1
|
Shao M, Botvinov J, Banerjee D, Girirajan S, Lüscher B. Transcriptome signatures of the medial prefrontal cortex underlying GABAergic control of resilience to chronic stress exposure. Mol Psychiatry 2025; 30:2197-2209. [PMID: 39550415 PMCID: PMC12014471 DOI: 10.1038/s41380-024-02832-x] [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: 07/10/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
Analyses of postmortem human brains and preclinical studies of rodents have identified somatostatin (SST)-positive, dendrite-targeting GABAergic interneurons as key elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, genetically induced disinhibition of SST neurons (induced by Cre-mediated deletion of the γ2 GABAA receptor subunit gene selectively from SST neurons, SSTCre:γ2f/f mice) results in stress resilience. Similarly, chronic chemogenetic activation of SST neurons in the medial prefrontal cortex (mPFC) results in stress resilience but only in male and not in female mice. Here, we used RNA sequencing of the mPFC of SSTCre:γ2f/f mice to characterize the transcriptome changes underlying GABAergic control of stress resilience. We found that stress resilience of male but not female SSTCre:γ2f/f mice is characterized by resilience to chronic stress-induced transcriptome changes in the mPFC. Interestingly, the transcriptome of non-stressed SSTCre:γ2f/f (stress-resilient) male mice resembled that of chronic stress-exposed SSTCre (stress-vulnerable) mice. However, the behavior and the serum corticosterone levels of non-stressed SSTCre:γ2f/f mice showed no signs of physiological stress. Most strikingly, chronic stress exposure of SSTCre:γ2f/f mice was associated with an almost complete reversal of their chronic stress-like transcriptome signature, along with pathway changes suggesting stress-induced enhancement of mRNA translation. Behaviorally, the SSTCre:γ2f/f mice were not only resilient to chronic stress-induced anhedonia - they also showed an inversed, anxiolytic-like behavioral response to chronic stress exposure that mirrored the chronic stress-induced reversal of the chronic stress-like transcriptome signature. We conclude that GABAergic dendritic inhibition by SST neurons exerts bidirectional control over behavioral vulnerability and resilience to chronic stress exposure that is mirrored in bidirectional changes in the expression of putative stress resilience genes, through a sex-specific brain substrate.
Collapse
Affiliation(s)
- Meiyu Shao
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Julia Botvinov
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Deepro Banerjee
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Santhosh Girirajan
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Lüscher
- Department of Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
2
|
Park E, Kuljis DA, Swindell RA, Ray A, Zhu M, Christian JA, Barth AL. Somatostatin neurons detect stimulus-reward contingencies to reduce neocortical inhibition during learning. Cell Rep 2025; 44:115606. [PMID: 40257862 DOI: 10.1016/j.celrep.2025.115606] [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: 04/12/2023] [Revised: 12/02/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
Learning involves the association of discrete events in the world to infer causality, likely through a cascade of changes at input- and target-specific synapses. Transient or sustained disinhibition may initiate cortical circuit plasticity important for association learning, but the cellular networks involved have not been well defined. Using recordings in acute brain slices, we show that whisker-dependent sensory association learning drives a durable, target-specific reduction in inhibition from somatostatin (SST)-expressing GABAergic neurons onto pyramidal (Pyr) neurons in superficial but not deep layers of mouse somatosensory cortex. Critically, SST output was not altered when stimuli and rewards were unpaired, indicating that these neurons are sensitive to stimulus-reward contingency. Depression of SST output onto Pyr neurons could be phenocopied by chemogenetic suppression of SST activity outside of the training context. Thus, neocortical SST neuron output can undergo long-lasting modifications to selectively disinhibit superficial layers of sensory neocortex during learning.
Collapse
Affiliation(s)
- Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Dika A Kuljis
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Rachel A Swindell
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Mo Zhu
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Joseph A Christian
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
3
|
Song SC, Froemke RC. Lateralized local circuit tuning in female mouse auditory cortex. Neurosci Res 2025:S0168-0102(25)00068-9. [PMID: 40189152 DOI: 10.1016/j.neures.2025.03.009] [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: 03/28/2025] [Revised: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Most offspring are born helpless, requiring intense caregiving from parents especially during the first few days of neonatal life. For many species, infant cries are a primary signal used by parents to provide caregiving. Previously we and others documented how maternal left auditory cortex rapidly becomes sensitized to pup calls over hours of parental experience, enabled by oxytocin. The speed and robustness of this maternal plasticity suggests cortical pre-tuning or initial bias for pup call stimulus features. Here we examine the circuit basis of left-lateralized tuning to vocalization features with whole-cell recordings in brain slices. We found that layer 2/3 pyramidal cells of female left auditory cortex show selective suppression of inhibitory inputs with repeated stimulation at the fundamental pup call rate (inter-stimulus interval ∼150 msec) in pup-naïve females and expanded with maternal experience. However, optogenetic stimulation of cortical inhibitory cells showed that inputs from somatostatin-positive and oxytocin-receptor-expressing interneurons were less suppressed at these rates. This suggested that disynaptic inhibition rather than monosynaptic depression was a major mechanism underlying pre-tuning of cortical excitatory neurons, confirmed with simulations. Thus cortical interneuron specializations can augment neuroplasticity mechanisms to ensure fast appropriate caregiving in response to infant cries.
Collapse
Affiliation(s)
- Soomin C Song
- Ion Laboratory, New York University Langone Health, New York, NY, USA; Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Robert C Froemke
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
4
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. Neuropsychopharmacology 2025; 50:673-684. [PMID: 39390105 PMCID: PMC11845475 DOI: 10.1038/s41386-024-02002-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA.
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT, 06519, USA
| |
Collapse
|
5
|
Welle TM, Smith KR. Release your inhibitions: The cell biology of GABAergic postsynaptic plasticity. Curr Opin Neurobiol 2025; 90:102952. [PMID: 39721557 PMCID: PMC11839402 DOI: 10.1016/j.conb.2024.102952] [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: 09/20/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
GABAergic synaptic inhibition controls circuit function by regulating neuronal plasticity, excitability, and firing. To achieve these goals, inhibitory synapses themselves undergo several forms of plasticity via diverse mechanisms, strengthening and weakening phasic inhibition in response to numerous activity-induced stimuli. These mechanisms include changing the number and arrangement of functional GABAARs within the inhibitory postsynaptic domain (iPSD), which can profoundly regulate inhibitory synapse strength. Here, we explore recent advances in our molecular understanding of inhibitory postsynaptic plasticity, with a focus on modulation of the trafficking, protein-protein interactions, nanoscale-organization, and posttranscriptional regulation of GABAARs and iPSD proteins. What has emerged is a complex mechanistic picture of how synaptic inhibition is controlled, with critical ramifications for cognition under typical and pathogenic conditions.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
6
|
Michałowski MA, Kłopotowski K, Wiera G, Czyżewska MM, Mozrzymas JW. Molecular mechanisms of the GABA type A receptor function. Q Rev Biophys 2025; 58:e3. [PMID: 39806800 DOI: 10.1017/s0033583524000179] [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: 01/16/2025]
Abstract
The GABA type A receptor (GABAAR) belongs to the family of pentameric ligand-gated ion channels and plays a key role in inhibition in adult mammalian brains. Dysfunction of this macromolecule may lead to epilepsy, anxiety disorders, autism, depression, and schizophrenia. GABAAR is also a target for multiple physiologically and clinically relevant modulators, such as benzodiazepines (BDZs), general anesthetics, and neurosteroids. The first GABAAR structure appeared in 2014, but the past years have brought a particularly abundant surge in structural data for these receptors with various ligands and modulators. Although the open conformation remains elusive, this novel information has pushed the structure-function studies to an unprecedented level. Electrophysiology, mutagenesis, photolabeling, and in silico simulations, guided by novel structural information, shed new light on the molecular mechanisms of receptor functioning. The main goal of this review is to present the current knowledge of GABAAR functional and structural properties. The review begins with an outline of the functional and structural studies of GABAAR, accompanied by some methodological considerations, especially biophysical methods, enabling the reader to follow how major breakthroughs in characterizing GABAAR features have been achieved. The main section provides a comprehensive analysis of the functional significance of specific structural elements in GABAARs. We additionally summarize the current knowledge on the binding sites for major GABAAR modulators, referring to the molecular underpinnings of their action. The final chapter of the review moves beyond examining GABAAR as an isolated macromolecule and describes the interactions of the receptor with other proteins in a broader context of inhibitory plasticity. In the final section, we propose a general conclusion that agonist binding to the orthosteric binding sites appears to rely on local interactions, whereas conformational transitions of bound macromolecule (gating) and allosteric modulation seem to reflect more global phenomena involving vast portions of the macromolecule.
Collapse
Affiliation(s)
- Michał A Michałowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Karol Kłopotowski
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Grzegorz Wiera
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Marta M Czyżewska
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | - Jerzy W Mozrzymas
- Faculty of Medicine, Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
7
|
Krueger-Burg D. Understanding GABAergic synapse diversity and its implications for GABAergic pharmacotherapy. Trends Neurosci 2025; 48:47-61. [PMID: 39779392 DOI: 10.1016/j.tins.2024.11.007] [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: 04/11/2024] [Revised: 10/17/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Despite the substantial contribution of disruptions in GABAergic inhibitory neurotransmission to the etiology of psychiatric, neurodevelopmental, and neurodegenerative disorders, surprisingly few drugs targeting the GABAergic system are currently available, partly due to insufficient understanding of circuit-specific GABAergic synapse biology. In addition to GABA receptors, GABAergic synapses contain an elaborate organizational protein machinery that regulates the properties of synaptic transmission. Until recently, this machinery remained largely unexplored, but key methodological advances have now led to the identification of a wealth of new GABAergic organizer proteins. Notably, many of these proteins appear to function only at specific subsets of GABAergic synapses, creating a diversity of organizer complexes that may serve as circuit-specific targets for pharmacotherapies. The present review aims to summarize the methodological developments that underlie this newfound knowledge and provide a current overview of synapse-specific GABAergic organizer complexes, as well as outlining future avenues and challenges in translating this knowledge into clinical applications.
