride administration is associated with impaired synaptic plasticity and cognitive behavior in male rats. J Psychiatr Res 2024; 174:304-318. [PMID: 38685188 DOI: 10.1016/j.jpsychires.2024.04.029] [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: 07/24/2023] [Revised: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024]
Abstract
Finasteride, a 5α-Reductase inhibitor, is used to treat male pattern baldness and benign prostatic hyperplasia. Several clinical studies show that chronic finasteride treatment induces persistent depression, suicidal thoughts and cognitive impairment and these symptoms are persistent even after its withdrawal. Previous results from our lab showed that repeated administration of finasteride for six days induces depression-like behavior. However, whether short-term finasteride administration induces anxiety-like behavior and memory impairment and alters synaptic plasticity are not known, which formed the basis of this study. Finasteride was administered to 2-2.5 months old male Wistar rats for six days and subjected to behavioral evaluation, biochemical estimation and synaptic plasticity assessment. Anxiety-like behavior was evaluated in the elevated plus maze (EPM), open field test (OFT), light/dark test (LDT), and novelty suppressed feeding test (NSFT), and learning and memory using novel object recognition test (NORT) and novel object location test (NOLT) and depression-like behavior in the sucrose preference test (SPT). Synaptic plasticity in the hippocampal Schaffer collateral-CA1 was evaluated using slice field potential recordings. Plasma corticosterone levels were estimated using ELISA. Finasteride administration induced anxiety-like behavior in the EPM, OFT, LDT and NSFT, and depression-like behavior in the SPT. Further, finasteride induced hippocampal dependent spatial learning and memory impairment in the NOLT. In addition, finasteride decreased basal synaptic plasticity and long-term potentiation (LTP) in the hippocampus. A trend of increased plasma corticosterone levels was observed following repeated finasteride administration. These results indicate the potential role of corticosterone and synaptic plasticity in finasteride-induced effects and further studies will pave way for the development of novel neurosteroid-based therapeutics in neuropsychiatric diseases.
Collapse
Affiliation(s)
- R B Sasibhushana
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - B S Shankaranarayana Rao
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, India
| | - Bettadapura N Srikumar
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Hosur Road, Bengaluru, 560029, India.
| |
Collapse
|
18
|
Tsotsokou G, Trompoukis G, Papatheodoropoulos C. Muscarinic Modulation of Synaptic Transmission and Short-Term Plasticity in the Dorsal and Ventral Hippocampus. Mol Cell Neurosci 2024; 129:103935. [PMID: 38703973 DOI: 10.1016/j.mcn.2024.103935] [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: 09/29/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
Muscarinic neurotransmission is fundamentally involved in supporting several brain functions by modulating flow of information in brain neural circuits including the hippocampus which displays a remarkable functional segregation along its longitudinal axis. However, how muscarinic neuromodulation contributes to the functional segregation along the hippocampus remains unclear. In this study we show that the nonselective muscarinic receptor agonist carbachol similarly suppresses basal synaptic transmission in the dorsal and ventral CA1 hippocampal field, in a concentration-depended manner. Furthermore, using a ten-pulse stimulation train of varying frequency we found that carbachol changes the frequency filtering properties more in ventral than dorsal hippocampus by facilitating synaptic inputs at a wide range of input frequencies in the ventral compared with dorsal hippocampus. Using the M2 receptor antagonist gallamine and the M4 receptor antagonist tropicamide, we found that M2 receptors are involved in controlling basal synaptic transmission and short-term synaptic plasticity (STSP) in the ventral but not the dorsal hippocampus, while M4 receptors participate in modulating basal synaptic transmission and STSP in both segments of the hippocampus. These results were corroborated by the higher protein expression levels of M2 receptors in the ventral compared with dorsal hippocampus. We conclude that muscarinic transmission modulates excitatory synaptic transmission and short-term synaptic plasticity along the entire rat hippocampus by acting through M4 receptors and recruiting M2 receptors only in the ventral hippocampus. Furthermore, M4 receptors appear to exert a permissive role on the actions of M2 receptors on STSP in the ventral hippocampus. This dorsoventral differentiation of muscarinic modulation is expected to have important implications in information processing along the endogenous hippocampal circuitry.
Collapse
Affiliation(s)
- Giota Tsotsokou
- Laboratory of Physiology, University of Patras, Department of Medicine, Rion, Greece
| | - George Trompoukis
- Laboratory of Physiology, University of Patras, Department of Medicine, Rion, Greece
| | | |
Collapse
|
19
|
Dossi E, Zonca L, Pivonkova H, Milior G, Moulard J, Vargova L, Chever O, Holcman D, Rouach N. Astroglial gap junctions strengthen hippocampal network activity by sustaining afterhyperpolarization via KCNQ channels. Cell Rep 2024; 43:114158. [PMID: 38722742 DOI: 10.1016/j.celrep.2024.114158] [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: 12/15/2023] [Revised: 03/01/2024] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
Throughout the brain, astrocytes form networks mediated by gap junction channels that promote the activity of neuronal ensembles. Although their inputs on neuronal information processing are well established, how molecular gap junction channels shape neuronal network patterns remains unclear. Here, using astroglial connexin-deficient mice, in which astrocytes are disconnected and neuronal bursting patterns are abnormal, we show that astrocyte networks strengthen bursting activity via dynamic regulation of extracellular potassium levels, independently of glutamate homeostasis or metabolic support. Using a facilitation-depression model, we identify neuronal afterhyperpolarization as the key parameter underlying bursting pattern regulation by extracellular potassium in mice with disconnected astrocytes. We confirm this prediction experimentally and reveal that astroglial network control of extracellular potassium sustains neuronal afterhyperpolarization via KCNQ voltage-gated K+ channels. Altogether, these data delineate how astroglial gap junctions mechanistically strengthen neuronal population bursts and point to approaches for controlling aberrant activity in neurological diseases.
Collapse
Affiliation(s)
- Elena Dossi
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Lou Zonca
- Group of Data Modeling and Computational Biology, Institute of Biology, Ecole Normale Superieure, CNRS, INSERM, Université PSL, Paris, France; ED386, Ecole Doctorale de Sciences Mathématiques Paris Centre, Paris, France
| | - Helena Pivonkova
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France; Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic; 2(nd) Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Giampaolo Milior
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Julien Moulard
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Lydia Vargova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic; 2(nd) Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - David Holcman
- Group of Data Modeling and Computational Biology, Institute of Biology, Ecole Normale Superieure, CNRS, INSERM, Université PSL, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France.
| |
Collapse
|
20
|
Öttl M, Toonen RF, Verhage M. Reduced synaptic depression in human neurons carrying homozygous disease-causing STXBP1 variant L446F. Hum Mol Genet 2024; 33:991-1000. [PMID: 38484778 PMCID: PMC11102591 DOI: 10.1093/hmg/ddae035] [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: 12/19/2023] [Revised: 02/02/2024] [Indexed: 05/20/2024] Open
Abstract
MUNC18-1 is an essential protein of the regulated secretion machinery. De novo, heterozygous mutations in STXBP1, the human gene encoding this protein, lead to a severe neurodevelopmental disorder. Here, we describe the electrophysiological characteristics of a unique case of STXBP1-related disorder caused by a homozygous mutation (L446F). We engineered this mutation in induced pluripotent stem cells from a healthy donor (STXBP1LF/LF) to establish isogenic cell models. We performed morphological and electrophysiological analyses on single neurons grown on glial micro-islands. Human STXBP1LF/LF neurons displayed normal morphology and normal basal synaptic transmission but increased paired-pulse ratios and charge released, and reduced synaptic depression compared to control neurons. Immunostainings revealed normal expression levels but impaired recognition by a mutation-specific MUNC18-1 antibody. The electrophysiological gain-of-function phenotype is in line with earlier overexpression studies in Stxbp1 null mouse neurons, with some potentially human-specific features. Therefore, the present study highlights important differences between mouse and human neurons critical for the translatability of pre-clinical studies.
