1
|
Ghazvineh S, Mooziri M, Salimi A, Mirnajafi-Zadeh J, Raoufy MR. Olfactory epithelium electrical stimulation mitigates memory and synaptic deficits caused by mechanical ventilation. Sci Rep 2025; 15:12197. [PMID: 40204831 PMCID: PMC11982190 DOI: 10.1038/s41598-025-96661-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Mechanical ventilation (MV) causes a wide range of cognitive impairments. Unfortunately, to date, we are lacking knowledge about its underlying neural mechanisms and significant treatment options for the condition. Here, we show that MV-induced memory impairment in rats stems from dysfunctions in the olfactory bulb-medial prefrontal cortex-ventral hippocampus network and hippocampal synaptic currents imbalance. More importantly, we introduce a novel treatment approach, namely olfactory epithelium electrical stimulation (OEES) that shows promising preclinical results in mitigating the mentioned behavioral and neural disorders caused by MV. These results pave the way for research on non-invasive brain stimulation approaches and introduce the olfactory system as a potential target for treating cognitive or psychiatric disorders induced by MV.
Collapse
Affiliation(s)
- Sepideh Ghazvineh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Morteza Mooziri
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alireza Salimi
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for Brain and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Hazra JD, Shrivastava K, Wüstner LS, Anunu R, Chervinsky E, Hazra S, Beuter S, Kriebel M, Maroun M, Volkmer H, Richter-Levin G. Effects of TrkB-related induced metaplasticity within the BLA on anxiety, extinction learning, and plasticity in BLA-modulated brain regions. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:4. [PMID: 40033342 PMCID: PMC11874401 DOI: 10.1186/s12993-025-00267-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Neuronal plasticity within the basolateral amygdala (BLA) is fundamental for fear learning. Metaplasticity, the regulation of plasticity states, has emerged as a key mechanism mediating the subsequent impact of emotional and stressful experiences. After mRNA knockdown of synaptic plasticity-related TrkB, we examined the impact of chronically altered activity in the rat BLA (induced metaplasticity) on anxiety-like behavior, fear memory-related behaviors, and neural plasticity in brain regions modulated by the BLA. These effects were investigated under both basal conditions and following exposure to acute trauma (UWT). RESULTS Under basal conditions, TrkB knockdown increased anxiety-like behavior and impaired extinction learning. TrkBKD also reduced LTP in the vSub-mPFC pathway but not in the dentate gyrus. Compared with those of control animals, acute trauma exposure led to increased anxiety-like behavior and impaired extinction learning in both the trauma-exposed group (CTR-UWT) and the trauma-exposed group on the background of TrkB knockdown (TrkBKD-UWT). However, the deficit in extinction learning was more pronounced in the TrkBKD-UWT group than in the CTR-UWT group. Accordingly, TrkBKD-UWT, but not CTR-UWT, resulted in impaired LTP in the vSub- mPFC pathway. Since LTP in this pathway is independent of BLA involvement, this result suggests that lasting intra-BLA-induced metaplasticity may also lead to transregional metaplasticity within the mPFC, as suggested previously. CONCLUSIONS Taken together, these findings reveal the dissociative involvement of BLA function, on the one hand, in anxiety, which is affected by the knockdown of TrkB, and, on the other hand, in extinction learning, which is more significantly affected by the combination of intra-BLA-induced metaplasticity and exposure to emotional trauma.
Collapse
Affiliation(s)
- Joyeeta Dutta Hazra
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Kuldeep Shrivastava
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Lisa-Sophie Wüstner
- Department Molecular-Neurobiology, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Rachel Anunu
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Erez Chervinsky
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Somoday Hazra
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Simone Beuter
- Department Molecular-Neurobiology, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Martin Kriebel
- Department Molecular-Neurobiology, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Mouna Maroun
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel
| | - Hansjuergen Volkmer
- Department Molecular-Neurobiology, Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstr. 55, 72770, Reutlingen, Germany
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, 3498838, Haifa, Israel.
- Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, Israel.
- Psychology Department, University of Haifa, 3498838, Haifa, Israel.