Collapse
Affiliation(s)
- Dilja Krueger-Burg
- Laboratory of Cell Biology and Neuroscience, Institute of Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, 55128 Mainz, Germany.
| |
Collapse
|
8
|
Kitchigina VF. Colocalization of Neurotransmitters in Hippocampus and Afferent Systems: Possible Functional Role. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:61-78. [PMID: 40058974 DOI: 10.1134/s0006297924603915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 05/13/2025]
Abstract
In neurophysiology, the transmitter phenotype is considered as an indicator of neuronal identity. It has become known at the end of last century that a nerve cell can produce and use several different molecules to communicate with other neurons. These could be "classical" transmitters: glutamate or gamma-aminobutyric acid (or acetylcholine, serotonin, norepinephrine), as well as secondary messengers, mainly neuropeptides released from the same neurons. In the case, when classical neurotransmitters are released together from the same nerve cell, this event is called cotransmission or corelease (release from the same vesicles). In this review article, the term "cotransmission" is used in a broad sense, denoting neurons that can release more than one classical mediator. Since transmitters are often intermediate products of metabolism and are found in many cells, the neuron classification is currently based on the carrier proteins (transporters) that "pack" neurotransmitters synthesized in the cytoplasm into vesicles. Here, we limit the issue of colocalization of the main neurotransmitters in mammals to the neurons of hippocampus and those structures that send their pathways to it. The review considers problems concerning the mechanisms of multitransmitter signaling, as well as probable functional role of mediator colocalization in the work of hippocampus, which yet has been poorly understood. It has been suggested that co-expression of different mediator phenotypes is involved in maintaining the balance of excitation and inhibition in different regions of hippocampus, facilitates rapid selection of information processing mode, induction of long-term potentiation, maintenance of spatial coding by place cells, as well as ensuring flexibility of learning and formation of working memory. However, the functional role of mediator colocalization, as well as the mechanisms of release of "dual" transmitters, have not been fully elucidated. The solution of these problems will advance some areas of fundamental neuroscience and help in the treatment of those diseases, where disruption of the balance between excitation and inhibition is detected, such as, for example, in epilepsy, Alzheimer's disease, and many others.
Collapse
Affiliation(s)
- Valentina F Kitchigina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
9
|
Cupolillo D, Regio V, Barberis A. Synaptic microarchitecture: the role of spatial interplay between excitatory and inhibitory inputs in shaping dendritic plasticity and neuronal output. Front Cell Neurosci 2024; 18:1513602. [PMID: 39758273 PMCID: PMC11695373 DOI: 10.3389/fncel.2024.1513602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Affiliation(s)
| | | | - Andrea Barberis
- Istituto Italiano di Tecnologia, Synaptic Plasticity of Inhibitory Networks, Genova, Italy
| |
Collapse
|
10
|
Ceballos CC, Ma L, Qin M, Zhong H. Widespread co-release of glutamate and GABA throughout the mouse brain. Commun Biol 2024; 7:1502. [PMID: 39537846 PMCID: PMC11560972 DOI: 10.1038/s42003-024-07198-y] [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: 06/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Several brain neuronal populations transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA. However, it remains largely unknown whether these opposing neurotransmitters are co-released simultaneously or are independently transmitted at different times and locations. By recording from acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons. This observation cannot be explained by accidental coincidence of monophasic excitatory and inhibitory minis. Interestingly, these biphasic minis could either be an excitatory current leading an inhibitory current or vice versa. Deletion of dopaminergic neurons did not eliminate biphasic minis, indicating that they originate from another source. Importantly, we found that both types of biphasic minis were present in multiple striatal neuronal types and in nine out of ten other brain regions. Overall, co-release of glutamate and GABA appears to be a widespread mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
11
|
Asim M, Qianqian G, Waris A, Wang H, Lai Y, Chen X. Unraveling the role of cholecystokinin in epilepsy: Mechanistic insight into neuroplasticity. Neurochem Int 2024; 180:105870. [PMID: 39343303 DOI: 10.1016/j.neuint.2024.105870] [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: 08/15/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Epilepsy is a disorder characterized by an imbalance between excitability and inhibition, leading to uncontrolled hyperexcitability of neurons in the central nervous system. Despite the prevalence of epileptic seizures, the underlying mechanisms driving this hyperexcitability remain poorly understood. This review article aims to enhance our understanding of the mechanisms of epilepsy, with a specific focus on the role of cholecystokinin (CCK) in this debilitating disease. We will begin with an introduction to the topic, followed by an examination of the role of GABAergic neurons and the synaptic plasticity mechanisms associated with seizures. As we delve deeper, we will elucidate how CCK and its receptors contribute to seizure behavior. Finally, we will discuss the CCK-dependent synaptic plasticity mechanisms and highlight their potential implications in seizure activity. Through a comprehensive examination of these aspects, this review provides valuable insights into the involvement of CCK and its receptors in epilepsy. By improving our understanding of the mechanisms underlying this condition, particularly the role of CCK, we aim to contribute to the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong.
| | - Gao Qianqian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Abdul Waris
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Huajie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Yuanying Lai
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong
| |
Collapse
|
12
|
Welle TM, Rajgor D, Kareemo DJ, Garcia JD, Zych SM, Wolfe SE, Gookin SE, Martinez TP, Dell'Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. EMBO Rep 2024; 25:5141-5168. [PMID: 39294503 PMCID: PMC11549329 DOI: 10.1038/s44319-024-00253-z] [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: 12/15/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024] Open
Abstract
Activity-dependent protein synthesis is crucial for long-lasting forms of synaptic plasticity. However, our understanding of translational mechanisms controlling GABAergic synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the mechanisms controlling plasticity-induced gephyrin translation remain unknown. We identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting inhibitory synaptic structure and function. iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and promoting de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Together, we delineate a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah M Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah E Wolfe
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sara E Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Tyler P Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Wiera G, Jabłońska J, Lech AM, Mozrzymas JW. Input specificity of NMDA-dependent GABAergic plasticity in the hippocampus. Sci Rep 2024; 14:20463. [PMID: 39242672 PMCID: PMC11379801 DOI: 10.1038/s41598-024-70278-w] [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: 04/12/2024] [Accepted: 08/14/2024] [Indexed: 09/09/2024] Open
Abstract
Sensory experiences and learning induce long-lasting changes in both excitatory and inhibitory synapses, thereby providing a crucial substrate for memory. However, the co-tuning of excitatory long-term potentiation (eLTP) or depression (eLTD) with the simultaneous changes at inhibitory synapses (iLTP/iLTD) remains unclear. Herein, we investigated the co-expression of NMDA-induced synaptic plasticity at excitatory and inhibitory synapses in hippocampal CA1 pyramidal cells (PCs) using a combination of electrophysiological, optogenetic, and pharmacological approaches. We found that inhibitory inputs from somatostatin (SST) and parvalbumin (PV)-positive interneurons onto CA1 PCs display input-specific long-term plastic changes following transient NMDA receptor activation. Notably, synapses from SST-positive interneurons consistently exhibited iLTP, irrespective of the direction of excitatory plasticity, whereas synapses from PV-positive interneurons predominantly showed iLTP concurrent with eLTP, rather than eLTD. As neuroplasticity is known to depend on the extracellular matrix, we tested the impact of metalloproteinases (MMP) inhibition. MMP3 blockade interfered with GABAergic plasticity for all inhibitory inputs, whereas MMP9 inhibition selectively blocked eLTP and iLTP in SST-CA1PC synapses co-occurring with eLTP but not eLTD. These findings demonstrate the dissociation of excitatory and inhibitory plasticity co-expression. We propose that these mechanisms of plasticity co-expression may be involved in maintaining excitation-inhibition balance and modulating neuronal integration modes.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368, Wroclaw, Poland.
| | - Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368, Wroclaw, Poland
| | - Anna Maria Lech
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368, Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368, Wroclaw, Poland.
| |
Collapse
|
14
|
Wen W, Turrigiano GG. Keeping Your Brain in Balance: Homeostatic Regulation of Network Function. Annu Rev Neurosci 2024; 47:41-61. [PMID: 38382543 DOI: 10.1146/annurev-neuro-092523-110001] [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: 02/23/2024]
Abstract
To perform computations with the efficiency necessary for animal survival, neocortical microcircuits must be capable of reconfiguring in response to experience, while carefully regulating excitatory and inhibitory connectivity to maintain stable function. This dynamic fine-tuning is accomplished through a rich array of cellular homeostatic plasticity mechanisms that stabilize important cellular and network features such as firing rates, information flow, and sensory tuning properties. Further, these functional network properties can be stabilized by different forms of homeostatic plasticity, including mechanisms that target excitatory or inhibitory synapses, or that regulate intrinsic neuronal excitability. Here we discuss which aspects of neocortical circuit function are under homeostatic control, how this homeostasis is realized on the cellular and molecular levels, and the pathological consequences when circuit homeostasis is impaired. A remaining challenge is to elucidate how these diverse homeostatic mechanisms cooperate within complex circuits to enable them to be both flexible and stable.
Collapse
Affiliation(s)
- Wei Wen
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| | - Gina G Turrigiano
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
15
|
Fogaça MV, Daher F, Picciotto MR. Effects of ketamine on GABAergic and glutamatergic activity in the mPFC: biphasic recruitment of GABA function in antidepressant-like responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605610. [PMID: 39131322 PMCID: PMC11312475 DOI: 10.1101/2024.07.29.605610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Major depressive disorder (MDD) is associated with disruptions in glutamatergic and GABAergic activity in the medial prefrontal cortex (mPFC), leading to altered synaptic formation and function. Low doses of ketamine rapidly rescue these deficits, inducing fast and sustained antidepressant effects. While it is suggested that ketamine produces a rapid glutamatergic enhancement in the mPFC, the temporal dynamics and the involvement of GABA interneurons in its sustained effects remain unclear. Using simultaneous photometry recordings of calcium activity in mPFC pyramidal and GABA neurons, as well as chemogenetic approaches in Gad1-Cre mice, we explored the hypothesis that initial effects of ketamine on glutamate signaling trigger subsequent enhancement of GABAergic responses, contributing to its sustained antidepressant responses. Calcium recordings revealed a biphasic effect of ketamine on activity of mPFC GABA neurons, characterized by an initial transient decrease (phase 1, <30 min) followed by an increase (phase 2, >60 min), in parallel with a transient increase in excitation/inhibition levels (10 min) and lasting enhancement of glutamatergic activity (30-120 min). Previous administration of ketamine enhanced GABA neuron activity during the sucrose splash test (SUST) and novelty suppressed feeding test (NSFT), 24 h and 72 h post-treatment, respectively. Chemogenetic inhibition of GABA interneurons during the surge of GABAergic activity (phase 2), or immediately before the SUST or NSFT, occluded ketamine's behavioral actions. These results indicate that time-dependent modulation of GABAergic activity is required for the sustained antidepressant-like responses induced by ketamine, suggesting that approaches to enhance GABAergic plasticity and function are promising therapeutic targets for antidepressant development.