Collapse
Affiliation(s)
- Miriam Öttl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081HV, the Netherlands
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, University Medical Center, De Boelelaan 1117, Amsterdam 1081HV, the Netherlands
| |
Collapse
|
21
|
Asadi nejad H, Yousefi Nejad A, Akbari S, Naseh M, Shid Moosavi SM, Haghani M. The low and high doses administration of lutein improves memory and synaptic plasticity impairment through different mechanisms in a rat model of vascular dementia. PLoS One 2024; 19:e0302850. [PMID: 38748711 PMCID: PMC11095768 DOI: 10.1371/journal.pone.0302850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/12/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND AND AIM Vascular dementia (VD) is a common type of dementia. This study aimed to evaluate the effects of low and high doses of lutein administration in bilateral-carotid vessel occlusion (2VO) rats. EXPERIMENTAL PROCEDURE The rats were divided into the following groups: the control, sham-, vehicle (2VO+V) groups, and two groups after 2VO were treated with lutein 0.5 (2VO+LUT-o.5) and 5mg/kg (2VO+LUT-5). The passive-avoidance and Morris water maze were performed to examine fear and spatial memory. The field-potential recording was used to investigate the properties of basal synaptic transmission (BST), paired-pulse ratio (PPR), as an index for measurement of neurotransmitter release, and long-term potentiation (LTP). The hippocampus was removed to evaluate hippocampal cells, volume, and MDA level. RESULT Treatment with low and high doses improves spatial memory and LTP impairment in VD rats, but only the high dose restores the fear memory, hippocampal cell loss, and volume and MDA level. Interestingly, low-dose, but not high-dose, increased PPR. However, BST recovered only in the high-dose treated group. CONCLUSIONS Treatment with a low dose might affect neurotransmitter release probability, but a high dose affects postsynaptic processes. It seems likely that low and high doses improve memory and LTP through different mechanisms.
Collapse
Affiliation(s)
- Hamideh Asadi nejad
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Yousefi Nejad
- Faculty of Veterinary Medicine, Department of Veterinary Medicine Islamic Azad University of Kazeroon, Shiraz, Iran
| | - Somayeh Akbari
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
Jones G, Akter Y, Shifflett V, Hruska M. Nanoscale analysis of functionally diverse glutamatergic synapses in the neocortex reveals input and layer-specific organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592008. [PMID: 38746319 PMCID: PMC11092571 DOI: 10.1101/2024.05.01.592008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Discovery of synaptic nanostructures suggests a molecular logic for the flexibility of synaptic function. We still have little understanding of how functionally diverse synapses in the brain organize their nanoarchitecture due to challenges associated with super-resolution imaging in complex brain tissue. Here, we characterized single-domain camelid nanobodies for the 3D quantitative multiplex imaging of synaptic nano-organization in 6 µm brain cryosections using STED nanoscopy. We focused on thalamocortical (TC) and corticocortical (CC) synapses along the apical-basal axis of layer 5 pyramidal neurons as models of functionally diverse glutamatergic synapses in the brain. Spines receiving TC input were larger than CC spines in all layers examined. However, TC synapses on apical and basal dendrites conformed to different organizational principles. TC afferents on apical dendrites frequently contacted spines with multiple aligned PSD-95/Bassoon nanomodules, which are larger. TC spines on basal dendrites contained mostly one aligned PSD-95/Bassoon nanocluster. However, PSD-95 nanoclusters were larger and scaled with spine volume. The nano-organization of CC synapses did not change across cortical layers. These results highlight striking nanoscale diversity of functionally distinct glutamatergic synapses, relying on afferent input and sub-cellular localization of individual synaptic connections.
Collapse
|
23
|
Kabeiseman E, Paulsen RT, Burrell BD. Characterization of a Fatty Acid Amide Hydrolase (FAAH) in Hirudo verbana. RESEARCH SQUARE 2024:rs.3.rs-4271305. [PMID: 38699363 PMCID: PMC11065068 DOI: 10.21203/rs.3.rs-4271305/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
The endocannabinoid system plays a critical role in modulating both peripheral and central nervous system function. Despite being present throughout the animal kingdom, there has been relatively little investigation of the endocannabinoid system beyond the traditional animal model systems. In this study, we report on the identification and characterization of a fatty acid aminohydrolase (FAAH) in the medicinal leech, Hirudo verbana. FAAH is the primary enzyme responsible for metabolizing the endocannabinoid signaling molecule arachidonoyl ethanolamide (anandamide or AEA) and therefore plays a critical role in regulating AEA levels in the nervous system. This Hirudo FAAH (HirFAAH) is expressed in the leech central nervous system (CNS) and is an orthologue of FAAH-2 observed in vertebrates. Functionally, HirFAAH has serine hydrolase activity based on activity-based protein profiling (ABPP) studies using the fluorophosphonate probe TAMRA-FP. HirFAAH also hydrolyzes arachidonyl 7-amino, 4-methyl coumarin amide (AAMCA), a substrate specific to FAAH. Hydrolase activity during both the ABPP and AAMCA assays was eliminated by mutation at a conserved activity-binding site. Activity was also blocked by the known FAAH inhibitor, URB597. Treatment of Hirudo ganglia with URB597 potentiated synapses made by the pressure-sensitive mechanosensory neuron (P cell), mimicking the effects of exogenously applied AEA. The Hirudo CNS has been a useful system in which to study properties of endocannabinoid modulation of nociception relevant to vertebrates. Therefore, this characterization of HirFAAH is an important contribution to comparative studies of the endocannabinoid system.
Collapse
|
24
|
Pereira-Obilinovic U, Hou H, Svoboda K, Wang XJ. Brain mechanism of foraging: Reward-dependent synaptic plasticity versus neural integration of values. Proc Natl Acad Sci U S A 2024; 121:e2318521121. [PMID: 38551832 PMCID: PMC10998608 DOI: 10.1073/pnas.2318521121] [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: 11/01/2023] [Accepted: 01/16/2024] [Indexed: 04/02/2024] Open
Abstract
During foraging behavior, action values are persistently encoded in neural activity and updated depending on the history of choice outcomes. What is the neural mechanism for action value maintenance and updating? Here, we explore two contrasting network models: synaptic learning of action value versus neural integration. We show that both models can reproduce extant experimental data, but they yield distinct predictions about the underlying biological neural circuits. In particular, the neural integrator model but not the synaptic model requires that reward signals are mediated by neural pools selective for action alternatives and their projections are aligned with linear attractor axes in the valuation system. We demonstrate experimentally observable neural dynamical signatures and feasible perturbations to differentiate the two contrasting scenarios, suggesting that the synaptic model is a more robust candidate mechanism. Overall, this work provides a modeling framework to guide future experimental research on probabilistic foraging.
Collapse
Affiliation(s)
- Ulises Pereira-Obilinovic
- Center for Neural Science, New York University, New York, NY10003
- Allen Institute for Neural Dynamics, Seattle, WA98109
| | - Han Hou
- Allen Institute for Neural Dynamics, Seattle, WA98109
| | - Karel Svoboda
- Allen Institute for Neural Dynamics, Seattle, WA98109
| | - Xiao-Jing Wang
- Center for Neural Science, New York University, New York, NY10003
| |
Collapse
|
25
|
Meijer M, Öttl M, Yang J, Subkhangulova A, Kumar A, Feng Z, van Voorst TW, Groffen AJ, van Weering JRT, Zhang Y, Verhage M. Tomosyns attenuate SNARE assembly and synaptic depression by binding to VAMP2-containing template complexes. Nat Commun 2024; 15:2652. [PMID: 38531902 PMCID: PMC10965968 DOI: 10.1038/s41467-024-46828-1] [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: 10/02/2023] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Tomosyns are widely thought to attenuate membrane fusion by competing with synaptobrevin-2/VAMP2 for SNARE-complex assembly. Here, we present evidence against this scenario. In a novel mouse model, tomosyn-1/2 deficiency lowered the fusion barrier and enhanced the probability that synaptic vesicles fuse, resulting in stronger synapses with faster depression and slower recovery. While wild-type tomosyn-1m rescued these phenotypes, substitution of its SNARE motif with that of synaptobrevin-2/VAMP2 did not. Single-molecule force measurements indeed revealed that tomosyn's SNARE motif cannot substitute synaptobrevin-2/VAMP2 to form template complexes with Munc18-1 and syntaxin-1, an essential intermediate for SNARE assembly. Instead, tomosyns extensively bind synaptobrevin-2/VAMP2-containing template complexes and prevent SNAP-25 association. Structure-function analyses indicate that the C-terminal polybasic region contributes to tomosyn's inhibitory function. These results reveal that tomosyns regulate synaptic transmission by cooperating with synaptobrevin-2/VAMP2 to prevent SNAP-25 binding during SNARE assembly, thereby limiting initial synaptic strength and equalizing it during repetitive stimulation.