| |
Collapse
|
3
|
Lozano-Ortiz K, Felix-Ortiz AC, Terrell JM, Gonzalez C, Whitehorn KR, Kanke AE, Villalon SA, Ramos AR, Miller AN, Rodriguez-Romaguera J, Burgos-Robles A. The prelimbic prefrontal cortex mediates the development of lasting social avoidance as a consequence of social threat conditioning. Neuropsychopharmacology 2025:10.1038/s41386-025-02073-8. [PMID: 40016364 DOI: 10.1038/s41386-025-02073-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 01/21/2025] [Accepted: 02/11/2025] [Indexed: 03/01/2025]
Abstract
Social avoidance is highly detrimental for natural behavior. Despite much research on this topic, the mechanisms underlying the development of social avoidance as a consequence of social-related traumatic experiences remain highly elusive. To investigate this issue, we adapted a mouse model of social threat conditioning in which mice received shock punishment during exploration of an unfamiliar conspecific. This resulted in prominent and lasting reductions in social behavior, effects that were not observed in mice that received shock punishment in the absence of a social stimulus. Furthermore, the effects of social threat conditioning were independent of contextual settings, sex variables, and particular identity of the unfamiliar conspecifics that predicted shock punishment. Shedding new light into the neurobiological bases for this phenomenon, we found that optogenetic silencing of the prelimbic (PL), but not infralimbic (IL), prefrontal cortex during social threat conditioning produced profound forgetting and restoration of social behavior during subsequent sociability tests. Significant forgetting and recovery of social behavior was also observed with prelimbic inhibition of NMDARs. Collectively, these findings are consistent with the notion that social-related trauma is a prominent risk factor for social avoidance, and that traumatic experiences that involve social elements engage learning-related mechanisms in corticolimbic networks to promote long-term representations of social threat.
Collapse
Affiliation(s)
- Kelly Lozano-Ortiz
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Ada C Felix-Ortiz
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
- Department of Cellular and Integrative Physiology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Jaelyn M Terrell
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Carolina Gonzalez
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Kamryn R Whitehorn
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Addison E Kanke
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Stephanie A Villalon
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Angelica R Ramos
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Ashley N Miller
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jose Rodriguez-Romaguera
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Institute of Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA.
| | - Anthony Burgos-Robles
- Department of Neuroscience, Developmental, and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA.
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
4
|
Dentel B, Angeles-Perez L, Flores AY, Lei K, Ren C, Sanchez AP, Tsai PT. Neuronal cell type specific roles for Nprl2 in neurodevelopmental disorder-relevant behaviors. Neurobiol Dis 2025; 205:106790. [PMID: 39765274 DOI: 10.1016/j.nbd.2025.106790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Loss of function in the subunits of the GTPase-activating protein (GAP) activity toward Rags-1 (GATOR1) complex, an amino-acid sensitive negative regulator of the mechanistic target of rapamycin complex 1 (mTORC1), is implicated in both genetic familial epilepsies and Neurodevelopmental Disorders (NDDs) (Baldassari et al., 2018). Previous studies have found seizure phenotypes and increased activity resulting from conditional deletion of GATOR1 function from forebrain excitatory neurons (Yuskaitis et al., 2018; Dentel et al., 2022); however, studies focused on understanding mechanisms contributing to NDD-relevant behaviors are lacking, especially studies understanding the contributions of GATOR1's critical GAP catalytic subunit, nitrogen permease regulator like-2 (Nprl2). Given the clinical phenotypes observed in patients with Nprl2 mutations, in this study, we sought to investigate the neuronal cell type contributions of Nprl2 to NDD behaviors. We conditionally deleted Nprl2 broadly in most neurons (Synapsin1cre), in inhibitory neurons only (Vgatcre), and in Purkinje cells within the cerebellum (L7cre). Broad neuronal deletion of Nprl2 resulted in seizures, social and learning deficits, and hyperactivity. In contrast, deleting Nprl2 from inhibitory neurons led to increased motor learning, hyperactive behavior, in addition to social and learning deficits. Lastly, Purkinje cell (PC) loss of Nprl2 also led to learning and social deficits but did not affect locomotor activity. These phenotypes enhance understanding of the spectrum of disease found in human populations with GATOR1 loss of function and highlight the significance of distinct cellular populations to NDD-related behaviors. SIGNIFICANCE STATEMENT: We aim to elucidate the neuronal-specific contributions of nitrogen permease regulator like-2 (Nprl2) to its neurodevelopmental disorder (NDD)-relevant phenotypes. We conditionally deleted Nprl2 broadly in neurons (Syn1cre), in inhibitory neurons (Vgatcre), and in cerebellar Purkinje cells (L7cre). We identify seizures only in the Syn1cre conditional mutant (cKO); hyperactivity, learning difficulties, social deficits, and impulsivity in the Syn1cre and Vgatcre cKOs; and social deficits, and fear learning deficits in L7cre cKOs. To our knowledge, we are the first to describe the behavioral contributions of Nprl2's function across multiple cell types. Our findings highlight both critical roles for Nprl2 in learning and behavior and also distinct contributions of select neuronal populations to these NDD-relevant behaviors.