Collapse
Affiliation(s)
- Manoela V. Fogaça
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| | - Fernanda Daher
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06519, USA
| |
Collapse
|
16
|
Horton S, Mastrolia V, Jackson RE, Kemlo S, Pereira Machado PM, Carbajal MA, Hindges R, Fleck RA, Aguiar P, Neves G, Burrone J. Excitatory and inhibitory synapses show a tight subcellular correlation that weakens over development. Cell Rep 2024; 43:114361. [PMID: 38900634 DOI: 10.1016/j.celrep.2024.114361] [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: 02/09/2024] [Revised: 04/24/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024] Open
Abstract
Neurons receive correlated levels of excitation and inhibition, a feature that is important for proper brain function. However, how this relationship between excitatory and inhibitory inputs is established during the dynamic period of circuit wiring remains unexplored. Using multiple techniques, including in utero electroporation, electron microscopy, and electrophysiology, we reveal a tight correlation in the distribution of excitatory and inhibitory synapses along the dendrites of developing CA1 hippocampal neurons. This correlation was present within short dendritic stretches (<20 μm) and, surprisingly, was most pronounced during early development, sharply declining with maturity. The tight matching between excitation and inhibition was unexpected, as inhibitory synapses lacked an active zone when formed and exhibited compromised evoked release. We propose that inhibitory synapses form as a stabilizing scaffold to counterbalance growing excitation levels. This relationship diminishes over time, suggesting a critical role for a subcellular balance in early neuronal function and circuit formation.
Collapse
Affiliation(s)
- 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
| | - Vincenzo Mastrolia
- 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
| | - Rachel E Jackson
- 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
| | - Sarah Kemlo
- 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
| | - Robert Hindges
- 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
| | - Roland A Fleck
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Paulo Aguiar
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - 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.
| | - 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
|
17
|
Kulbay M, Tuli N, Akdag A, Kahn Ali S, Qian CX. Optogenetics and Targeted Gene Therapy for Retinal Diseases: Unravelling the Fundamentals, Applications, and Future Perspectives. J Clin Med 2024; 13:4224. [PMID: 39064263 PMCID: PMC11277578 DOI: 10.3390/jcm13144224] [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: 06/18/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
With a common aim of restoring physiological function of defective cells, optogenetics and targeted gene therapies have shown great clinical potential and novelty in the branch of personalized medicine and inherited retinal diseases (IRDs). The basis of optogenetics aims to bypass defective photoreceptors by introducing opsins with light-sensing capabilities. In contrast, targeted gene therapies, such as methods based on CRISPR-Cas9 and RNA interference with noncoding RNAs (i.e., microRNA, small interfering RNA, short hairpin RNA), consists of inducing normal gene or protein expression into affected cells. Having partially leveraged the challenges limiting their prompt introduction into the clinical practice (i.e., engineering, cell or tissue delivery capabilities), it is crucial to deepen the fields of knowledge applied to optogenetics and targeted gene therapy. The aim of this in-depth and novel literature review is to explain the fundamentals and applications of optogenetics and targeted gene therapies, while providing decision-making arguments for ophthalmologists. First, we review the biomolecular principles and engineering steps involved in optogenetics and the targeted gene therapies mentioned above by bringing a focus on the specific vectors and molecules for cell signalization. The importance of vector choice and engineering methods are discussed. Second, we summarize the ongoing clinical trials and most recent discoveries for optogenetics and targeted gene therapies for IRDs. Finally, we then discuss the limits and current challenges of each novel therapy. We aim to provide for the first time scientific-based explanations for clinicians to justify the specificity of each therapy for one disease, which can help improve clinical decision-making tasks.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3S5, Canada;
| | - Nicolas Tuli
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (A.A.)
| | - Arjin Akdag
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada (A.A.)
| | - Shigufa Kahn Ali
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, QC H1T 2M4, Canada;
| | - Cynthia X. Qian
- Centre de Recherche de l’Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, QC H1T 2M4, Canada;
- Department of Ophthalmology, Centre Universitaire d’Ophtalmologie (CUO), Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, QC H1T 2M4, Canada
| |
Collapse
|
18
|
Shao M, Botvinov J, Banerjee D, Girirajan S, Lüscher B. Transcriptome signatures of the medial prefrontal cortex underlying GABAergic control of resilience to chronic stress exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602959. [PMID: 39026878 PMCID: PMC11257543 DOI: 10.1101/2024.07.10.602959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Analyses of postmortem human brains and preclinical studies of rodents have identified somatostatin (SST)-positive interneurons as key elements that regulate the vulnerability to stress-related psychiatric disorders. Conversely, genetically induced disinhibition of SST neurons or brain region-specific chemogenetic activation of SST neurons in mice results in stress resilience. Here, we used RNA sequencing of mice with disinhibited SST neurons to characterize the transcriptome changes underlying GABAergic control of stress resilience. We found that stress resilience of male but not female mice with disinhibited SST neurons is characterized by resilience to chronic stress-induced transcriptome changes in the medial prefrontal cortex. Interestingly, the transcriptome of non-stressed stress-resilient male mice resembled the transcriptome of chronic stress-exposed stress-vulnerable mice. However, the behavior and the serum corticosterone levels of non-stressed stress-resilient mice showed no signs of physiological stress. Most strikingly, chronic stress exposure of stress-resilient mice was associated with an almost complete reversal of their chronic stress-like transcriptome signature, along with pathway changes indicating stress-induced enhancement of mRNA translation. Behaviorally, the mice with disinhibited SST neurons were not only resilient to chronic stress-induced anhedonia - they also showed an inversed anxiolytic-like response to chronic stress exposure that mirrored the chronic stress-induced reversal of the chronic stress-like transcriptome signature. We conclude that GABAergic dendritic inhibition by SST neurons exerts bidirectional control over behavioral vulnerability and resilience to chronic stress exposure that is mirrored in bidirectional changes in expression of putative stress resilience genes, through a sex-specific brain substrate.
Collapse
Affiliation(s)
- Meiyu Shao
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| | - Julia Botvinov
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
| | - Deepro Banerjee
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| | - Santhosh Girirajan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| | - Bernhard Lüscher
- Department of Biology, The Pennsylvania State University, University Park, PA 16802
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
19
|
Pinto MJ, Bizien L, Fabre JM, Ðukanović N, Lepetz V, Henderson F, Pujol M, Sala RW, Tarpin T, Popa D, Triller A, Léna C, Fabre V, Bessis A. Microglial TNFα controls daily changes in synaptic GABAARs and sleep slow waves. J Cell Biol 2024; 223:e202401041. [PMID: 38695719 PMCID: PMC11070559 DOI: 10.1083/jcb.202401041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 05/08/2024] Open
Abstract
Microglia sense the changes in their environment. How microglia actively translate these changes into suitable cues to adapt brain physiology is unknown. We reveal an activity-dependent regulation of cortical inhibitory synapses by microglia, driven by purinergic signaling acting on P2RX7 and mediated by microglia-derived TNFα. We demonstrate that sleep induces microglia-dependent synaptic enrichment of GABAARs in a manner dependent on microglial TNFα and P2RX7. We further show that microglia-specific depletion of TNFα alters slow waves during NREM sleep and blunt memory consolidation in sleep-dependent learning tasks. Together, our results reveal that microglia orchestrate sleep-intrinsic plasticity of synaptic GABAARs, sculpt sleep slow waves, and support memory consolidation.
Collapse
Affiliation(s)
- Maria Joana Pinto
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Lucy Bizien
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Julie M.J. Fabre
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Nina Ðukanović
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Valentin Lepetz
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Fiona Henderson
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Marine Pujol
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Romain W. Sala
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Thibault Tarpin
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Daniela Popa
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Antoine Triller
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Clément Léna
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Véronique Fabre
- Neurosciences Paris Seine—Institut de Biologie Paris Seine (NPS—IBPS), CNRS, INSERM, Sorbonne Universités, Paris, France
| | - Alain Bessis
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
20
|
Burch AM, Garcia JD, O'Leary H, Haas A, Orfila JE, Tiemeier E, Chalmers N, Smith KR, Quillinan N, Herson PS. TRPM2 and CaMKII Signaling Drives Excessive GABAergic Synaptic Inhibition Following Ischemia. J Neurosci 2024; 44:e1762232024. [PMID: 38565288 PMCID: PMC11079974 DOI: 10.1523/jneurosci.1762-23.2024] [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: 09/22/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Excitotoxicity and the concurrent loss of inhibition are well-defined mechanisms driving acute elevation in excitatory/inhibitory (E/I) balance and neuronal cell death following an ischemic insult to the brain. Despite the high prevalence of long-term disability in survivors of global cerebral ischemia (GCI) as a consequence of cardiac arrest, it remains unclear whether E/I imbalance persists beyond the acute phase and negatively affects functional recovery. We previously demonstrated sustained impairment of long-term potentiation (LTP) in hippocampal CA1 neurons correlating with deficits in learning and memory tasks in a murine model of cardiac arrest/cardiopulmonary resuscitation (CA/CPR). Here, we use CA/CPR and an in vitro ischemia model to elucidate mechanisms by which E/I imbalance contributes to ongoing hippocampal dysfunction in male mice. We reveal increased postsynaptic GABAA receptor (GABAAR) clustering and function in the CA1 region of the hippocampus that reduces the E/I ratio. Importantly, reduced GABAAR clustering observed in the first 24 h rebounds to an elevation of GABAergic clustering by 3 d postischemia. This increase in GABAergic inhibition required activation of the Ca2+-permeable ion channel transient receptor potential melastatin-2 (TRPM2), previously implicated in persistent LTP and memory deficits following CA/CPR. Furthermore, we find Ca2+-signaling, likely downstream of TRPM2 activation, upregulates Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity, thereby driving the elevation of postsynaptic inhibitory function. Thus, we propose a novel mechanism by which inhibitory synaptic strength is upregulated in the context of ischemia and identify TRPM2 and CaMKII as potential pharmacological targets to restore perturbed synaptic plasticity and ameliorate cognitive function.