Collapse
Affiliation(s)
- Marieke Meijer
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands.
| | - Miriam Öttl
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Jie Yang
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA.
| | - Aygul Subkhangulova
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Avinash Kumar
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Zicheng Feng
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Torben W van Voorst
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Alexander J Groffen
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands
| | - Jan R T van Weering
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands
| | - Yongli Zhang
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, 06511, USA.
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
| | - Matthijs Verhage
- Department of Human Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam University Medical Center, 1081HV, Amsterdam, The Netherlands.
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Di Castro MA, Garofalo S, Mormino A, Carbonari L, Di Pietro E, De Felice E, Catalano M, Maggi L, Limatola C. Interleukin-15 alters hippocampal synaptic transmission and impairs episodic memory formation in mice. Brain Behav Immun 2024; 115:652-666. [PMID: 37992787 DOI: 10.1016/j.bbi.2023.11.015] [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: 07/14/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/24/2023] Open
Abstract
Cytokines are potent immunomodulators exerting pleiotropic effects in the central nervous system (CNS). They influence neuronal functions and circuit activities with effects on memory processes and behaviors. Here, we unravel a neuromodulatory activity of interleukin-15 (IL-15) in mouse brain. Acute exposure of hippocampal slices to IL-15 enhances gamma-aminobutyricacid (GABA) release and reduces glutamatergic currents, while chronic treatment with IL-15 increases the frequency of hippocampal miniature inhibitory synaptic transmission and impairs memory formation in the novel object recognition (NOR) test. Moreover, we describe that serotonin is involved in mediating the hippocampal effects of IL-15, because a selective 5-HT3A receptor antagonist prevents the effects on inhibitory neurotransmission and ameliorates mice performance in the NOR test. These findings provide new insights into the modulatory activities of cytokines in the CNS, with implications on behavior.
Collapse
Affiliation(s)
- Maria Amalia Di Castro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Alessandro Mormino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Laura Carbonari
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Erika Di Pietro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Eleonora De Felice
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Cristina Limatola
- IRCCS Neuromed Via Atinese 18, 86077 Pozzilli, Italy; Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur, Italy.
| |
Collapse
|
27
|
Neher E. Interpretation of presynaptic phenotypes of synaptic plasticity in terms of a two-step priming process. J Gen Physiol 2024; 156:e202313454. [PMID: 38112713 PMCID: PMC10730358 DOI: 10.1085/jgp.202313454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Studies on synaptic proteins involved in neurotransmitter release often aim at distinguishing between their roles in vesicle priming (the docking of synaptic vesicles to the plasma membrane and the assembly of a release machinery) as opposed to the process of vesicle fusion. This has traditionally been done by estimating two parameters, the size of the pool of fusion-competent vesicles (the readily releasable pool, RRP) and the probability that such vesicles are released by an action potential, with the aim of determining how these parameters are affected by molecular perturbations. Here, it is argued that the assumption of a homogeneous RRP may be too simplistic and may blur the distinction between vesicle priming and fusion. Rather, considering priming as a dynamic and reversible multistep process allows alternative interpretations of mutagenesis-induced changes in synaptic transmission and suggests mechanisms for variability in synaptic strength and short-term plasticity among synapses, as well as for interactions between short- and long-term plasticity. In many cases, assigned roles of proteins or causes for observed phenotypes are shifted from fusion- to priming-related when considering multistep priming. Activity-dependent enhancement of priming is an essential element in this alternative view and its variation among synapse types can explain why some synapses show depression and others show facilitation at low to intermediate stimulation frequencies. Multistep priming also suggests a mechanism for frequency invariance of steady-state release, which can be observed in some synapses involved in sensory processing.
Collapse
Affiliation(s)
- Erwin Neher
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
28
|
Dósa Z, Nieto-Gonzalez JL, Elfving B, Hougaard KS, Holm MM, Wegener G, Jensen K. Reduction in hippocampal GABAergic transmission in a low birth weight rat model of depression. Acta Neuropsychiatr 2023; 35:315-327. [PMID: 36896595 DOI: 10.1017/neu.2023.18] [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] [Indexed: 03/11/2023]
Abstract
Prenatal stress is believed to increase the risk of developing neuropsychiatric disorders, including major depression. Adverse genetic and environmental impacts during early development, such as glucocorticoid hyper-exposure, can lead to changes in the foetal brain, linked to mental illnesses developed in later life. Dysfunction in the GABAergic inhibitory system is associated with depressive disorders. However, the pathophysiology of GABAergic signalling is poorly understood in mood disorders. Here, we investigated GABAergic neurotransmission in the low birth weight (LBW) rat model of depression. Pregnant rats, exposed to dexamethasone, a synthetic glucocorticoid, during the last week of gestation, yielded LBW offspring showing anxiety- and depressive-like behaviour in adulthood. Patch-clamp recordings from dentate gyrus granule cells in brain slices were used to examine phasic and tonic GABAA receptor-mediated currents. The transcriptional levels of selected genes associated with synaptic vesicle proteins and GABAergic neurotransmission were investigated. The frequency of spontaneous inhibitory postsynaptic currents (sIPSC) was similar in control and LBW rats. Using a paired-pulse protocol to stimulate GABAergic fibres impinging onto granule cells, we found indications of decreased probability of GABA release in LBW rats. However, tonic GABAergic currents and miniature IPSCs, reflecting quantal vesicle release, appeared normal. Additionally, we found elevated expression levels of two presynaptic proteins, Snap-25 and Scamp2, components of the vesicle release machinery. The results suggest that altered GABA release may be an essential feature in the depressive-like phenotype of LBW rats.
Collapse
Affiliation(s)
- Zita Dósa
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mai Marie Holm
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Pharmaceutical Research Center of Excellence, North-West University, Potchefstroom, South Africa
| | - Kimmo Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
29
|
Sharifi M, Oryan S, Komaki A, Barkley V, Sarihi A, Mirnajafi-Zadeh J. Comparing the synaptic potentiation in schaffer collateral-CA1 synapses in dorsal and intermediate regions of the hippocampus in normal and kindled rats. IBRO Neurosci Rep 2023; 15:252-261. [PMID: 37841086 PMCID: PMC10570600 DOI: 10.1016/j.ibneur.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/26/2023] [Accepted: 09/30/2023] [Indexed: 10/17/2023] Open
Abstract
There is growing evidence that the hippocampus comprises diverse neural circuits that exhibit longitudinal variation in their properties, however, the intermediate region of the hippocampus has received comparatively little attention. Therefore, this study was designed to compared short- and long-term synaptic plasticity between the dorsal and intermediate regions of the hippocampus in normal and PTZ-kindled rats. Short-term plasticity was assessed by measuring the ratio of field excitatory postsynaptic potentials' (fEPSPs) slope in response to paired-pulse stimulation at three different inter-pulse intervals (20, 80, and 160 ms), while long-term plasticity was assessed using primed burst stimulation (PBS). The results showed that the basal synaptic strength differed between the dorsal and intermediate regions of the hippocampus in both control and kindled rats. In the control group, paired-pulse stimulation of Schaffer collaterals resulted in a significantly lower fEPSP slope in the intermediate part of the hippocampus compared to the dorsal region. Additionally, the magnitude of long-term potentiation (LTP) was significantly lower in the intermediate part of the hippocampus compared to the dorsal region. In PTZ-kindled rats, both short-term facilitation and long-term potentiation were impaired in both regions of the hippocampus. Interestingly, there was no significant difference in synaptic plasticity between the dorsal and intermediate regions in PTZ-kindled rats, despite impairments in both regions. This suggests that seizures eliminate the regional difference between the dorsal and intermediate parts of the hippocampus, resulting in similar electrophysiological activity in both regions in kindled animals. Future studies should consider this when investigating the responses of the dorsal and intermediate regions of the hippocampus following PTZ kindling.