Collapse
Affiliation(s)
- Brianne Dentel
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Lidiette Angeles-Perez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Abigail Y Flores
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Katherine Lei
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Chongyu Ren
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Andrea Pineda Sanchez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Peter T Tsai
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Psychiatry, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Pediatrics, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Neuroscience; O'Donnell Brain Institute, Dallas, TX, United States of America.
| |
Collapse
|
5
|
Zerlaut Y, Tzilivaki A. Interneuronal modulations as a functional switch for cortical computations: mechanisms and implication for disease. Front Cell Neurosci 2025; 18:1479579. [PMID: 39916937 PMCID: PMC11799556 DOI: 10.3389/fncel.2024.1479579] [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: 08/12/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025] Open
Abstract
Understanding cortical inhibition and its diverse roles remains a key challenge in neurophysiological research. Traditionally, inhibition has been recognized for controlling the stability and rhythmicity of network dynamics, or refining the spatiotemporal properties of cortical representations. In this perspective, we propose that specific types of interneurons may play a complementary role, by modulating the computational properties of neural networks. We review experimental and theoretical evidence, mainly from rodent sensory cortices, that supports this view. Additionally, we explore how dysfunctions in these interneurons may disrupt the network's ability to switch between computational modes, impacting the flexibility of cortical processing and potentially contributing to various neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Yann Zerlaut
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, Berlin, Germany
- Einstein Center for Neurosciences, Chariteplatz, Berlin, Germany
- NeuroCure Cluster of Excellence, Chariteplatz, Berlin, Germany
| |
Collapse
|
6
|
Hu YB, Deng X, Liu L, Cao CC, Su YW, Gao ZJ, Cheng X, Kong D, Li Q, Shi YW, Wang XG, Ye X, Zhao H. Distinct roles of excitatory and inhibitory neurons in the medial prefrontal cortex in the expression and reconsolidation of methamphetamine-associated memory in male mice. Neuropsychopharmacology 2024; 49:1827-1838. [PMID: 38730034 PMCID: PMC11473735 DOI: 10.1038/s41386-024-01879-2] [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: 12/06/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
Methamphetamine, a commonly abused drug, is known for its high relapse rate. The persistence of addictive memories associated with methamphetamine poses a significant challenge in preventing relapse. Memory retrieval and subsequent reconsolidation provide an opportunity to disrupt addictive memories. However, the key node in the brain network involved in methamphetamine-associated memory retrieval has not been clearly defined. In this study, using the conditioned place preference in male mice, whole brain c-FOS mapping and functional connectivity analysis, together with chemogenetic manipulations of neural circuits, we identified the medial prefrontal cortex (mPFC) as a critical hub that integrates inputs from the retrosplenial cortex and the ventral tegmental area to support both the expression and reconsolidation of methamphetamine-associated memory during its retrieval. Surprisingly, with further cell-type specific analysis and manipulation, we also observed that methamphetamine-associated memory retrieval activated inhibitory neurons in the mPFC to facilitate memory reconsolidation, while suppressing excitatory neurons to aid memory expression. These findings provide novel insights into the neural circuits and cellular mechanisms involved in the retrieval process of addictive memories. They suggest that targeting the balance between excitation and inhibition in the mPFC during memory retrieval could be a promising treatment strategy to prevent relapse in methamphetamine addiction.
Collapse
Affiliation(s)
- Yu-Bo Hu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xi Deng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Lu Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Can-Can Cao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ya-Wen Su
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhen-Jie Gao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Deshan Kong
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Qi Li
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| | - Hu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
7
|
Báldi R, Muthuswamy S, Loomba N, Patel S. Synaptic Organization-Function Relationships of Amygdala Interneurons Supporting Associative Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599631. [PMID: 38948865 PMCID: PMC11212985 DOI: 10.1101/2024.06.18.599631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Coordinated activity of basolateral amygdala (BLA) GABAergic interneurons (INs) and glutamatergic principal cells (PCs) is critical for associative learning, however the microcircuit organization-function relationships of distinct IN classes remain uncertain. Here, we show somatostatin (SOM) INs provide inhibition onto, and are excited by, local PCs, whereas vasoactive intestinal peptide (VIP) INs are driven by extrinsic afferents. Parvalbumin (PV) INs inhibit PCs and are activated by local and extrinsic inputs. Thus, SOM and VIP INs exhibit complementary roles in feedback and feedforward inhibition, respectively, while PV INs contribute to both microcircuit motifs. Functionally, each IN subtype reveals unique activity patterns across fear- and extinction learning with SOM and VIP INs showing most divergent characteristics, and PV INs display an intermediate phenotype parallelling synaptic data. Finally, SOM and PV INs dynamically track behavioral state transitions across learning. These data provide insight into the synaptic microcircuit organization-function relationships of distinct BLA IN classes.