Collapse
Affiliation(s)
- Amelia M Burch
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Heather O'Leary
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Ami Haas
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - James E Orfila
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Erika Tiemeier
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nicholas Chalmers
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Nidia Quillinan
- Neuronal Injury & Plasticity Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Paco S Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, Ohio 43210
| |
Collapse
|
21
|
Kundu S, Paul B, Reuevni I, Lamprecht R, Barkai E. Learning-induced bidirectional enhancement of inhibitory synaptic metaplasticity. J Physiol 2024; 602:2343-2358. [PMID: 38654583 DOI: 10.1113/jp284761] [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: 03/28/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Training rodents in a particularly difficult olfactory-discrimination (OD) task results in the acquisition of the ability to perform the task well, termed 'rule learning'. In addition to enhanced intrinsic excitability and synaptic excitation in piriform cortex pyramidal neurons, rule learning results in increased synaptic inhibition across the whole cortical network to the point where it precisely maintains the balance between inhibition and excitation. The mechanism underlying such precise inhibitory enhancement remains to be explored. Here, we use brain slices from transgenic mice (VGAT-ChR2-EYFP), enabling optogenetic stimulation of single GABAergic neurons and recordings of unitary synaptic events in pyramidal neurons. Quantal analysis revealed that learning-induced enhanced inhibition is mediated by increased quantal size of the evoked inhibitory events. Next, we examined the plasticity of synaptic inhibition induced by long-lasting, intrinsically evoked spike firing in post-synaptic neurons. Repetitive depolarizing current pulses from depolarized (-70 mV) or hyperpolarized (-90 mV) membrane potentials induced long-term depression (LTD) and long-term potentiation (LTP) of synaptic inhibition, respectively. We found a profound bidirectional increase in the ability to induce both LTD, mediated by L-type calcium channels, and LTP, mediated by R-type calcium channels after rule learning. Blocking the GABAB receptor reversed the effect of intrinsic stimulation at -90 mV from LTP to LTD. We suggest that learning greatly enhances the ability to modify the strength of synaptic inhibition of principal neurons in both directions. Such plasticity of synaptic plasticity allows fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule. KEY POINTS: Olfactory discrimination rule learning results in long-lasting enhancement of synaptic inhibition on piriform cortex pyramidal neurons. Quantal analysis of unitary inhibitory synaptic events, evoked by optogenetic minimal stimulation, revealed that enhanced synaptic inhibition is mediated by increased quantal size. Surprisingly, metaplasticity of synaptic inhibition, induced by intrinsically evoked repetitive spike firing, is increased bidirectionally. The susceptibility to both long-term depression (LTD) and long-term potentiation (LTP) of inhibition is enhanced after learning. LTD of synaptic inhibition is mediated by L-type calcium channels and LTP by R-type calcium channels. LTP is also dependent on activation of GABAB receptors. We suggest that learning-induced changes in the metaplasticity of synaptic inhibition enable the fine-tuning of inhibition on each particular neuron, thereby stabilizing the network while maintaining the memory of the rule.
Collapse
Affiliation(s)
- Sankhanava Kundu
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Blesson Paul
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Iris Reuevni
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
22
|
McFarlan AR, Guo C, Gomez I, Weinerman C, Liang TA, Sjöström PJ. The spike-timing-dependent plasticity of VIP interneurons in motor cortex. Front Cell Neurosci 2024; 18:1389094. [PMID: 38706517 PMCID: PMC11066220 DOI: 10.3389/fncel.2024.1389094] [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: 02/20/2024] [Accepted: 04/09/2024] [Indexed: 05/07/2024] Open
Abstract
The plasticity of inhibitory interneurons (INs) plays an important role in the organization and maintenance of cortical microcircuits. Given the many different IN types, there is an even greater diversity in synapse-type-specific plasticity learning rules at excitatory to excitatory (E→I), I→E, and I→I synapses. I→I synapses play a key disinhibitory role in cortical circuits. Because they typically target other INs, vasoactive intestinal peptide (VIP) INs are often featured in I→I→E disinhibition, which upregulates activity in nearby excitatory neurons. VIP IN dysregulation may thus lead to neuropathologies such as epilepsy. In spite of the important activity regulatory role of VIP INs, their long-term plasticity has not been described. Therefore, we characterized the phenomenology of spike-timing-dependent plasticity (STDP) at inputs and outputs of genetically defined VIP INs. Using a combination of whole-cell recording, 2-photon microscopy, and optogenetics, we explored I→I STDP at layer 2/3 (L2/3) VIP IN outputs onto L5 Martinotti cells (MCs) and basket cells (BCs). We found that VIP IN→MC synapses underwent causal long-term depression (LTD) that was presynaptically expressed. VIP IN→BC connections, however, did not undergo any detectable plasticity. Conversely, using extracellular stimulation, we explored E→I STDP at inputs to VIP INs which revealed long-term potentiation (LTP) for both causal and acausal timings. Taken together, our results demonstrate that VIP INs possess synapse-type-specific learning rules at their inputs and outputs. This suggests the possibility of harnessing VIP IN long-term plasticity to control activity-related neuropathologies such as epilepsy.
Collapse
Affiliation(s)
- Amanda R. McFarlan
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Connie Guo
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Isabella Gomez
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Chaim Weinerman
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Tasha A. Liang
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - P. Jesper Sjöström
- Centre for Research in Neuroscience, BRaIN Program, Department of Neurology and Neurosurgery, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| |
Collapse
|
23
|
Pinto MJ, Ragozzino D, Bessis A, Audinat E. Microglial Modulation of Synaptic Maturation, Activity, and Plasticity. ADVANCES IN NEUROBIOLOGY 2024; 37:209-219. [PMID: 39207694 DOI: 10.1007/978-3-031-55529-9_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia, which are the resident immune cells of the CNS, also have important functions in physiological conditions. In this chapter, we review the experimental evidence that microglia modulate neuronal and synaptic activity during normal development and in adults. We show that microglia can regulate the maturation and function of both inhibitory and excitatory synapses that can be stimulated or repressed. We further review the fact that these regulations occur in various brain regions, through soluble and membrane molecules, directly or through other cell partners. This review emphasizes the fact that microglia are genuine and highly context-dependent and thus adaptable regulators of neuronal activity.
Collapse
Affiliation(s)
- Maria Joana Pinto
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
- Center for Neuroscience and Cell Biology (CNC), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
| | - Alain Bessis
- Institut de Biologie de l'École normale supérieure (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Etienne Audinat
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
24
|
Welle TM, Rajgor D, Garcia JD, Kareemo D, Zych SM, Gookin SE, Martinez TP, Dell’Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.570420. [PMID: 38168421 PMCID: PMC10760056 DOI: 10.1101/2023.12.12.570420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Activity-dependent protein synthesis is crucial for many long-lasting forms of synaptic plasticity. However, our understanding of the translational mechanisms controlling inhibitory synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the precise mechanisms controlling gephyrin translation during this process remain unknown. Here, we identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting GABAergic synaptic structure and function. We find that iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and allowing for increased de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Overall, this work delineates a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M. Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Joshua D. Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Dean Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sarah M. Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sara E. Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Tyler P. Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Matthew J. Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| |
Collapse
|
25
|
Shao W, Zheng H, Zhu J, Li W, Li Y, Hu W, Zhang J, Jing L, Wang K, Jiang X. Deletions of Cacna2d3 in parvalbumin-expressing neurons leads to autistic-like phenotypes in mice. Neurochem Int 2023; 169:105569. [PMID: 37419212 DOI: 10.1016/j.neuint.2023.105569] [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: 03/30/2023] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Autism spectrum disorder (ASD) is a series of highly inherited neurodevelopmental disorders. Loss-of-function (LOF) mutations in the CACNA2D3 gene are associated with ASD. However, the underlying mechanism is unknown. Dysfunction of cortical interneurons (INs) is strongly implicated in ASD. Parvalbumin-expressing (PV) INs and somatostatin-expressing (SOM) INs are the two most subtypes. Here, we characterized a mouse knockout of the Cacna2d3 gene in PV-expressing neurons (PVCre;Cacna2d3f/f mice) or in SOM-expressing neurons (SOMCre;Cacna2d3f/f mice), respectively. PVCre;Cacna2d3f/f mice showed deficits in the core ASD behavioral domains (including impaired sociability and increased repetitive behavior), as well as anxiety-like behavior and improved spatial memory. Furthermore, loss of Cacna2d3 from a subset of PV neurons results in a reduction of GAD67 and PV expression in the medial prefrontal cortex (mPFC). These may underlie the increased neuronal excitability in the mPFC, which contribute to the abnormal social behavior in PVCre;Cacna2d3f/f mice. Whereas, SOMCre;Cacna2d3f/f mice showed no obvious deficits in social, cognitive, or emotional phenotypes. Our findings provide the first evidence suggesting the causal role of Cacna2d3 insufficiency in PV neurons in autism.
Collapse
Affiliation(s)
- Wei Shao
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
| | - Hang Zheng
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
| | - Jingwen Zhu
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
| | - Wenhao Li
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
| | - Yifan Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juanjuan Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liang Jing
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China.
| | - Kai Wang
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Center for Neuropsychiatric Disorders and Mental Health, Hefei, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China.
| | - Xiao Jiang
- The School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China; Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China.
| |
Collapse
|
26
|
Ferguson KA, Salameh J, Alba C, Selwyn H, Barnes C, Lohani S, Cardin JA. VIP interneurons regulate cortical size tuning and visual perception. Cell Rep 2023; 42:113088. [PMID: 37682710 PMCID: PMC10618959 DOI: 10.1016/j.celrep.2023.113088] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Cortical circuit function is regulated by extensively interconnected, diverse populations of GABAergic interneurons that may play key roles in shaping circuit operation according to behavioral context. A specialized population of interneurons that co-express vasoactive intestinal peptides (VIP-INs) are activated during arousal and innervate other INs and pyramidal neurons (PNs). Although state-dependent modulation of VIP-INs has been extensively studied, their role in regulating sensory processing is less well understood. We examined the impact of VIP-INs in the primary visual cortex of awake behaving mice. Loss of VIP-IN activity alters the behavioral state-dependent modulation of somatostatin-expressing INs (SST-INs) but not PNs. In contrast, reduced VIP-IN activity globally disrupts visual feature selectivity for stimulus size. Moreover, the impact of VIP-INs on perceptual behavior varies with context and is more acute for small than large visual cues. VIP-INs thus contribute to both state-dependent modulation of cortical activity and sensory context-dependent perceptual performance.