Collapse
Affiliation(s)
- Maryam Sharifi
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Victoria Barkley
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
30
|
Pati E, Franceschi Biagioni A, Casani R, Lozano N, Kostarelos K, Cellot G, Ballerini L. Delivery of graphene oxide nanosheets modulates glutamate release and normalizes amygdala synaptic plasticity to improve anxiety-related behavior. NANOSCALE 2023; 15:18581-18591. [PMID: 37955642 DOI: 10.1039/d3nr04490d] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Graphene oxide nanosheets (GO) were reported to alter neurobiological processes involving cell membrane dynamics. GO ability to reversibly downregulate specifically glutamatergic synapses underpins their potential in future neurotherapeutic developments. Aberrant glutamate plasticity contributes to stress-related psychopathology and drugs which target dysregulated glutamate represent promising treatments. We find that in a rat model of post-traumatic stress disorder (PTSD), a single injection of GO to the lateral amygdala following the stressful event induced PTSD-related behavior remission and reduced dendritic spine densities. We explored from a mechanistic perspective how GO could impair glutamate synaptic plasticity. By simultaneous patch clamp pair recordings of unitary synaptic currents, live-imaging of presynaptic vesicle release and confocal microscopy, we report that GO nanosheets altered the probability of release enhancing the extinction of synaptic plasticity in the amygdala. These findings show that the modulation of presynaptic glutamate release might represent an unexplored target for (nano)pharmacological interventions of stress-related disorders.
Collapse
Affiliation(s)
- Elisa Pati
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | | | - Raffaele Casani
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | - Neus Lozano
- Nanomedicine Lab Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
| | - Kostas Kostarelos
- Nanomedicine Lab Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, 08193 Barcelona, Spain
- Nanomedicine Lab, and Faculty of Biology, Medicine & Health, The National Graphene Institute, University of Manchester, Manchester, M13 9PL, UK
| | - Giada Cellot
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| | - Laura Ballerini
- Neuroscience Area, International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy.
| |
Collapse
|
31
|
Nair JD, Wilkinson KA, Yucel BP, Mulle C, Vissel B, Mellor J, Henley JM. GluK2 Q/R editing regulates kainate receptor signaling and long-term potentiation of AMPA receptors. iScience 2023; 26:107708. [PMID: 37720087 PMCID: PMC10504484 DOI: 10.1016/j.isci.2023.107708] [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: 05/22/2023] [Revised: 07/18/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023] Open
Abstract
Q/R editing of the kainate receptor (KAR) subunit GluK2 radically alters recombinant KAR properties, but the effects on endogenous KARs in vivo remain largely unexplored. Here, we compared GluK2 editing-deficient mice that express ∼95% unedited GluK2(Q) to wild-type counterparts that express ∼85% edited GluK2(R). At mossy fiber-CA3 (MF-CA3) synapses GluK2(Q) mice displayed increased postsynaptic KAR function and KAR-mediated presynaptic facilitation, demonstrating enhanced ionotropic function. Conversely, GluK2(Q) mice exhibited reduced metabotropic KAR function, assessed by KAR-mediated inhibition of slow after-hyperpolarization currents (ISAHP). GluK2(Q) mice also had fewer GluA1-and GluA3-containing AMPA receptors (AMPARs) and reduced postsynaptic AMPAR currents at both MF-CA3 and CA1-Schaffer collateral synapses. Moreover, long-term potentiation of AMPAR-mediated transmission at CA1-Schaffer collateral synapses was reduced in GluK2(Q) mice. These findings suggest that GluK2 Q/R editing influences ionotropic/metabotropic balance of KAR signaling to regulate synaptic expression of AMPARs and plasticity.
Collapse
Affiliation(s)
- Jithin D. Nair
- Centre for Synaptic Plasticity, School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Kevin A. Wilkinson
- Centre for Synaptic Plasticity, School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Busra P. Yucel
- Centre for Synaptic Plasticity, School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Christophe Mulle
- CNRS UMR 5297, Interdisciplinary Institute of Neuroscience, University of Bordeaux, France
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Jack Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - Jeremy M. Henley
- Centre for Synaptic Plasticity, School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, UK
| |
Collapse
|
32
|
Çalışkan G, Demiray YE, Stork O. Comparison of three common inbred mouse strains reveals substantial differences in hippocampal GABAergic interneuron populations and in vitro network oscillations. Eur J Neurosci 2023; 58:3383-3401. [PMID: 37550182 DOI: 10.1111/ejn.16112] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 08/09/2023]
Abstract
A major challenge in neuroscience is to pinpoint neurobiological correlates of specific cognitive and neuropsychiatric traits. At the mesoscopic level, promising candidates for establishing such connections are brain oscillations that can be robustly recorded as local field potentials with varying frequencies in the hippocampus in vivo and in vitro. Inbred mouse strains show natural variation in hippocampal synaptic plasticity (e.g. long-term potentiation), a cellular correlate of learning and memory. However, their diversity in expression of different types of hippocampal network oscillations has not been fully explored. Here, we investigated hippocampal network oscillations in three widely used inbred mouse strains: C57BL/6J (B6J), C57BL/6NCrl (B6N) and 129S2/SvPasCrl (129) with the aim to identify common oscillatory characteristics in inbred mouse strains that show aberrant emotional/cognitive behaviour (B6N and 129) and compare them to "control" B6J strain. First, we detected higher gamma oscillation power in the hippocampal CA3 of both B6N and 129 strains. Second, higher incidence of hippocampal sharp wave-ripple (SPW-R) transients was evident in these strains. Third, we observed prominent differences in the densities of distinct interneuron types and CA3 associative network activity, which are indispensable for sustainment of mesoscopic network oscillations. Together, these results add further evidence to profound physiological differences among inbred mouse strains commonly used in neuroscience research.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Research Group "Synapto-Oscillopathies", Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Yunus E Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying MentalHealth (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Site Jena-Magdeburg-Halle, Jena-Magdeburg-Halle, Germany
| |
Collapse
|
33
|
Nagaraja RY, Stiles MA, Sherry DM, Agbaga MP, Ahmad M. Synapse-Specific Defects in Synaptic Transmission in the Cerebellum of W246G Mutant ELOVL4 Rats-a Model of Human SCA34. J Neurosci 2023; 43:5963-5974. [PMID: 37491316 PMCID: PMC10436685 DOI: 10.1523/jneurosci.0378-23.2023] [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: 03/01/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023] Open
Abstract
Elongation of very long fatty acids-4 (ELOVL4) mediates biosynthesis of very long chain-fatty acids (VLC-FA; ≥28 carbons). Various mutations in this enzyme result in spinocerebellar ataxia-34 (SCA34). We generated a rat model of human SCA34 by knock-in of a naturally occurring c.736T>G, p.W246G mutation in the Elovl4 gene. Our previous analysis of homozygous W246G mutant ELOVL4 rats (MUT) revealed early-onset gait disturbance and impaired synaptic transmission and plasticity at parallel fiber-Purkinje cell (PF-PC) and climbing fiber-Purkinje cell (CF-PC) synapses. However, the underlying mechanisms that caused these defects remained unknown. Here, we report detailed patch-clamp recordings from Purkinje cells that identify impaired synaptic mechanisms. Our results show that miniature EPSC (mEPSC) frequency is reduced in MUT rats with no change in mEPSC amplitude, suggesting a presynaptic defect of excitatory synaptic transmission on Purkinje cells. We also find alterations in inhibitory synaptic transmission as miniature IPSC (mIPSC) frequency and amplitude are increased in MUT Purkinje cells. Paired-pulse ratio is reduced at PF-PC synapses but increased at CF-PC synapses in MUT rats, which along with results from high-frequency stimulation suggest opposite changes in the release probability at these two synapses. In contrast, we identify exaggerated persistence of EPSC amplitude at CF-PC and PF-PC synapses in MUT cerebellum, suggesting a larger readily releasable pool (RRP) at both synapses. Furthermore, the dendritic spine density is reduced in MUT Purkinje cells. Thus, our results uncover novel mechanisms of action of VLC-FA at cerebellar synapses, and elucidate the synaptic dysfunction underlying SCA34 pathology.SIGNIFICANCE STATEMENT Very long chain-fatty acids (VLC-FA) are an understudied class of fatty acids that are present in the brain. They are critical for brain function as their deficiency caused by mutations in elongation of very long fatty acids-4 (ELOVL4), the enzyme that mediates their biosynthesis, results in neurologic diseases including spinocerebellar ataxia-34 (SCA34), neuroichthyosis, and Stargardt-like macular dystrophy. In this study, we investigated the synaptic defects present in a rat model of SCA34 and identified defects in presynaptic neurotransmitter release and dendritic spine density at synapses in the cerebellum, a brain region involved in motor coordination. These results advance our understanding of the synaptic mechanisms regulated by VLC-FA and describe the synaptic dysfunction that leads to motor incoordination in SCA34.