Collapse
|
8
|
Duggins P, Eliasmith C. A scalable spiking amygdala model that explains fear conditioning, extinction, renewal and generalization. Eur J Neurosci 2024; 59:3093-3116. [PMID: 38616566 DOI: 10.1111/ejn.16338] [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: 09/07/2023] [Revised: 02/03/2024] [Accepted: 03/11/2024] [Indexed: 04/16/2024]
Abstract
The amygdala (AMY) is widely implicated in fear learning and fear behaviour, but it remains unclear how the many biological components present within AMY interact to achieve these abilities. Building on previous work, we hypothesize that individual AMY nuclei represent different quantities and that fear conditioning arises from error-driven learning on the synapses between AMY nuclei. We present a computational model of AMY that (a) recreates the divisions and connections between AMY nuclei and their constituent pyramidal and inhibitory neurons; (b) accommodates scalable high-dimensional representations of external stimuli; (c) learns to associate complex stimuli with the presence (or absence) of an aversive stimulus; (d) preserves feature information when mapping inputs to salience estimates, such that these estimates generalize to similar stimuli; and (e) induces a diverse profile of neural responses within each nucleus. Our model predicts (1) defensive responses and neural activities in several experimental conditions, (2) the consequence of artificially ablating particular nuclei and (3) the tendency to generalize defensive responses to novel stimuli. We test these predictions by comparing model outputs to neural and behavioural data from animals and humans. Despite the relative simplicity of our model, we find significant overlap between simulated and empirical data, which supports our claim that the model captures many of the neural mechanisms that support fear conditioning. We conclude by comparing our model to other computational models and by characterizing the theoretical relationship between pattern separation and fear generalization in healthy versus anxious individuals.
Collapse
Affiliation(s)
- Peter Duggins
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Chris Eliasmith
- Centre for Theoretical Neuroscience, University of Waterloo, Waterloo, Ontario, Canada
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
- Department of Philosophy, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
9
|
Mota B, Brás AR, Araújo-Andrade L, Silva A, Pereira PA, Madeira MD, Cardoso A. High-Caloric Diets in Adolescence Impair Specific GABAergic Subpopulations, Neurogenesis, and Alter Astrocyte Morphology. Int J Mol Sci 2024; 25:5524. [PMID: 38791562 PMCID: PMC11122083 DOI: 10.3390/ijms25105524] [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: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
We compared the effects of two different high-caloric diets administered to 4-week-old rats for 12 weeks: a diet rich in sugar (30% sucrose) and a cafeteria diet rich in sugar and high-fat foods. We focused on the hippocampus, particularly on the gamma-aminobutyric acid (GABA)ergic system, including the Ca2+-binding proteins parvalbumin (PV), calretinin (CR), calbindin (CB), and the neuropeptides somatostatin (SST) and neuropeptide Y (NPY). We also analyzed the density of cholinergic varicosities, brain-derived neurotrophic factor (BDNF), reelin (RELN), and cyclin-dependent kinase-5 (CDK-5) mRNA levels, and glial fibrillary acidic protein (GFAP) expression. The cafeteria diet reduced PV-positive neurons in the granular layer, hilus, and CA1, as well as NPY-positive neurons in the hilus, without altering other GABAergic populations or overall GABA levels. The high-sugar diet induced a decrease in the number of PV-positive cells in CA3 and an increase in CB-positive cells in the hilus and CA1. No alterations were observed in the cholinergic varicosities. The cafeteria diet also reduced the relative mRNA expression of RELN without significant changes in BDNF and CDK5 levels. The cafeteria diet increased the number but reduced the length of the astrocyte processes. These data highlight the significance of determining the mechanisms mediating the observed effects of these diets and imply that the cognitive impairments previously found might be related to both the neuroinflammation process and the reduction in PV, NPY, and RELN expression in the hippocampal formation.