Collapse
Affiliation(s)
- Katie A Ferguson
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jenna Salameh
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Christopher Alba
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hannah Selwyn
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Clayton Barnes
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Sweyta Lohani
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Jessica A Cardin
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
27
|
Wyroślak M, Dobrzański G, Mozrzymas JW. Bidirectional plasticity of GABAergic tonic inhibition in hippocampal somatostatin- and parvalbumin-containing interneurons. Front Cell Neurosci 2023; 17:1193383. [PMID: 37448697 PMCID: PMC10336215 DOI: 10.3389/fncel.2023.1193383] [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: 03/24/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
GABAA receptors present in extrasynaptic areas mediate tonic inhibition in hippocampal neurons regulating the performance of neural networks. In this study, we investigated the effect of NMDA-induced plasticity on tonic inhibition in somatostatin- and parvalbumin-containing interneurons. Using pharmacological methods and transgenic mice (SST-Cre/PV-Cre x Ai14), we induced the plasticity of GABAergic transmission in somatostatin- and parvalbumin-containing interneurons by a brief (3 min) application of NMDA. In the whole-cell patch-clamp configuration, we measured tonic currents enhanced by specific agonists (etomidate or gaboxadol). Furthermore, in both the control and NMDA-treated groups, we examined to what extent these changes depend on the regulation of distinct subtypes of GABAA receptors. Tonic conductance in the somatostatin-containing (SST+) interneurons is enhanced after NMDA application, and the observed effect is associated with an increased content of α5-containing GABAARs. Both fast-spiking and non-fast-spiking parvalbumin-positive (PV+) cells showed a reduction of tonic inhibition after plasticity induction. This effect was accompanied in both PV+ interneuron types by a strongly reduced proportion of δ-subunit-containing GABAARs and a relatively small increase in currents mediated by α5-containing GABAARs. Both somatostatin- and parvalbumin-containing interneurons show cell type-dependent and opposite sign plasticity of tonic inhibition. The underlying mechanisms depend on the cell-specific balance of plastic changes in the contents of α5 and δ subunit-containing GABAARs.
Collapse
Affiliation(s)
- Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| | | | - Jerzy W. Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
28
|
Kang J, Lu N, Yang S, Guo B, Zhu Y, Wu S, Huang X, Wong-Riley MTT, Liu YY. Alterations in synapses and mitochondria induced by acute or chronic intermittent hypoxia in the pre-Bötzinger complex of rats: an ultrastructural triple-labeling study with immunocytochemistry and histochemistry. Front Cell Neurosci 2023; 17:1132241. [PMID: 37396926 PMCID: PMC10312010 DOI: 10.3389/fncel.2023.1132241] [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: 12/27/2022] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction The pre-Bötzinger complex (pre-BötC), a kernel of inspiratory rhythmogenesis, is a heterogeneous network with excitatory glutamatergic and inhibitory GABAergic and glycinergic neurons. Inspiratory rhythm generation relies on synchronous activation of glutamatergic neuron, whilst inhibitory neurons play a critical role in shaping the breathing pattern, endowing the rhythm with flexibility in adapting to environmental, metabolic, and behavioral needs. Here we report ultrastructural alterations in excitatory, asymmetric synapses (AS) and inhibitory, symmetric synapses (SS), especially perforated synapses with discontinuous postsynaptic densities (PSDs) in the pre-BötC in rats exposed to daily acute intermittent hypoxia (dAIH) or chronic (C) IH. Methods We utilized for the first time a combination of somatostatin (SST) and neurokinin 1 receptor (NK1R) double immunocytochemistry with cytochrome oxidase histochemistry, to reveal synaptic characteristics and mitochondrial dynamic in the pre-BötC. Results We found perforated synapses with synaptic vesicles accumulated in distinct pools in apposition to each discrete PSD segments. dAIH induced significant increases in the PSD size of macular AS, and the proportion of perforated synapses. AS were predominant in the dAIH group, whereas SS were in a high proportion in the CIH group. dAIH significantly increased SST and NK1R expressions, whereas CIH led to a decrease. Desmosome-like contacts (DLC) were characterized for the first time in the pre-BötC. They were distributed alongside of synapses, especially SS. Mitochondria appeared in more proximity to DLC than synapses, suggestive of a higher energy demand of the DLC. Findings of single spines with dual AS and SS innervation provide morphological evidence of excitation-inhibition interplay within a single spine in the pre-BötC. In particular, we characterized spine-shaft microdomains of concentrated synapses coupled with mitochondrial positioning that could serve as a structural basis for synchrony of spine-shaft communication. Mitochondria were found within spines and ultrastructural features of mitochondrial fusion and fission were depicted for the first time in the pre-BötC. Conclusion We provide ultrastructural evidence of excitation-inhibition synapses in shafts and spines, and DLC in association with synapses that coincide with mitochondrial dynamic in their contribution to respiratory plasticity in the pre-BötC.
Collapse
Affiliation(s)
- Junjun Kang
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Naining Lu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Shoujing Yang
- Department of Pathology, The Fourth Military Medical University, Xi’an, China
| | - Baolin Guo
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Yuanyuan Zhu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Shengxi Wu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| | - Xiaofeng Huang
- Department of Pathology, Xi’an Gaoxin Hospital, Xi’an, China
| | - Margaret T. T. Wong-Riley
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ying-Ying Liu
- Department of Neurobiology, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
29
|
Rindner DJ, Lur G. Practical considerations in an era of multicolor optogenetics. Front Cell Neurosci 2023; 17:1160245. [PMID: 37293628 PMCID: PMC10244638 DOI: 10.3389/fncel.2023.1160245] [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: 02/06/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
The ability to control synaptic communication is indispensable to modern neuroscience. Until recently, only single-pathway manipulations were possible due to limited availability of opsins activated by distinct wavelengths. However, extensive protein engineering and screening efforts have drastically expanded the optogenetic toolkit, ushering in an era of multicolor approaches for studying neural circuits. Nonetheless, opsins with truly discrete spectra are scarce. Experimenters must therefore take care to avoid unintended cross-activation of optogenetic tools (crosstalk). Here, we demonstrate the multidimensional nature of crosstalk in a single model synaptic pathway, testing stimulus wavelength, irradiance, duration, and opsin choice. We then propose a "lookup table" method for maximizing the dynamic range of opsin responses on an experiment-by-experiment basis.
Collapse
Affiliation(s)
| | - Gyorgy Lur
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
30
|
Ferguson KA, Salameh J, Alba C, Selwyn H, Barnes C, Lohani S, Cardin JA. VIP interneurons regulate cortical size tuning and visual perception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532664. [PMID: 37162871 PMCID: PMC10168200 DOI: 10.1101/2023.03.14.532664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Local cortical circuit function is regulated by diverse populations of GABAergic interneurons with distinct properties and extensive interconnectivity. Inhibitory-to-inhibitory interactions between interneuron populations may play key roles in shaping circuit operation according to behavioral context. A specialized population of GABAergic interneurons that co-express vasoactive intestinal peptide (VIP-INs) are activated during arousal and locomotion and innervate other local interneurons and pyramidal neurons. Although modulation of VIP-IN activity by behavioral state has been extensively studied, their role in regulating information processing and selectivity is less well understood. Using a combination of cellular imaging, short and long-term manipulation, and perceptual behavior, we examined the impact of VIP-INs on their synaptic target populations in the primary visual cortex of awake behaving mice. We find that loss of VIP-IN activity alters the behavioral state-dependent modulation of somatostatin-expressing interneurons (SST-INs) but not pyramidal neurons (PNs). In contrast, reduced VIP-IN activity disrupts visual feature selectivity for stimulus size in both populations. Inhibitory-to inhibitory interactions thus directly shape the selectivity of GABAergic interneurons for sensory stimuli. Moreover, the impact of VIP-IN activity on perceptual behavior varies with visual context and is more acute for small than large visual cues. VIP-INs thus contribute to both state-dependent modulation of cortical circuit activity and sensory context-dependent perceptual performance.
Collapse
Affiliation(s)
- Katie A Ferguson
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Jenna Salameh
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Christopher Alba
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Hannah Selwyn
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Clayton Barnes
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Sweyta Lohani
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Jessica A Cardin
- Department of Neuroscience, Kavli Institute for Neuroscience, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06510 USA
| |
Collapse
|
31
|
Brzdąk P, Lebida K, Wyroślak M, Mozrzymas JW. GABAergic synapses onto SST and PV interneurons in the CA1 hippocampal region show cell-specific and integrin-dependent plasticity. Sci Rep 2023; 13:5079. [PMID: 36977728 PMCID: PMC10050003 DOI: 10.1038/s41598-023-31882-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
It is known that GABAergic transmission onto pyramidal neurons shows different forms of plasticity. However, GABAergic cells innervate also other inhibitory interneurons and plasticity phenomena at these projections remain largely unknown. Several mechanisms underlying plastic changes, both at inhibitory and excitatory synapses, show dependence on integrins, key proteins mediating interaction between intra- and extracellular environment. We thus used hippocampal slices to address the impact of integrins on long-term plasticity of GABAergic synapses on specific inhibitory interneurons (containing parvalbumin, PV + or somatostatin, SST +) known to innervate distinct parts of principal cells. Administration of RGD sequence-containing peptide induced inhibitory long-term potentiation (iLTP) at fast-spiking (FS) PV + as well as on SST + interneurons. Interestingly, treatment with a more specific peptide GA(C)RRETAWA(C)GA (RRETAWA), affecting α5β1 integrins, resulted in iLTP in SST + and iLTD in FS PV + interneurons. Brief exposure to NMDA is known to induce iLTP at GABAergic synapses on pyramidal cells. Intriguingly, application of this protocol for considered interneurons evoked iLTP in SST + and iLTD in PV + interneurons. Moreover, we showed that in SST + cells, NMDA-evoked iLTP depends on the incorporation of GABAA receptors containing α5 subunit to the synapses, and this iLTP is occluded by RRETAWA peptide, indicating a key role of α5β1 integrins. Altogether, our results revealed that plasticity of inhibitory synapses at GABAergic cells shows interneuron-specificity and show differences in the underlying integrin-dependent mechanisms. This is the first evidence that neuronal disinhibition may be a highly plastic process depending on interneuron type and integrins' activity.
Collapse
Affiliation(s)
- Patrycja Brzdąk
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland.
| | - Marcin Wyroślak
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-367, Wroclaw, Poland
| |
Collapse
|
32
|
Spike timing-dependent plasticity and memory. Curr Opin Neurobiol 2023; 80:102707. [PMID: 36924615 DOI: 10.1016/j.conb.2023.102707] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 03/16/2023]
Abstract
Spike timing-dependent plasticity (STDP) is a bidirectional form of synaptic plasticity discovered about 30 years ago and based on the relative timing of pre- and post-synaptic spiking activity with a millisecond precision. STDP is thought to be involved in the formation of memory but the millisecond-precision spike-timing required for STDP is difficult to reconcile with the much slower timescales of behavioral learning. This review therefore aims to expose and discuss recent findings about i) the multiple STDP learning rules at both excitatory and inhibitory synapses in vitro, ii) the contribution of STDP-like synaptic plasticity in the formation of memory in vivo and iii) the implementation of STDP rules in artificial neural networks and memristive devices.