Collapse
Affiliation(s)
- Raghavendra Y Nagaraja
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Megan A Stiles
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Martin-Paul Agbaga
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Mohiuddin Ahmad
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
34
|
Lumeij LB, van Huijstee AN, Cappaert NLM, Kessels HW. Variance analysis as a method to predict the locus of plasticity at populations of non-uniform synapses. Front Cell Neurosci 2023; 17:1232541. [PMID: 37528963 PMCID: PMC10388551 DOI: 10.3389/fncel.2023.1232541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Our knowledge on synaptic transmission in the central nervous system has often been obtained by evoking synaptic responses to populations of synapses. Analysis of the variance in synaptic responses can be applied as a method to predict whether a change in synaptic responses is a consequence of altered presynaptic neurotransmitter release or postsynaptic receptors. However, variance analysis is based on binomial statistics, which assumes that synapses are uniform. In reality, synapses are far from uniform, which questions the reliability of variance analysis when applying this method to populations of synapses. To address this, we used an in silico model for evoked synaptic responses and compared variance analysis outcomes between populations of uniform versus non-uniform synapses. This simulation revealed that variance analysis produces similar results irrespectively of the grade of uniformity of synapses. We put this variance analysis to the test with an electrophysiology experiment using a model system for which the loci of plasticity are well established: the effect of amyloid-β on synapses. Variance analysis correctly predicted that postsynaptically produced amyloid-β triggered predominantly a loss of synapses and a minor reduction of postsynaptic currents in remaining synapses with little effect on presynaptic release probability. We propose that variance analysis can be reliably used to predict the locus of synaptic changes for populations of non-uniform synapses.
Collapse
|
35
|
Liu J, Ke P, Guo H, Gu J, Liu Y, Tian X, Wang X, Xiao F. Activation of TLR7-mediated autophagy increases epileptic susceptibility via reduced KIF5A-dependent GABA A receptor transport in a murine model. Exp Mol Med 2023; 55:1159-1173. [PMID: 37258573 PMCID: PMC10317981 DOI: 10.1038/s12276-023-01000-5] [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: 09/02/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 06/02/2023] Open
Abstract
The pathophysiological mechanisms underlying epileptogenesis are poorly understood but are considered to actively involve an imbalance between excitatory and inhibitory synaptic transmission. Excessive activation of autophagy, a cellular pathway that leads to the removal of proteins, is known to aggravate the disease. Toll-like receptor (TLR) 7 is an innate immune receptor that regulates autophagy in infectious and noninfectious diseases. However, the relationship between TLR7, autophagy, and synaptic transmission during epileptogenesis remains unclear. We found that TLR7 was activated in neurons in the early stage of epileptogenesis. TLR7 knockout significantly suppressed seizure susceptibility and neuronal excitability. Furthermore, activation of TLR7 induced autophagy and decreased the expression of kinesin family member 5 A (KIF5A), which influenced interactions with γ-aminobutyric acid type A receptor (GABAAR)-associated protein and GABAARβ2/3, thus producing abnormal GABAAR-mediated postsynaptic transmission. Our results indicated that TLR7 is an important factor in regulating epileptogenesis, suggesting a possible therapeutic target for epilepsy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
- Department of Neurology, Chongqing University Three Gorges Hospital, 165 Xincheng Road, Chongqing, 404100, China
| | - Pingyang Ke
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
| | - Haokun Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
| | - Juan Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
| | - Yan Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China.
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China.
- Institute for Brain Science and Disease of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
36
|
Rigby M, Grillo FW, Compans B, Neves G, Gallinaro J, Nashashibi S, Horton S, Pereira Machado PM, Carbajal MA, Vizcay-Barrena G, Levet F, Sibarita JB, Kirkland A, Fleck RA, Clopath C, Burrone J. Multi-synaptic boutons are a feature of CA1 hippocampal connections in the stratum oriens. Cell Rep 2023; 42:112397. [PMID: 37074915 PMCID: PMC10695768 DOI: 10.1016/j.celrep.2023.112397] [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: 05/20/2022] [Revised: 01/21/2023] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
Excitatory synapses are typically described as single synaptic boutons (SSBs), where one presynaptic bouton contacts a single postsynaptic spine. Using serial section block-face scanning electron microscopy, we found that this textbook definition of the synapse does not fully apply to the CA1 region of the hippocampus. Roughly half of all excitatory synapses in the stratum oriens involved multi-synaptic boutons (MSBs), where a single presynaptic bouton containing multiple active zones contacted many postsynaptic spines (from 2 to 7) on the basal dendrites of different cells. The fraction of MSBs increased during development (from postnatal day 22 [P22] to P100) and decreased with distance from the soma. Curiously, synaptic properties such as active zone (AZ) or postsynaptic density (PSD) size exhibited less within-MSB variation when compared with neighboring SSBs, features that were confirmed by super-resolution light microscopy. Computer simulations suggest that these properties favor synchronous activity in CA1 networks.
Collapse
Affiliation(s)
- Mark Rigby
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Federico W Grillo
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Benjamin Compans
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Guilherme Neves
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Julia Gallinaro
- Bioengineering Department, Imperial College London, London, UK
| | - Sophie Nashashibi
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Sally Horton
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Pedro M Pereira Machado
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Maria Alejandra Carbajal
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Florian Levet
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; University Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR3420, US 4, 33000 Bordeaux, France
| | - Jean-Baptiste Sibarita
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Angus Kirkland
- The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Roland A Fleck
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Claudia Clopath
- Bioengineering Department, Imperial College London, London, UK
| | - Juan Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| |
Collapse
|
37
|
Fatemeh B, Koorosh S, Amir S, Yaghoub F, Javad MZ. Intra-hippocampal cis-P tau microinjection induces long-term changes in behavior and synaptic plasticity in mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:9. [PMID: 37231523 DOI: 10.1186/s12993-023-00211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Alzheimer's disease is accompanied by an abnormal high accumulation of cis-P tau. However, the long-term changes in behavior following tau accumulation remains under debate. The present study investigated the long-term effects of tauopathy on learning and memory, synaptic plasticity, and hippocampal cell numbers. RESULTS Cis-P tau was microinjected into the dorsal hippocampus to generate Alzheimer's like-disease model in C57BL/6 mice. Cis-P tau injected animals showed a significant impairment in learning and memory in Y-maze and Barnes maze tests. In another group of animals, the generation of long-term potentiation (LTP) was evaluated in hippocampal slices 7 months after cis-P tau injection. LTP induction was disrupted only in the dorsal but not ventral hippocampal slices. The basal synaptic transmission was also reduced in dorsal hippocampal slices. In addition, hippocampal sampling was done, and the number of cells was assessed by Nissl staining. Obtained results indicated that the number of survived cells was significantly reduced in the dorsal and ventral hippocampus of cis P-tau injected animals compared to the animals in control group. However, the decrement of cell number was higher in the dorsal compared to the ventral hippocampus. CONCLUSIONS In conclusion, intra-hippocampal cis-P tau injection produced learning and memory impairment at 7 months after its injection. This impairment might result from LTP disruption and a significant decrease in the number of neurons in the dorsal hippocampus.