Collapse
Affiliation(s)
- Bárbara Mota
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Rita Brás
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
| | - Leonardo Araújo-Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Silva
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Pedro A. Pereira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - M. Dulce Madeira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Armando Cardoso
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
10
|
Sugimoto C, Perna MK, Regan SL, Tepe EA, Liou R, Fritz AL, Williams MT, Vorhees CV, Skelton MR. A Gad2 specific Slc6a8 deletion recapitulates the contextual and cued freezing deficits seen in Slc6a8 -/y mice. Brain Res 2024; 1825:148690. [PMID: 38030104 PMCID: PMC10875619 DOI: 10.1016/j.brainres.2023.148690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023]
Abstract
The creatine (Cr)-phosphocreatine shuttle is essential for ATP homeostasis. In humans, the absence of brain Cr causes significant intellectual disability, epilepsy, and language delay. Mutations of the creatine transporter (SLC6A8) are the most common cause of Cr deficiency. In rodents, Slc6a8 deletion causes deficits in spatial learning, novel object recognition (NOR), as well as in contextual and cued freezing. The mechanisms that underlie these cognitive deficits are not known. Due to the heterogeneous nature of the brain, it is important to determine which systems are affected by a loss of Cr. In this study, we generated mice lacking Slc6a8 in GABAergic neurons by crossing Slc6a8FL mice with Gad2-Cre mice. These Gad2-specific Slc6a8 knockout (cKO) mice, along with the ubiquitous Slc6a8 KO (Slc6a8-/y), Gad2-Cre+, and wild-type (WT) mice were tested in the Morris water maze, NOR, conditioned freezing, and the radial water maze. Similar to the Slc6a8-/y mice, cKO mice had reduced contextual and cued freezing compared with WT mice. The cKO mice had a mild spatial learning deficit during the reversal phase of the MWM, however they were not as pronounced as in Slc6a8-/y mice. In NOR, the Gad2-Cre mice spent less time with the novel object, similar to the reduced novel time in the cKO mice. There were no changes in radial water maze performance. Slc6a8 deletion in GABAergic neurons is sufficient to recapitulate the conditioned freezing deficits seen in Slc6a8-/y mice.
Collapse
Affiliation(s)
- Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Marla K Perna
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Samantha L Regan
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Erin A Tepe
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Rosalyn Liou
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Adam L Fritz
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| |
Collapse
|
11
|
Sohn J. Synaptic configuration and reconfiguration in the neocortex are spatiotemporally selective. Anat Sci Int 2024; 99:17-33. [PMID: 37837522 PMCID: PMC10771605 DOI: 10.1007/s12565-023-00743-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 10/16/2023]
Abstract
Brain computation relies on the neural networks. Neurons extend the neurites such as dendrites and axons, and the contacts of these neurites that form chemical synapses are the biological basis of signal transmissions in the central nervous system. Individual neuronal outputs can influence the other neurons within the range of the axonal spread, while the activities of single neurons can be affected by the afferents in their somatodendritic fields. The morphological profile, therefore, binds the functional role each neuron can play. In addition, synaptic connectivity among neurons displays preference based on the characteristics of presynaptic and postsynaptic neurons. Here, the author reviews the "spatial" and "temporal" connection selectivity in the neocortex. The histological description of the neocortical circuitry depends primarily on the classification of cell types, and the development of gene engineering techniques allows the cell type-specific visualization of dendrites and axons as well as somata. Using genetic labeling of particular cell populations combined with immunohistochemistry and imaging at a subcellular spatial resolution, we revealed the "spatial selectivity" of cortical wirings in which synapses are non-uniformly distributed on the subcellular somatodendritic domains in a presynaptic cell type-specific manner. In addition, cortical synaptic dynamics in learning exhibit presynaptic cell type-dependent "temporal selectivity": corticocortical synapses appear only transiently during the learning phase, while learning-induced new thalamocortical synapses persist, indicating that distinct circuits may supervise learning-specific ephemeral synapse and memory-specific immortal synapse formation. The selectivity of spatial configuration and temporal reconfiguration in the neural circuitry may govern diverse functions in the neocortex.
Collapse
Affiliation(s)
- Jaerin Sohn
- Department of Systematic Anatomy and Neurobiology, Graduate School of Dentistry, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
12
|
Gräff J. Engrams of Fear Memory Attenuation. ADVANCES IN NEUROBIOLOGY 2024; 38:149-161. [PMID: 39008015 DOI: 10.1007/978-3-031-62983-9_9] [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: 07/16/2024]
Abstract
Fear attenuation is an etiologically relevant process for animal survival, since once acquired information needs to be continuously updated in the face of changing environmental contingencies. Thus, when situations are encountered that were originally perceived as fearful but are no longer so, fear must be attenuated, otherwise, it risks becoming maladaptive. But what happens to the original memory trace of fear during fear attenuation? In this chapter, we review the studies that have started to approach this question from an engram perspective. We find evidence pointing to both the original memory trace of fear being suppressed, as well as it being updated towards safety. These seemingly conflicting results reflect a well-established dichotomy in the field of fear memory attenuation, namely whether fear attenuation is mediated by an inhibitory mechanism that suppresses fear expression, called extinction, or by an updating mechanism that allows the fear memory to reconsolidate in a different form, called reconsolidation-updating. Which of these scenarios takes the upper hand is ultimately influenced by the behavioral paradigms used to induce fear attenuation, but is an important area for further study as the precise cell populations underlying fear attenuation and the molecular mechanisms therein can now be understood at unprecedented resolution.