Collapse
|
33
|
McFarlan AR, Chou CYC, Watanabe A, Cherepacha N, Haddad M, Owens H, Sjöström PJ. The plasticitome of cortical interneurons. Nat Rev Neurosci 2023; 24:80-97. [PMID: 36585520 DOI: 10.1038/s41583-022-00663-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/31/2022]
Abstract
Hebb postulated that, to store information in the brain, assemblies of excitatory neurons coding for a percept are bound together via associative long-term synaptic plasticity. In this view, it is unclear what role, if any, is carried out by inhibitory interneurons. Indeed, some have argued that inhibitory interneurons are not plastic. Yet numerous recent studies have demonstrated that, similar to excitatory neurons, inhibitory interneurons also undergo long-term plasticity. Here, we discuss the many diverse forms of long-term plasticity that are found at inputs to and outputs from several types of cortical inhibitory interneuron, including their plasticity of intrinsic excitability and their homeostatic plasticity. We explain key plasticity terminology, highlight key interneuron plasticity mechanisms, extract overarching principles and point out implications for healthy brain functionality as well as for neuropathology. We introduce the concept of the plasticitome - the synaptic plasticity counterpart to the genome or the connectome - as well as nomenclature and definitions for dealing with this rich diversity of plasticity. We argue that the great diversity of interneuron plasticity rules is best understood at the circuit level, for example as a way of elucidating how the credit-assignment problem is solved in deep biological neural networks.
Collapse
Affiliation(s)
- Amanda R McFarlan
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Christina Y C Chou
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Airi Watanabe
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Nicole Cherepacha
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Maria Haddad
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Hannah Owens
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.,Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - P Jesper Sjöström
- Centre for Research in Neuroscience, Department of Medicine, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
34
|
Miehl C, Gjorgjieva J. Stability and learning in excitatory synapses by nonlinear inhibitory plasticity. PLoS Comput Biol 2022; 18:e1010682. [PMID: 36459503 PMCID: PMC9718420 DOI: 10.1371/journal.pcbi.1010682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
Synaptic changes are hypothesized to underlie learning and memory formation in the brain. But Hebbian synaptic plasticity of excitatory synapses on its own is unstable, leading to either unlimited growth of synaptic strengths or silencing of neuronal activity without additional homeostatic mechanisms. To control excitatory synaptic strengths, we propose a novel form of synaptic plasticity at inhibitory synapses. Using computational modeling, we suggest two key features of inhibitory plasticity, dominance of inhibition over excitation and a nonlinear dependence on the firing rate of postsynaptic excitatory neurons whereby inhibitory synaptic strengths change with the same sign (potentiate or depress) as excitatory synaptic strengths. We demonstrate that the stable synaptic strengths realized by this novel inhibitory plasticity model affects excitatory/inhibitory weight ratios in agreement with experimental results. Applying a disinhibitory signal can gate plasticity and lead to the generation of receptive fields and strong bidirectional connectivity in a recurrent network. Hence, a novel form of nonlinear inhibitory plasticity can simultaneously stabilize excitatory synaptic strengths and enable learning upon disinhibition.
Collapse
Affiliation(s)
- Christoph Miehl
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- School of Life Sciences, Technical University of Munich, Freising, Germany
- * E-mail: (CM); (JG)
| | - Julijana Gjorgjieva
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- School of Life Sciences, Technical University of Munich, Freising, Germany
- * E-mail: (CM); (JG)
| |
Collapse
|
35
|
Wu YK, Miehl C, Gjorgjieva J. Regulation of circuit organization and function through inhibitory synaptic plasticity. Trends Neurosci 2022; 45:884-898. [PMID: 36404455 DOI: 10.1016/j.tins.2022.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/15/2022]
Abstract
Diverse inhibitory neurons in the mammalian brain shape circuit connectivity and dynamics through mechanisms of synaptic plasticity. Inhibitory plasticity can establish excitation/inhibition (E/I) balance, control neuronal firing, and affect local calcium concentration, hence regulating neuronal activity at the network, single neuron, and dendritic level. Computational models can synthesize multiple experimental results and provide insight into how inhibitory plasticity controls circuit dynamics and sculpts connectivity by identifying phenomenological learning rules amenable to mathematical analysis. We highlight recent studies on the role of inhibitory plasticity in modulating excitatory plasticity, forming structured networks underlying memory formation and recall, and implementing adaptive phenomena and novelty detection. We conclude with experimental and modeling progress on the role of interneuron-specific plasticity in circuit computation and context-dependent learning.
Collapse
Affiliation(s)
- Yue Kris Wu
- School of Life Sciences, Technical University of Munich, Freising, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Christoph Miehl
- School of Life Sciences, Technical University of Munich, Freising, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Julijana Gjorgjieva
- School of Life Sciences, Technical University of Munich, Freising, Germany; Max Planck Institute for Brain Research, Frankfurt, Germany.
| |
Collapse
|
36
|
Sleep and wake cycles dynamically modulate hippocampal inhibitory synaptic plasticity. PLoS Biol 2022; 20:e3001812. [PMID: 36318572 PMCID: PMC9624398 DOI: 10.1371/journal.pbio.3001812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/30/2022] [Indexed: 01/01/2023] Open
Abstract
Sleep is an essential process that consolidates memories by modulating synapses through poorly understood mechanisms. Here, we report that GABAergic synapses in hippocampal CA1 pyramidal neurons undergo daily rhythmic alterations. Specifically, wake inhibits phasic inhibition, whereas it promotes tonic inhibition compared to sleep. We further utilize a model of chemically induced inhibitory long-term potentiation (iLTP) to examine inhibitory plasticity. Intriguingly, while CA1 pyramidal neurons in both wake and sleep mice undergo iLTP, wake mice have a much higher magnitude. We also employ optogenetics and observe that inhibitory inputs from parvalbumin-, but not somatostatin-, expressing interneurons contribute to dynamic iLTP during sleep and wake. Finally, we demonstrate that synaptic insertion of α5-GABAA receptors underlies the wake-specific enhancement of iLTP at parvalbumin-synapses, which is independent of time of the day. These data reveal a previously unappreciated daily oscillation of inhibitory LTP in hippocampal neurons and uncover a dynamic contribution of inhibitory synapses in memory mechanisms across sleep and wake.
Collapse
|
37
|
Wu K, Shepard RD, Castellano D, Han W, Tian Q, Dong L, Lu W. Shisa7 phosphorylation regulates GABAergic transmission and neurodevelopmental behaviors. Neuropsychopharmacology 2022; 47:2160-2170. [PMID: 35534528 PMCID: PMC9556544 DOI: 10.1038/s41386-022-01334-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/27/2022] [Accepted: 04/19/2022] [Indexed: 11/09/2022]
Abstract
GABA-A receptors (GABAARs) are crucial for development and function of the brain. Altered GABAergic transmission is hypothesized to be involved in neurodevelopmental disorders. Recently, we identified Shisa7 as a GABAAR auxiliary subunit that modulates GABAAR trafficking and GABAergic transmission. However, the underlying molecular mechanisms remain elusive. Here we generated a knock-in (KI) mouse line that is phospho-deficient at a phosphorylation site in Shisa7 (S405) and combined with electrophysiology, imaging and behavioral assays to illustrate the role of this site in GABAergic transmission and plasticity as well as behaviors. We found that expression of phospho-deficient mutants diminished α2-GABAAR trafficking in heterologous cells. Additionally, α1/α2/α5-GABAAR surface expression and GABAergic inhibition were decreased in hippocampal neurons in KI mice. Moreover, chemically induced inhibitory long-term potentiation was abolished in KI mice. Lastly, KI mice exhibited hyperactivity, increased grooming and impaired sleep homeostasis. Collectively, our study reveals a phosphorylation site critical for Shisa7-dependent GABAARs trafficking which contributes to behavioral endophenotypes displayed in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Kunwei Wu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ryan David Shepard
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Castellano
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wenyan Han
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qingjun Tian
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
38
|
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: 4.0] [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
|
39
|
Qi C, Luo LD, Feng I, Ma S. Molecular mechanisms of synaptogenesis. Front Synaptic Neurosci 2022; 14:939793. [PMID: 36176941 PMCID: PMC9513053 DOI: 10.3389/fnsyn.2022.939793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Synapses are the basic units for information processing and storage in the nervous system. It is only when the synaptic connection is established, that it becomes meaningful to discuss the structure and function of a circuit. In humans, our unparalleled cognitive abilities are correlated with an increase in the number of synapses. Additionally, genes involved in synaptogenesis are also frequently associated with neurological or psychiatric disorders, suggesting a relationship between synaptogenesis and brain physiology and pathology. Thus, understanding the molecular mechanisms of synaptogenesis is the key to the mystery of circuit assembly and neural computation. Furthermore, it would provide therapeutic insights for the treatment of neurological and psychiatric disorders. Multiple molecular events must be precisely coordinated to generate a synapse. To understand the molecular mechanisms underlying synaptogenesis, we need to know the molecular components of synapses, how these molecular components are held together, and how the molecular networks are refined in response to neural activity to generate new synapses. Thanks to the intensive investigations in this field, our understanding of the process of synaptogenesis has progressed significantly. Here, we will review the molecular mechanisms of synaptogenesis by going over the studies on the identification of molecular components in synapses and their functions in synaptogenesis, how cell adhesion molecules connect these synaptic molecules together, and how neural activity mobilizes these molecules to generate new synapses. Finally, we will summarize the human-specific regulatory mechanisms in synaptogenesis and results from human genetics studies on synaptogenesis and brain disorders.