Collapse
Affiliation(s)
- Bakhtiarzadeh Fatemeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
| | - Shahpasand Koorosh
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shojaei Amir
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Fathollahi Yaghoub
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran
| | - Mirnajafi-Zadeh Javad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-331, Tehran, 1411713116, Iran.
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
38
|
Franzen AD, Paulsen RT, Kabeiseman EJ, Burrell BD. Heterosynaptic long-term potentiation of non-nociceptive synapses requires endocannabinoids, NMDARs, CamKII, and PKCζ. J Neurophysiol 2023; 129:807-818. [PMID: 36883763 PMCID: PMC10085563 DOI: 10.1152/jn.00494.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Noxious stimuli or injury can trigger long-lasting sensitization to non-nociceptive stimuli (referred to as allodynia in mammals). Long-term potentiation (LTP) of nociceptive synapses has been shown to contribute to nociceptive sensitization (hyperalgesia) and there is even evidence of heterosynaptic spread of LTP contributing to this type of sensitization. This study will focus on how activation of nociceptors elicits heterosynaptic LTP (hetLTP) in non-nociceptive synapses. Previous studies in the medicinal leech (Hirudo verbana) have demonstrated that high-frequency stimulation (HFS) of nociceptors produces both homosynaptic LTP as well as hetLTP in non-nociceptive afferent synapses. This hetLTP involves endocannabinoid-mediated disinhibition of non-nociceptive synapses at the presynaptic level, but it is not clear if there are additional processes contributing to this synaptic potentiation. In this study, we found evidence for the involvement of postsynaptic level change and observed that postsynaptic N-methyl-d-aspartate (NMDA) receptors (NMDARs) were required for this potentiation. Next, Hirudo orthologs for known LTP signaling proteins, CamKII and PKCζ, were identified based on sequences from humans, mice, and the marine mollusk Aplysia. In electrophysiological experiments, inhibitors of CamKII (AIP) and PKCζ (ZIP) were found to interfere with hetLTP. Interestingly, CamKII was found to be necessary for both induction and maintenance of hetLTP, whereas PKCζ was only necessary for maintenance. These findings show that activation of nociceptors can elicit a potentiation of non-nociceptive synapses through a process that involves both endocannabinoid-mediated disinhibition and NMDAR-initiated signaling pathways.NEW & NOTEWORTHY Pain-related sensitization involves increases in signaling by non-nociceptive sensory neurons. This can allow non-nociceptive afferents to have access to nociceptive circuitry. In this study, we examine a form of synaptic potentiation in which nociceptor activity elicits increases in non-nociceptive synapses. This process involves endocannabinoids, "gating" the activation of NMDA receptors, which in turn activate CamKII and PKCζ. This study provides an important link in how nociceptive stimuli can enhance non-nociceptive signaling related to pain.
Collapse
Affiliation(s)
- Avery D Franzen
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, United States
| | - Riley T Paulsen
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, United States
| | - Emily J Kabeiseman
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, United States
| | - Brian D Burrell
- Division of Basic Biomedical Sciences, Center for Brain and Behavior Research, Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota, United States
| |
Collapse
|
39
|
Zador A, Escola S, Richards B, Ölveczky B, Bengio Y, Boahen K, Botvinick M, Chklovskii D, Churchland A, Clopath C, DiCarlo J, Ganguli S, Hawkins J, Körding K, Koulakov A, LeCun Y, Lillicrap T, Marblestone A, Olshausen B, Pouget A, Savin C, Sejnowski T, Simoncelli E, Solla S, Sussillo D, Tolias AS, Tsao D. Catalyzing next-generation Artificial Intelligence through NeuroAI. Nat Commun 2023; 14:1597. [PMID: 36949048 PMCID: PMC10033876 DOI: 10.1038/s41467-023-37180-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
Neuroscience has long been an essential driver of progress in artificial intelligence (AI). We propose that to accelerate progress in AI, we must invest in fundamental research in NeuroAI. A core component of this is the embodied Turing test, which challenges AI animal models to interact with the sensorimotor world at skill levels akin to their living counterparts. The embodied Turing test shifts the focus from those capabilities like game playing and language that are especially well-developed or uniquely human to those capabilities - inherited from over 500 million years of evolution - that are shared with all animals. Building models that can pass the embodied Turing test will provide a roadmap for the next generation of AI.
Collapse
Affiliation(s)
- Anthony Zador
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| | - Sean Escola
- Department of Psychiatry, Columbia University, New York, NY, 10027, USA
| | - Blake Richards
- Mila, Montréal, QC, H2S 3H1, Canada
- School of Computer Science, McGill University, Montreal, Canada
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, Canada
- Learning in Machines and Brains Program, CIFAR, Toronto, Canada
| | - Bence Ölveczky
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | | | - Kwabena Boahen
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | | | | | - Anne Churchland
- Department of Neurobiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, SW7 2BW, UK
| | - James DiCarlo
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA
| | - Surya Ganguli
- Department of Applied Physics, Stanford University, Stanford, CA, 94305, USA
| | | | - Konrad Körding
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alexei Koulakov
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Yann LeCun
- Meta, Menlo Park, CA, 94025, USA
- Department of Electrical and Computer Engineering, NYU, Brooklyn, NY, 11201, USA
| | | | | | - Bruno Olshausen
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Alexandre Pouget
- Department of Basic Neurosciences, University of Geneva, Genève, 1211, Switzerland
| | - Cristina Savin
- Center for Neural Science, NYU, New York, NY, 10003, USA
| | | | - Eero Simoncelli
- Departments of Neural Science, Mathematics, and Psychology, NYU, New York, NY, 10003, USA
| | - Sara Solla
- Department of Physiology, Northwestern University, Chicago, IL, 60611, USA
| | - David Sussillo
- Meta, Menlo Park, CA, 94025, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Doris Tsao
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
40
|
Liang SL, Liao WL, Chen RS. Perinatal blockade of neuronal glutamine transport sex-differentially alters glutamatergic synaptic transmission and organization of neurons in the ventrolateral ventral media hypothalamus of adult rats. J Neuroendocrinol 2023; 35:e13253. [PMID: 36949648 DOI: 10.1111/jne.13253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
Compared to male pups, perinatal female rats rely heavily on neuronal glutamine (Gln) transport for sustaining glutamatergic synaptic release in neurons of the ventrolateral ventral media nucleus of the hypothalamus (vlVMH). VMH mainly regulates female sexual behavior and increases glutamate release of perinatal hypothalamic neurons, permanently enhances dendrite spine numbers and is associated with brain and behavioral defeminization. We hypothesized that perinatal interruption of neuronal Gln transport may alter the glutamatergic synaptic transmission during adulthood. Perinatal rats of both sexes received an intracerebroventricular injection of a neuronal Gln uptake blocker, alpha-(methylamino) isobutyric acid (MeAIB, 5 mM), and were raised until adulthood. Whole-cell voltage-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) and evoked EPSCs (eEPSCs) of vlVMH neurons in adult rats with the perinatal pretreatment were conducted and neuron morphology was subjected to post hoc examination. Perinatal MeAIB treatment sex-differentially increased mEPSC frequency in males, but decreased mEPSC amplitude and synaptic Glu release in females. The pretreatment sex-differentially decreased eEPSC amplitude in males but increased AMPA/NMDA current ratio in females, and changed the morphology of vlVMH neurons of adult rats to that of the opposite sex. Most alterations in the glutamatergic synaptic transmission resembled the changes occurring during MeAIB acute exposure in perinatal rats of both sexes. We conclude that perinatal blockade of neuronal Gln transport mediates changes via different presynaptic and postsynaptic mechanisms to induce sex-differential alterations of the glutamatergic synaptic transmission and organization of vlVMH neurons in adult rats. These changes may be permanent and associated with brain and behavior feminization and/or defeminization in rats.