Collapse
Affiliation(s)
- Johannes Gräff
- Brain Mind Institute, School of Life Sciences, EPFL, Lausanne, Switzerland.
| |
Collapse
|
13
|
Stubbendorff C, Hale E, Day HLL, Smith J, Alvaro GS, Large CH, Stevenson CW. Pharmacological modulation of Kv3 voltage-gated potassium channels regulates fear discrimination and expression in a response-dependent manner. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110829. [PMID: 37451593 DOI: 10.1016/j.pnpbp.2023.110829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Various psychiatric diseases are characterized by aberrant cognition and emotional regulation. This includes inappropriately attributing affective salience to innocuous cues, which can be investigated using translationally relevant preclinical models of fear discrimination. Activity in the underpinning corticolimbic circuitry is governed by parvalbumin-expressing GABAergic interneurons, which also regulate fear discrimination. Kv3 voltage-gated potassium channels are highly expressed in these neurons and are important for controlling their activity, suggesting that pharmacological Kv3 modulation may regulate fear discrimination. We determined the effect of the positive Kv3 modulator AUT00206 given systemically to female rats undergoing limited or extended auditory fear discrimination training, which we have previously shown results in more discrimination or generalization, respectively, based on freezing at retrieval. We also characterized darting and other active fear-related responses. We found that limited training resulted in more discrimination based on freezing, which was unaffected by AUT00206. In contrast, extended training resulted in more generalization based on freezing and the emergence of discrimination based on darting during training and, to a lesser extent, at retrieval. Importantly, AUT00206 given before extended training had dissociable effects on fear discrimination and expression at retrieval depending on the response examined. While AUT00206 mitigated generalization without affecting expression based on freezing, it reduced expression without affecting discrimination based on darting, although darting levels were low overall. These results indicate that pharmacological Kv3 modulation regulates fear discrimination and expression in a response-dependent manner. They also raise the possibility that targeting Kv3 channels may ameliorate perturbed cognition and emotional regulation in psychiatric disease.
Collapse
Affiliation(s)
- Christine Stubbendorff
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Ed Hale
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Harriet L L Day
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Jessica Smith
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Giuseppe S Alvaro
- Autifony Therapeutics Limited, Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage SG1 2FX, UK
| | - Charles H Large
- Autifony Therapeutics Limited, Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage SG1 2FX, UK
| | - Carl W Stevenson
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| |
Collapse
|
14
|
Hijazi S, Smit AB, van Kesteren RE. Fast-spiking parvalbumin-positive interneurons in brain physiology and Alzheimer's disease. Mol Psychiatry 2023; 28:4954-4967. [PMID: 37419975 PMCID: PMC11041664 DOI: 10.1038/s41380-023-02168-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
Fast-spiking parvalbumin (PV) interneurons are inhibitory interneurons with unique morphological and functional properties that allow them to precisely control local circuitry, brain networks and memory processing. Since the discovery in 1987 that PV is expressed in a subset of fast-spiking GABAergic inhibitory neurons, our knowledge of the complex molecular and physiological properties of these cells has been expanding. In this review, we highlight the specific properties of PV neurons that allow them to fire at high frequency and with high reliability, enabling them to control network oscillations and shape the encoding, consolidation and retrieval of memories. We next discuss multiple studies reporting PV neuron impairment as a critical step in neuronal network dysfunction and cognitive decline in mouse models of Alzheimer's disease (AD). Finally, we propose potential mechanisms underlying PV neuron dysfunction in AD and we argue that early changes in PV neuron activity could be a causal step in AD-associated network and memory impairment and a significant contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Sara Hijazi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Nagy-Pál P, Veres JM, Fekete Z, Karlócai MR, Weisz F, Barabás B, Reéb Z, Hájos N. Structural Organization of Perisomatic Inhibition in the Mouse Medial Prefrontal Cortex. J Neurosci 2023; 43:6972-6987. [PMID: 37640552 PMCID: PMC10586541 DOI: 10.1523/jneurosci.0432-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Perisomatic inhibition profoundly controls neural function. However, the structural organization of inhibitory circuits giving rise to the perisomatic inhibition in the higher-order cortices is not completely known. Here, we performed a comprehensive analysis of those GABAergic cells in the medial prefrontal cortex (mPFC) that provide inputs onto the somata and proximal dendrites of pyramidal neurons. Our results show that most GABAergic axonal varicosities contacting the perisomatic region of superficial (layer 2/3) and deep (layer 5) pyramidal cells express parvalbumin (PV) or cannabinoid receptor type 1 (CB1). Further, we found that the ratio of PV/CB1 GABAergic inputs is larger on the somatic membrane surface of pyramidal tract neurons in comparison with those projecting to the contralateral hemisphere. Our morphologic analysis of in vitro labeled PV+ basket cells (PVBC) and CCK/CB1+ basket cells (CCKBC) revealed differences in many features. PVBC dendrites and axons arborized preferentially within the layer where their soma was located. In contrast, the axons of CCKBCs expanded throughout layers, although their dendrites were found preferentially either in superficial or deep layers. Finally, using anterograde trans-synaptic tracing we observed that PVBCs are preferentially innervated by thalamic and basal amygdala afferents in layers 5a and 5b, respectively. Thus, our results suggest that PVBCs can control the local circuit operation in a layer-specific manner via their characteristic arborization, whereas CCKBCs rather provide cross-layer inhibition in the mPFC.SIGNIFICANCE STATEMENT Inhibitory cells in cortical circuits are crucial for the precise control of local network activity. Nevertheless, in higher-order cortical areas that are involved in cognitive functions like decision-making, working memory, and cognitive flexibility, the structural organization of inhibitory cell circuits is not completely understood. In this study we show that perisomatic inhibitory control of excitatory cells in the medial prefrontal cortex is performed by two types of basket cells endowed with different morphologic properties that provide inhibitory inputs with distinct layer specificity on cells projecting to disparate areas. Revealing this difference in innervation strategy of the two basket cell types is a key step toward understanding how they fulfill their distinct roles in cortical network operations.