Collapse
Affiliation(s)
- Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Cai Qi,
| | - Li-Da Luo
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, United States
| | - Irena Feng
- Boston University School of Medicine, Boston, MA, United States
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
40
|
Almeida VN, Radanovic M. Semantic processing and neurobiology in Alzheimer's disease and Mild Cognitive Impairment. Neuropsychologia 2022; 174:108337. [DOI: 10.1016/j.neuropsychologia.2022.108337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/17/2022] [Accepted: 07/17/2022] [Indexed: 11/28/2022]
|
41
|
Wiera G, Brzdąk P, Lech AM, Lebida K, Jabłońska J, Gmerek P, Mozrzymas JW. Integrins Bidirectionally Regulate the Efficacy of Inhibitory Synaptic Transmission and Control GABAergic Plasticity. J Neurosci 2022; 42:5830-5842. [PMID: 35701161 PMCID: PMC9337602 DOI: 10.1523/jneurosci.1458-21.2022] [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: 07/16/2021] [Revised: 05/17/2022] [Accepted: 05/28/2022] [Indexed: 01/29/2023] Open
Abstract
For many decades, synaptic plasticity was believed to be restricted to excitatory transmission. However, in recent years, this view started to change, and now it is recognized that GABAergic synapses show distinct forms of activity-dependent long-term plasticity, but the underlying mechanisms remain obscure. Herein, we asked whether signaling mediated by β1 or β3 subunit-containing integrins might be involved in regulating the efficacy of GABAergic synapses, including the NMDA receptor-dependent inhibitory long-term potentiation (iLTP) in the hippocampus. We found that activation of β3 integrin with fibrinogen induced a stable depression, whereas inhibition of β1 integrin potentiated GABAergic synapses at CA1 pyramidal neurons in male mice. Additionally, compounds that interfere with the interaction of β1 or β3 integrins with extracellular matrix blocked the induction of NMDA-iLTP. In conclusion, we provide the first evidence that integrins are key players in regulating the endogenous modulatory mechanisms of GABAergic inhibition and plasticity in the hippocampus.SIGNIFICANCE STATEMENT Epilepsy, schizophrenia, and anxiety are just a few medical conditions associated with dysfunctional inhibitory synaptic transmission. GABAergic synapses are known for their extraordinary susceptibility to modulation by endogenous factors and exogenous pharmacological agents. We describe here that integrins, adhesion proteins, play a key role in the modulation of inhibitory synaptic transmission. Specifically, we show that interference with integrin-dependent adhesion results in a variety of effects on the amplitude and frequency of GABAergic mIPSCs. Activation of β3 subunit-containing integrins induces inhibitory long-term depression, whereas the inhibition of β1 subunit-containing integrins induces iLTP. Our results unveil an important mechanism controlling synaptic inhibition, which opens new avenues into the usage of integrin-aimed pharmaceuticals as modulators of GABAergic synapses.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Patrycja Brzdąk
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Anna Maria Lech
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Przemysław Gmerek
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
42
|
Xue X, Pan J, Zhang H, Lu Y, Mao Q, Ma K. Baihe Dihuang (Lilium Henryi Baker and Rehmannia Glutinosa) decoction attenuates somatostatin interneurons deficits in prefrontal cortex of depression via miRNA-144-3p mediated GABA synthesis and release. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115218. [PMID: 35337919 DOI: 10.1016/j.jep.2022.115218] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/19/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baihe Dihuang Decoction is a well-known traditional Chinese medicine prescription (Also known as Lilium Henryi Baker and Rehmannia Glutinosa Decoction, LBRD) composed of Lilium Henryi Baker bulb and raw juice from Rehmannia Glutinosa (Gaertn) DC with the curative efficacy of nourishing yin and clearing heat based on the Chinese herbal medicine theory. It has been used as routine medication in treating depression combined with conventional western medicine in China for years. AIM OF THE STUDY LBRD can attenuates GABAergic deficits in the medial prefrontal cortex (mPFC) of depression. This study aimed to investigate the mechanism of antidepressive properties of LBRD in the prefrontal GABAergic interneuron subtypes, including parvalbumin (PV), somatostatin (SST), vasoactive intestinal peptide (VIP)-positive neuron. MATERIALS AND METHODS In this project, chronic unpredicted mild stress paradigm was adopted to construct depression model. After treated with LBRD standard decoction and behaviors test, the level of GABA associated miRNA/mRNA and GABAergic subtype-specific markers were detected by qRT-PCR and Western blot. The lncRNAs/miRNAs/GABA regulatory axis was verified by luciferase reporter assay, RNA immunoprecipitation, RNA pull-down assay, and theses changes were measured in LBRD administration with the use of immunofluorescence staining and RNA-fluorescence in situ hybridization. RESULTS In the current study, we found that LBRD exhibited high efficacy based on the results of behavioral tests. Meanwhile, LBRD also improved the reduced GABA levels in depression by increasing the expression of lncRNA Neat1 and Malat1, as well as decreasing miRNA-144-3p and miRNA-15b-5p. Moreover, the level of Sst mRNA and protein that were harvested from the mPFC tissues of depression group was significantly lower than those in the control mice. While, these changes can be reverted by LBRD standard decoction administration. Whereas, neither chronic stress nor treatment can change the level of PV and VIP mRNAs and protein expression. In the SST-positive neuron of mPFC tissues, treatment with LBRD standard decoction resulted in the elevation of Gad-67, VGAT, GAT-3 and a reduction of miRNA-144-3p expression. CONCLUSIONS These findings suggested that LBRD antidepressant activities may be related to ameliorating the SST-positive neuron deficits via regulating the miRNA-144-3p mediated GABA synthesis and release.
Collapse
Affiliation(s)
- Xiaoyan Xue
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Jin Pan
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Hongxiu Zhang
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China; Institute of Virology, Jinan Municipal Center for Disease Control and Prevention, Jinan, 250021, PR China
| | - Yanting Lu
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Qiancheng Mao
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China
| | - Ke Ma
- Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan, 250355, PR China.
| |
Collapse
|
43
|
Tipton AE, Russek SJ. Regulation of Inhibitory Signaling at the Receptor and Cellular Level; Advances in Our Understanding of GABAergic Neurotransmission and the Mechanisms by Which It Is Disrupted in Epilepsy. Front Synaptic Neurosci 2022; 14:914374. [PMID: 35874848 PMCID: PMC9302637 DOI: 10.3389/fnsyn.2022.914374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Inhibitory signaling in the brain organizes the neural circuits that orchestrate how living creatures interact with the world around them and how they build representations of objects and ideas. Without tight control at multiple points of cellular engagement, the brain’s inhibitory systems would run down and the ability to extract meaningful information from excitatory events would be lost leaving behind a system vulnerable to seizures and to cognitive decline. In this review, we will cover many of the salient features that have emerged regarding the dynamic regulation of inhibitory signaling seen through the lens of cell biology with an emphasis on the major building blocks, the ligand-gated ion channel receptors that are the first transduction point when the neurotransmitter GABA is released into the synapse. Epilepsy association will be used to indicate importance of key proteins and their pathways to brain function and to introduce novel areas for therapeutic intervention.
Collapse
Affiliation(s)
- Allison E. Tipton
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
| | - Shelley J. Russek
- Biomolecular Pharmacology Program, Boston University School of Medicine, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Boston University MD/PhD Training Program, Boston, MA, United States
- *Correspondence: Shelley J. Russek,
| |
Collapse
|
44
|
Simonova NA, Volgushev MA, Malyshev AY. Enhanced Non-Associative Long-Term Potentiation in Immature Granule Cells in the Dentate Gyrus of Adult Rats. Front Synaptic Neurosci 2022; 14:889947. [PMID: 35711669 PMCID: PMC9192440 DOI: 10.3389/fnsyn.2022.889947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/15/2022] [Indexed: 11/20/2022] Open
Abstract
The dentate gyrus is one of the few sites of neurogenesis in the adult brain. Integration of new-generated granule cells into the hippocampal circuitry provides a substrate for structural plasticity, fundamental for normal function of adult hippocampus. However, mechanisms of synaptic plasticity that mediate integration of new-generated granule cells into the existing circuitry remain poorly understood. Especially mechanisms of plasticity at GABA-ergic synapses remain elusive. Here, we show that postsynaptic spiking without presynaptic activation can induce heterosynaptic, non-associative plasticity at GABA-ergic inputs to both immature and mature granule cells. In both immature and mature neurons, plastic changes were bidirectional and individual inputs could express long-term potentiation (LTP) or long-term depression (LTD), or do not change. However, properties of non-associative plasticity dramatically change with maturation of newly generated granule cells: while in immature cells there was a clear predominance of non-associative LTP and net potentiation across the inputs, in mature neurons, potentiation and depression were balanced with no net change on average. We conclude that GABA-ergic inputs to granule cells are plastic, and that the rules for induction of non-associative plasticity change with maturation. We propose that potentiation-biased non-associative plasticity of GABA-ergic transmission might help to counter-balance an increase of excitatory drive that is facilitated by enhanced LTP at glutamatergic synapses in maturating granule cells. Such mechanism might help to build a strong GABA-ergic input to surviving active new cells, necessary for normal function of mature granule cells, which operate under a tight inhibitory control and generate sparse spiking activity.