Collapse
Affiliation(s)
- Shu-Ling Liang
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wen-Lin Liao
- Institute of Neuroscience, National Chengchi University, Taipei, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| |
Collapse
|
41
|
Jones ME, Büchler J, Dufor T, Palomer E, Teo S, Martin-Flores N, Boroviak K, Metzakopian E, Gibb A, Salinas PC. A genetic variant of the Wnt receptor LRP6 accelerates synapse degeneration during aging and in Alzheimer's disease. SCIENCE ADVANCES 2023; 9:eabo7421. [PMID: 36638182 PMCID: PMC10624429 DOI: 10.1126/sciadv.abo7421] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Synapse loss strongly correlates with cognitive decline in Alzheimer's disease (AD), but the underlying mechanisms are poorly understood. Deficient Wnt signaling contributes to synapse dysfunction and loss in AD. Consistently, a variant of the LRP6 receptor, (LRP6-Val), with reduced Wnt signaling, is linked to late-onset AD. However, the impact of LRP6-Val on the healthy and AD brain has not been examined. Knock-in mice, generated by gene editing, carrying this Lrp6 variant develop normally. However, neurons from Lrp6-val mice do not respond to Wnt7a, a ligand that promotes synaptic assembly through the Frizzled-5 receptor. Wnt7a stimulates the formation of the low-density lipoprotein receptor-related protein 6 (LRP6)-Frizzled-5 complex but not if LRP6-Val is present. Lrp6-val mice exhibit structural and functional synaptic defects that become pronounced with age. Lrp6-val mice present exacerbated synapse loss around plaques when crossed to the NL-G-F AD model. Our findings uncover a previously unidentified role for Lrp6-val in synapse vulnerability during aging and AD.
Collapse
Affiliation(s)
- Megan E. Jones
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Johanna Büchler
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Tom Dufor
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Ernest Palomer
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Samuel Teo
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Nuria Martin-Flores
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Katharina Boroviak
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 0AH, UK
| | - Alasdair Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| |
Collapse
|
42
|
Fischer M, Zimmerman A, Zhang E, Kolis J, Dickey A, Burdette MK, Chander P, Foulger SH, Brigman JL, Weick JP. Distribution and inflammatory cell response to intracranial delivery of radioluminescent Y2(SiO4)O:Ce particles. PLoS One 2023; 18:e0276819. [PMID: 36634053 PMCID: PMC9836305 DOI: 10.1371/journal.pone.0276819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 10/13/2022] [Indexed: 01/13/2023] Open
Abstract
Due to increasing advances in their manufacture and functionalization, nanoparticle-based systems have become a popular tool for in vivo drug delivery and biodetection. Recently, scintillating nanoparticles such as yttrium orthosilicate doped with cerium (Y2(SiO4)O:Ce) have come under study for their potential utility in optogenetic applications, as they emit photons upon low levels of stimulation from remote x-ray sources. The utility of such nanoparticles in vivo is hampered by rapid clearance from circulation by the mononuclear phagocytic system, which heavily restricts nanoparticle accumulation at target tissues. Local transcranial injection of nanoparticles may deliver scintillating nanoparticles to highly specific brain regions by circumventing the blood-brain barrier and avoiding phagocytic clearance. Few studies to date have examined the distribution and response to nanoparticles following localized delivery to cerebral cortex, a crucial step in understanding the therapeutic potential of nanoparticle-based biodetection in the brain. Following the synthesis and surface modification of these nanoparticles, two doses (1 and 3 mg/ml) were introduced into mouse secondary motor cortex (M2). This region was chosen as the site for RLP delivery, as it represents a common target for optogenetic manipulations of mouse behavior, and RLPs could eventually serve as an injectable x-ray inducible light delivery system. The spread of particles through the target tissue was assessed 24 hours, 72 hours, and 9 days post-injection. Y2(SiO4)O:Ce nanoparticles were found to be detectable in the brain for up to 9 days, initially diffusing through the tissue until 72 hours before achieving partial clearance by the final endpoint. Small transient increases in the presence of IBA-1+ microglia and GFAP+ astrocytic cell populations were detected near nanoparticle injection sites of both doses tested 24 hours after surgery. Taken together, these data provide evidence that Y2(SiO4)O:Ce nanoparticles coated with BSA can be injected directly into mouse cortex in vivo, where they persist for days and are broadly tolerated, such that they may be potentially utilized for remote x-ray activated stimulation and photon emission for optogenetic experiments in the near future.
Collapse
Affiliation(s)
- Máté Fischer
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Amber Zimmerman
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Eric Zhang
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Joseph Kolis
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Ashley Dickey
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Mary K. Burdette
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
| | - Praveen Chander
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Stephen H. Foulger
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina, United States of America
- Center for Optical Materials Science and Engineering Technologies, Clemson University, Clemson, South Carolina, United States of America
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Jonathan L. Brigman
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
- Center for Brain Recovery and Repair, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| | - Jason P. Weick
- Department of Neurosciences, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
- Center for Brain Recovery and Repair, University of New Mexico HSC, Albuquerque, New Mexico, United States of America
| |
Collapse
|
43
|
Tan SY, Jiang JX, Huang HX, Mo XP, Feng JR, Chen Y, Yang L, Long C. Neural mechanism underlies CYLD modulation of morphology and synaptic function of medium spiny neurons in dorsolateral striatum. Front Mol Neurosci 2023; 16:1107355. [PMID: 36846565 PMCID: PMC9945542 DOI: 10.3389/fnmol.2023.1107355] [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: 11/24/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Although the deubiquitinase cylindromatosis (CYLD), an abundant protein in the postsynaptic density fraction, plays a crucial role in mediating the synaptic activity of the striatum, the precise molecular mechanism remains largely unclear. Here, using a Cyld-knockout mouse model, we demonstrate that CYLD regulates dorsolateral striatum (DLS) neuronal morphology, firing activity, excitatory synaptic transmission, and plasticity of striatal medium spiny neurons via, likely, interaction with glutamate receptor 1 (GluA1) and glutamate receptor 2 (GluA2), two key subunits of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs). CYLD deficiency reduces levels of GluA1 and GluA2 surface protein and increases K63-linked ubiquitination, resulting in functional impairments both in AMPAR-mediated excitatory postsynaptic currents and in AMPAR-dependent long-term depression. The results demonstrate a functional association of CYLD with AMPAR activity, which strengthens our understanding of the role of CYLD in striatal neuronal activity.
Collapse
Affiliation(s)
- Shu-Yi Tan
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jin-Xiang Jiang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hui-Xian Huang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiu-Ping Mo
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Jing-Ru Feng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yu Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
44
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
45
|
Myeong J, Klyachko VA. Rapid astrocyte-dependent facilitation amplifies multi-vesicular release in hippocampal synapses. Cell Rep 2022; 41:111820. [PMID: 36516768 PMCID: PMC9805313 DOI: 10.1016/j.celrep.2022.111820] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Synaptic facilitation is a major form of short-term plasticity typically driven by an increase in residual presynaptic calcium. Using near-total internal reflection fluorescence (near-TIRF) imaging of single vesicle release in cultured hippocampal synapses, we demonstrate a distinctive, release-dependent form of facilitation in which probability of vesicle release is higher following a successful glutamate release event than following a failure. This phenomenon has an onset of ≤500 ms and lasts several seconds, resulting in clusters of successful release events. The release-dependent facilitation requires neuronal contact with astrocytes and astrocytic glutamate uptake by EAAT1. It is not observed in neurons grown alone or in the presence of astrocyte-conditioned media. This form of facilitation dynamically amplifies multi-vesicular release. Facilitation-evoked release events exhibit spatial clustering and have a preferential localization toward the active zone center. These results uncover a rapid astrocyte-dependent form of facilitation acting via modulation of multi-vesicular release and displaying distinctive spatiotemporal properties.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63132, USA
| | - Vitaly A. Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63132, USA,Lead contact,Correspondence:
| |
Collapse
|
46
|
Browne LP, Crespo A, Grubb MS. Rapid presynaptic maturation in naturally regenerating axons of the adult mouse olfactory nerve. Cell Rep 2022; 41:111750. [PMID: 36476871 DOI: 10.1016/j.celrep.2022.111750] [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: 06/15/2022] [Revised: 09/26/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Successful neuronal regeneration requires the reestablishment of synaptic connectivity. This process requires the reconstitution of presynaptic neurotransmitter release, which we investigate here in a model of entirely natural regeneration. After toxin-induced injury, olfactory sensory neurons in the adult mouse olfactory epithelium can regenerate fully, sending axons via the olfactory nerve to reestablish synaptic contact with postsynaptic partners in the olfactory bulb. Using electrophysiological recordings in acute slices, we find that, after initial recontact, functional connectivity in this system is rapidly established. Reconnecting presynaptic terminals have almost mature functional properties, including high release probability and strong capacity for presynaptic inhibition. Release probability then matures quickly, rendering reestablished terminals functionally indistinguishable from controls just 1 week after initial contact. These data show that successful synaptic regeneration in the adult mammalian brain is almost a "plug-and-play" process, with presynaptic terminals undergoing a rapid phase of functional maturation as they reintegrate into target networks.