Collapse
Affiliation(s)
- Petra Nagy-Pál
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Judit M Veres
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Zsuzsanna Fekete
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Mária R Karlócai
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Filippo Weisz
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Bence Barabás
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Zsófia Reéb
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Norbert Hájos
- Eötvös Loránd Research Network Institute of Experimental Medicine, 1083 Budapest, Hungary
- Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Bloomington, Indiana 47405
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, Indiana 47405
| |
Collapse
|
16
|
Tzilivaki A, Tukker JJ, Maier N, Poirazi P, Sammons RP, Schmitz D. Hippocampal GABAergic interneurons and memory. Neuron 2023; 111:3154-3175. [PMID: 37467748 PMCID: PMC10593603 DOI: 10.1016/j.neuron.2023.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
One of the most captivating questions in neuroscience revolves around the brain's ability to efficiently and durably capture and store information. It must process continuous input from sensory organs while also encoding memories that can persist throughout a lifetime. What are the cellular-, subcellular-, and network-level mechanisms that underlie this remarkable capacity for long-term information storage? Furthermore, what contributions do distinct types of GABAergic interneurons make to this process? As the hippocampus plays a pivotal role in memory, our review focuses on three aspects: (1) delineation of hippocampal interneuron types and their connectivity, (2) interneuron plasticity, and (3) activity patterns of interneurons during memory-related rhythms, including the role of long-range interneurons and disinhibition. We explore how these three elements, together showcasing the remarkable diversity of inhibitory circuits, shape the processing of memories in the hippocampus.
Collapse
Affiliation(s)
- Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany
| | - John J Tukker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Nikolaus Maier
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Panayiota Poirazi
- Foundation for Research and Technology Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), N. Plastira 100, Heraklion, Crete, Greece
| | - Rosanna P Sammons
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany; Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Philippstrasse. 13, 10115 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
17
|
Crestani AP, Cicvaric A, Yiu AP. Editorial: New insights into synaptic plasticity in fear conditioning. Front Synaptic Neurosci 2023; 15:1270701. [PMID: 37779862 PMCID: PMC10535560 DOI: 10.3389/fnsyn.2023.1270701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Affiliation(s)
- Ana P. Crestani
- Center for Neuroscience, University of California, Davis, Davis, CA, United States
| | - Ana Cicvaric
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | | |
Collapse
|
18
|
Sun XY, Liu L, Song YT, Wu T, Zheng T, Hao JR, Cao JL, Gao C. Two parallel medial prefrontal cortex-amygdala pathways mediate memory deficits via glutamatergic projection in surgery mice. Cell Rep 2023; 42:112719. [PMID: 37392387 DOI: 10.1016/j.celrep.2023.112719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/19/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023] Open
Abstract
The neural circuit mechanisms underlying postoperative cognitive dysfunction (POCD) remain elusive. We hypothesized that projections from the medial prefrontal cortex (mPFC) to the amygdala are involved in POCD. A mouse model of POCD in which isoflurane (1.5%) combined with laparotomy was used. Virally assisted tracing techniques were used to label the relevant pathways. Fear conditioning, immunofluorescence, whole-cell patch-clamp recordings, and chemogenetic and optogenetic techniques were applied to investigate the role of mPFC-amygdala projections in POCD. We find that surgery impairs memory consolidation but not retrieval of consolidated memories. In POCD mice, the glutamatergic pathway from the prelimbic cortex to the basolateral amygdala (PL-BLA) shows reduced activity, whereas the glutamatergic pathway from the infralimbic cortex to the basomedial amygdala (IL-BMA) shows enhanced activity. Our study indicates that the hypoactivity in the PL-BLA pathway interrupts memory consolidation, whereas the hyperactivity in the IL-BMA promotes memory extinction, in POCD mice.