Collapse
Affiliation(s)
- Natalia A. Simonova
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Maxim A. Volgushev
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, United States
- *Correspondence: Maxim A. Volgushev
| | - Alexey Y. Malyshev
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
45
|
Hauser D, Behr K, Konno K, Schreiner D, Schmidt A, Watanabe M, Bischofberger J, Scheiffele P. Targeted proteoform mapping uncovers specific Neurexin-3 variants required for dendritic inhibition. Neuron 2022; 110:2094-2109.e10. [PMID: 35550065 PMCID: PMC9275415 DOI: 10.1016/j.neuron.2022.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 02/05/2022] [Accepted: 04/15/2022] [Indexed: 12/21/2022]
Abstract
The diversification of cell adhesion molecules by alternative splicing is proposed to underlie molecular codes for neuronal wiring. Transcriptomic approaches mapped detailed cell-type-specific mRNA splicing programs. However, it has been hard to probe the synapse-specific localization and function of the resulting protein splice isoforms, or “proteoforms,” in vivo. We here apply a proteoform-centric workflow in mice to test the synapse-specific functions of the splice isoforms of the synaptic adhesion molecule Neurexin-3 (NRXN3). We uncover a major proteoform, NRXN3 AS5, that is highly expressed in GABAergic interneurons and at dendrite-targeting GABAergic terminals. NRXN3 AS5 abundance significantly diverges from Nrxn3 mRNA distribution and is gated by translation-repressive elements. Nrxn3 AS5 isoform deletion results in a selective impairment of dendrite-targeting interneuron synapses in the dentate gyrus without affecting somatic inhibition or glutamatergic perforant-path synapses. This work establishes cell- and synapse-specific functions of a specific neurexin proteoform and highlights the importance of alternative splicing regulation for synapse specification. Translational regulation guides alternative Neurexin proteoform expression NRXN3 AS5 proteoforms are concentrated at dendrite-targeting interneuron synapses A proteome-centric workflow uncovers NRXN3 AS5 interactors in vivo Loss of NRXN3 AS5 leads to selective impairments in dendritic inhibition
Collapse
Affiliation(s)
- David Hauser
- Biozentrum of the University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Katharina Behr
- Department of Biomedicine, University of Basel, Pestalozzistrasse 20, 4056 Basel, Switzerland
| | - Kohtarou Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Dietmar Schreiner
- Biozentrum of the University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Alexander Schmidt
- Biozentrum of the University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Josef Bischofberger
- Department of Biomedicine, University of Basel, Pestalozzistrasse 20, 4056 Basel, Switzerland
| | - Peter Scheiffele
- Biozentrum of the University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
46
|
Johnson C, Kretsge LN, Yen WW, Sriram B, O'Connor A, Liu RS, Jimenez JC, Phadke RA, Wingfield KK, Yeung C, Jinadasa TJ, Nguyen TPH, Cho ES, Fuchs E, Spevack ED, Velasco BE, Hausmann FS, Fournier LA, Brack A, Melzer S, Cruz-Martín A. Highly unstable heterogeneous representations in VIP interneurons of the anterior cingulate cortex. Mol Psychiatry 2022; 27:2602-2618. [PMID: 35246635 PMCID: PMC11128891 DOI: 10.1038/s41380-022-01485-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/09/2022]
Abstract
A hallmark of the anterior cingulate cortex (ACC) is its functional heterogeneity. Functional and imaging studies revealed its importance in the encoding of anxiety-related and social stimuli, but it is unknown how microcircuits within the ACC encode these distinct stimuli. One type of inhibitory interneuron, which is positive for vasoactive intestinal peptide (VIP), is known to modulate the activity of pyramidal cells in local microcircuits, but it is unknown whether VIP cells in the ACC (VIPACC) are engaged by particular contexts or stimuli. Additionally, recent studies demonstrated that neuronal representations in other cortical areas can change over time at the level of the individual neuron. However, it is not known whether stimulus representations in the ACC remain stable over time. Using in vivo Ca2+ imaging and miniscopes in freely behaving mice to monitor neuronal activity with cellular resolution, we identified individual VIPACC that preferentially activated to distinct stimuli across diverse tasks. Importantly, although the population-level activity of the VIPACC remained stable across trials, the stimulus-selectivity of individual interneurons changed rapidly. These findings demonstrate marked functional heterogeneity and instability within interneuron populations in the ACC. This work contributes to our understanding of how the cortex encodes information across diverse contexts and provides insight into the complexity of neural processes involved in anxiety and social behavior.
Collapse
Affiliation(s)
- Connor Johnson
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Lisa N Kretsge
- The Graduate Program for Neuroscience, Boston University, Boston, MA, USA
- Neurophotonics Center, Boston University, Boston, MA, USA
| | - William W Yen
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | | | - Alexandra O'Connor
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Ruichen Sky Liu
- MS in Statistical Practice Program, Boston University, Boston, MA, USA
| | - Jessica C Jimenez
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Rhushikesh A Phadke
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Kelly K Wingfield
- Neurophotonics Center, Boston University, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, USA
| | - Charlotte Yeung
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Tushare J Jinadasa
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Thanh P H Nguyen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Eun Seon Cho
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Erelle Fuchs
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Eli D Spevack
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Berta Escude Velasco
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Frances S Hausmann
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Luke A Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston, MA, USA
| | - Sarah Melzer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA.
- Neurophotonics Center, Boston University, Boston, MA, USA.
- Molecular Biology, Cell Biology and Biochemistry Program, Boston University, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- The Center for Network Systems Biology, Boston University, Boston, MA, USA.
| |
Collapse
|
47
|
Asgarihafshejani A, Honoré È, Michon FX, Laplante I, Lacaille JC. Long-term potentiation at pyramidal cell to somatostatin interneuron synapses controls hippocampal network plasticity and memory. iScience 2022; 25:104259. [PMID: 35521524 PMCID: PMC9062215 DOI: 10.1016/j.isci.2022.104259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/11/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
Abstract
Hippocampal somatostatin (SOM) cells are dendrite-projecting inhibitory interneurons. CA1 SOM cells receive major excitatory inputs from pyramidal cells (PC-SOM synapses) which show mGluR1a- and mTORC1-mediated long-term potentiation (LTP). PC-SOM synapse LTP contributes to CA1 network metaplasticity and memory consolidation, but whether it is sufficient to regulate these processes remains unknown. Here we used optogenetic stimulation of CA1 pyramidal cells and whole-cell recordings in slices to show that optogenetic theta-burst stimulation (TBSopto) produces LTP at PC-SOM synapses. At the network level, we found that TBSopto differentially regulates metaplasticity of pyramidal cell inputs: enhancing LTP at Schaffer collateral synapses and depressing LTP at temporo-ammonic synapses. At the behavioral level, we uncovered that in vivo TBSopto regulates learning-induced LTP at PC-SOM synapses, as well as contextual fear memory. Thus, LTP of PC-SOM synapses is a long-term feedback mechanism controlling pyramidal cell synaptic plasticity, sufficient to regulate memory consolidation. Optogenetic theta-burst (TBSopto) induces LTP at PC-SOM synapses TBSopto differentially regulates metaplasticity of pyramidal cell inputs In vivo TBSopto regulates PC-SOM plasticity and contextual fear memory PC-SOM synapse LTP grants durable feedback control of network plasticity and memory
Collapse
Affiliation(s)
- Azam Asgarihafshejani
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Ève Honoré
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - François-Xavier Michon
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Isabel Laplante
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Jean-Claude Lacaille
- Department of Neurosciences, Center for Interdisciplinary Research on Brain and Learning (CIRCA) and Research Group on Neural Signaling and Circuitry (GRSNC), Université de Montréal, Montreal, Quebec H3C 3J7, Canada
- Corresponding author
| |
Collapse
|
48
|
Widmer FC, O'Toole SM, Keller GB. NMDA receptors in visual cortex are necessary for normal visuomotor integration and skill learning. eLife 2022; 11:71476. [PMID: 35170429 PMCID: PMC8901170 DOI: 10.7554/elife.71476] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 02/16/2022] [Indexed: 11/24/2022] Open
Abstract
The experience of coupling between motor output and visual feedback is necessary for the development of visuomotor skills and shapes visuomotor integration in visual cortex. Whether these experience-dependent changes of responses in V1 depend on modifications of the local circuit or are the consequence of circuit changes outside of V1 remains unclear. Here, we probed the role of N-methyl-d-aspartate (NMDA) receptor-dependent signaling, which is known to be involved in neuronal plasticity, in mouse primary visual cortex (V1) during visuomotor development. We used a local knockout of NMDA receptors and a photoactivatable inhibition of CaMKII in V1 during the first visual experience to probe for changes in neuronal activity in V1 as well as the influence on performance in a visuomotor task. We found that a knockout of NMDA receptors before, but not after, first visuomotor experience reduced responses to unpredictable stimuli, diminished the suppression of predictable feedback in V1, and impaired visuomotor skill learning later in life. Our results demonstrate that NMDA receptor-dependent signaling in V1 is critical during the first visuomotor experience for shaping visuomotor integration and enabling visuomotor skill learning.
Collapse
Affiliation(s)
- Felix C Widmer
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Sean M O'Toole
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Georg B Keller
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
49
|
Ravasenga T, Ruben M, Regio V, Polenghi A, Petrini EM, Barberis A. Spatial regulation of coordinated excitatory and inhibitory synaptic plasticity at dendritic synapses. Cell Rep 2022; 38:110347. [PMID: 35139381 PMCID: PMC8844559 DOI: 10.1016/j.celrep.2022.110347] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 09/16/2021] [Accepted: 01/14/2022] [Indexed: 12/02/2022] Open
Abstract
The induction of synaptic plasticity at an individual dendritic glutamatergic spine can affect neighboring spines. This local modulation generates dendritic plasticity microdomains believed to expand the neuronal computational capacity. Here, we investigate whether local modulation of plasticity can also occur between glutamatergic synapses and adjacent GABAergic synapses. We find that the induction of long-term potentiation at an individual glutamatergic spine causes the depression of nearby GABAergic inhibitory synapses (within 3 μm), whereas more distant ones are potentiated. Notably, L-type calcium channels and calpain are required for this plasticity spreading. Overall, our data support a model whereby input-specific glutamatergic postsynaptic potentiation induces a spatially regulated rearrangement of inhibitory synaptic strength in the surrounding area through short-range heterosynaptic interactions. Such local coordination of excitatory and inhibitory synaptic plasticity is expected to influence dendritic information processing and integration. LTP of individual dendritic spines causes iLTD at neighboring GABAergic synapses Interaction between single-spine LTP and iLTD occurs in the spatial range of ±3 μm This iLTD depends on the local dendritic calcium increase and calpain activation iLTD is associated with reduced gephyrin clustering and increased GABAAR mobility
Collapse
Affiliation(s)
- Tiziana Ravasenga
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Massimo Ruben
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Vincenzo Regio
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Alice Polenghi
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Enrica Maria Petrini
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Andrea Barberis
- Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
50
|
Gao WJ, Yang SS, Mack NR, Chamberlin LA. Aberrant maturation and connectivity of prefrontal cortex in schizophrenia-contribution of NMDA receptor development and hypofunction. Mol Psychiatry 2022; 27:731-743. [PMID: 34163013 PMCID: PMC8695640 DOI: 10.1038/s41380-021-01196-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The neurobiology of schizophrenia involves multiple facets of pathophysiology, ranging from its genetic basis over changes in neurochemistry and neurophysiology, to the systemic level of neural circuits. Although the precise mechanisms associated with the neuropathophysiology remain elusive, one essential aspect is the aberrant maturation and connectivity of the prefrontal cortex that leads to complex symptoms in various stages of the disease. Here, we focus on how early developmental dysfunction, especially N-methyl-D-aspartate receptor (NMDAR) development and hypofunction, may lead to the dysfunction of both local circuitry within the prefrontal cortex and its long-range connectivity. More specifically, we will focus on an "all roads lead to Rome" hypothesis, i.e., how NMDAR hypofunction during development acts as a convergence point and leads to local gamma-aminobutyric acid (GABA) deficits and input-output dysconnectivity in the prefrontal cortex, which eventually induce cognitive and social deficits. Many outstanding questions and hypothetical mechanisms are listed for future investigations of this intriguing hypothesis that may lead to a better understanding of the aberrant maturation and connectivity associated with the prefrontal cortex.
Collapse
Affiliation(s)
- Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| | - Sha-Sha Yang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Nancy R Mack
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Linda A Chamberlin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| |
Collapse
|