Collapse
Affiliation(s)
- Lorcan P Browne
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Andres Crespo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| |
Collapse
|
47
|
Bhattacharya D, Bartley AF, Li Q, Dobrunz LE. Bicuculline restores frequency-dependent hippocampal I/E ratio and circuit function in PGC-1ɑ null mice. Neurosci Res 2022; 184:9-18. [PMID: 35842011 PMCID: PMC10865982 DOI: 10.1016/j.neures.2022.07.003] [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: 06/03/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 10/31/2022]
Abstract
Altered inhibition/excitation (I/E) balance contributes to various brain disorders. Dysfunctional GABAergic interneurons enhance or reduce inhibition, resulting in I/E imbalances. Differences in short-term plasticity between excitation and inhibition cause frequency-dependence of the I/E ratio, which can be altered by GABAergic dysfunction. However, it is unknown whether I/E imbalances can be rescued pharmacologically using a single dose when the imbalance magnitude is frequency-dependent. Loss of PGC-1α (peroxisome proliferator activated receptor γ coactivator 1α) causes transcriptional dysregulation in hippocampal GABAergic interneurons. PGC-1α-/- slices have enhanced baseline inhibition onto CA1 pyramidal cells, causing increased I/E ratio and impaired circuit function. High frequency stimulation reduces the I/E ratio and recovers circuit function in PGC-1α-/- slices. Here we tested if using a low dose of bicuculline that can restore baseline I/E ratio can also rescue the frequency-dependent I/E imbalances in these mice. Remarkably, bicuculline did not reduce the I/E ratio below that of wild type during high frequency stimulation. Interestingly, bicuculline enhanced the paired-pulse ratio (PPR) of disynaptic inhibition without changing the monosynaptic inhibition PPR, suggesting that bicuculline modifies interneuron recruitment and not GABA release. Bicuculline improved CA1 output in PGC-1α-/- slices, enhancing EPSP-spike coupling to wild type levels at high and low frequencies. Our results show that it is possible to rescue frequency-dependent I/E imbalances in an animal model of transcriptional dysregulation with a single treatment.
Collapse
Affiliation(s)
- Dwipayan Bhattacharya
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Aundrea F Bartley
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Qin Li
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Lynn E Dobrunz
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States.
| |
Collapse
|
48
|
Chipman PH, Fetter RD, Panzera LC, Bergerson SJ, Karmelic D, Yokoyama S, Hoppa MB, Davis GW. NMDAR-dependent presynaptic homeostasis in adult hippocampus: Synapse growth and cross-modal inhibitory plasticity. Neuron 2022; 110:3302-3317.e7. [PMID: 36070750 PMCID: PMC9588671 DOI: 10.1016/j.neuron.2022.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/11/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
Homeostatic plasticity (HP) encompasses a suite of compensatory physiological processes that counteract neuronal perturbations, enabling brain resilience. Currently, we lack a complete description of the homeostatic processes that operate within the mammalian brain. Here, we demonstrate that acute, partial AMPAR-specific antagonism induces potentiation of presynaptic neurotransmitter release in adult hippocampus, a form of compensatory plasticity that is consistent with the expression of presynaptic homeostatic plasticity (PHP) documented at peripheral synapses. We show that this compensatory plasticity can be induced within minutes, requires postsynaptic NMDARs, and is expressed via correlated increases in dendritic spine volume, active zone area, and docked vesicle number. Further, simultaneous postsynaptic genetic reduction of GluA1, GluA2, and GluA3 in triple heterozygous knockouts induces potentiation of presynaptic release. Finally, induction of compensatory plasticity at excitatory synapses induces a parallel, NMDAR-dependent potentiation of inhibitory transmission, a cross-modal effect consistent with the anti-epileptic activity of AMPAR-specific antagonists used in humans.
Collapse
Affiliation(s)
- Peter H Chipman
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94941, USA
| | - Richard D Fetter
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94941, USA
| | - Lauren C Panzera
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Samuel J Bergerson
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Daniel Karmelic
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94941, USA
| | - Sae Yokoyama
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94941, USA
| | - Michael B Hoppa
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Graeme W Davis
- Department of Biochemistry and Biophysics, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94941, USA.
| |
Collapse
|
49
|
Dürst CD, Wiegert JS, Schulze C, Helassa N, Török K, Oertner TG. Vesicular release probability sets the strength of individual Schaffer collateral synapses. Nat Commun 2022; 13:6126. [PMID: 36253353 PMCID: PMC9576736 DOI: 10.1038/s41467-022-33565-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 09/22/2022] [Indexed: 12/24/2022] Open
Abstract
Information processing in the brain is controlled by quantal release of neurotransmitters, a tightly regulated process. From ultrastructural analysis, it is known that presynaptic boutons along single axons differ in the number of vesicles docked at the active zone. It is not clear whether the probability of these vesicles to get released (pves) is homogenous or also varies between individual boutons. Here, we optically measure evoked transmitter release at individual Schaffer collateral synapses at different calcium concentrations, using the genetically encoded glutamate sensor iGluSnFR. Fitting a binomial model to measured response amplitude distributions allowed us to extract the quantal parameters N, pves, and q. We find that Schaffer collateral boutons typically release single vesicles under low pves conditions and switch to multivesicular release in high calcium saline. The potency of individual boutons is highly correlated with their vesicular release probability while the number of releasable vesicles affects synaptic output only under high pves conditions.
Collapse
Affiliation(s)
- Céline D Dürst
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
- Department of Basic Neurosciences, Center for Neurosciences (CMU), University of Geneva, 1211, Geneva, Switzerland
| | - J Simon Wiegert
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
- Research Group Synaptic Wiring and Information Processing, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
| | - Christian Schulze
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany
| | - Nordine Helassa
- Cell Biology and Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, SW17 0RE, UK
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L69 3BX, UK
| | - Katalin Török
- Cell Biology and Genetics Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, SW17 0RE, UK
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology Hamburg (ZMNH), 20251, Hamburg, Germany.
| |
Collapse
|
50
|
Linders LE, Supiot LF, Du W, D’Angelo R, Adan RAH, Riga D, Meye FJ. Studying Synaptic Connectivity and Strength with Optogenetics and Patch-Clamp Electrophysiology. Int J Mol Sci 2022; 23:ijms231911612. [PMID: 36232917 PMCID: PMC9570045 DOI: 10.3390/ijms231911612] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 02/07/2023] Open
Abstract
Over the last two decades the combination of brain slice patch clamp electrophysiology with optogenetic stimulation has proven to be a powerful approach to analyze the architecture of neural circuits and (experience-dependent) synaptic plasticity in such networks. Using this combination of methods, originally termed channelrhodopsin-assisted circuit mapping (CRACM), a multitude of measures of synaptic functioning can be taken. The current review discusses their rationale, current applications in the field, and their associated caveats. Specifically, the review addresses: (1) How to assess the presence of synaptic connections, both in terms of ionotropic versus metabotropic receptor signaling, and in terms of mono- versus polysynaptic connectivity. (2) How to acquire and interpret measures for synaptic strength and function, like AMPAR/NMDAR, AMPAR rectification, paired-pulse ratio (PPR), coefficient of variance and input-specific quantal sizes. We also address how synaptic modulation by G protein-coupled receptors can be studied with pharmacological approaches and advanced technology. (3) Finally, we elaborate on advances on the use of dual color optogenetics in concurrent investigation of multiple synaptic pathways. Overall, with this review we seek to provide practical insights into the methods used to study neural circuits and synapses, by combining optogenetics and patch-clamp electrophysiology.
Collapse
|