Collapse
Affiliation(s)
- Xiao-Yu Sun
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai 200127, China
| | - Le Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yu-Tong Song
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Tong Wu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Teng Zheng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jing-Ru Hao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jun-Li Cao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Can Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
19
|
Cummings KA, Bayshtok S, Dong TN, Kenny PJ, Clem RL. Control of fear by discrete prefrontal GABAergic populations encoding valence-specific information. Neuron 2022; 110:3036-3052.e5. [PMID: 35944526 PMCID: PMC10009874 DOI: 10.1016/j.neuron.2022.07.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 05/12/2022] [Accepted: 07/08/2022] [Indexed: 11/25/2022]
Abstract
Neurons activated by learning have been ascribed the unique potential to encode memory, but the functional contribution of discrete cell types remains poorly understood. In particular, it is unclear whether learning engages specific GABAergic interneurons and, if so, whether they differ functionally from interneurons recruited by other experiences. Here, we show that fear conditioning activates a heterogeneous neuronal population in the medial prefrontal cortex (mPFC) that is largely comprised of somatostatin-expressing interneurons (SST-INs). Using intersectional genetic approaches, we demonstrate that fear-learning-activated SST-INs exhibit distinct circuit properties and are selectively reactivated to mediate cue-evoked memory expression. In contrast, an orthogonal population of SST-INs activated by morphine experience exerts opposing control over fear and supports reward-like motivational effects. These results outline an important role for discrete subsets of GABAergic cells in emotional learning and point to an unappreciated capacity for functional specialization among SST-INs.
Collapse
Affiliation(s)
- Kirstie A Cummings
- Department of Neurobiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA.
| | - Sabina Bayshtok
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tri N Dong
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul J Kenny
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roger L Clem
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
20
|
Lv N, Wang Y, Liu Y, Tang J, Lei Q, Wang Y, Wei H. Decreased Microglia in Pax2 Mutant Mice Leads to Impaired Learning and Memory. ACS Chem Neurosci 2022; 13:2490-2502. [PMID: 35929805 DOI: 10.1021/acschemneuro.2c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Impaired learning and memory ability is one of the characteristics of a variety of neurological diseases, and its molecular mechanisms are complex and diverse and are regulated by a variety of factors. It is generally believed that synaptic plasticity plays an important role in the process of learning and memory. The protein encoded by the Pax2 gene is a transcription factor involved in neuron migration and cell fate determination during neural development. Mice knocked out of BDNF in the Pax2 lineage-derived interneuron precursor exhibited learning disabilities and severe cognitive impairment. In this study, Pax2 heterozygous gene (Pax2+/- mice) deletion mice were used as the research objects and behavioral tests were used to observe the effect of Pax2 gene deletion on learning and memory ability; morphological and molecular biological methods were used to observe the effect of Pax2 gene deletion on the neural structure. Single-cell transcriptome sequencing was used to observe the cell subtypes and differentially expressed genes (DEGs) and signaling pathways affected by Pax2 gene deletion and the possible molecular mechanisms. The results showed that Pax2+/- mice had impaired learning and memory ability, abnormal synaptic structure, and significantly reduced number of microglia clusters, and DEGs were associated with pro-inflammatory chemokines. Finally, we speculate that Pax2 gene deletion may lead to abnormal chemokines and chemokine receptors by affecting microglia.
Collapse
Affiliation(s)
- Na Lv
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Department of Physiology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Ying Wang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Yongfeng Liu
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China
| | - Jiaming Tang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Graduate College, Shanxi University of Chinese Medicine, Taiyuan 030024, China
| | - Qiang Lei
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Department of Physiology, School of Basic Medicine, Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Yizhuo Wang
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| | - Hongen Wei
- Department of Neurology, Shanxi Provincial People's Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan 030012, China.,Shanxi Key Laboratory of Brain Disease Control, Shanxi Provincial People's Hospital, Taiyuan 030012, China
| |
Collapse
|
21
|
Guzmán-Ramos K, Osorio-Gómez D, Bermúdez-Rattoni F. Cognitive impairment in alzheimer’s and metabolic diseases: A catecholaminergic hypothesis. Neuroscience 2022; 497:308-323. [DOI: 10.1016/j.neuroscience.2022.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022]
|
22
|
Canto-Bustos M, Friason FK, Bassi C, Oswald AMM. Disinhibitory Circuitry Gates Associative Synaptic Plasticity in Olfactory Cortex. J Neurosci 2022; 42:2942-2950. [PMID: 35181596 PMCID: PMC8985865 DOI: 10.1523/jneurosci.1369-21.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/02/2021] [Accepted: 12/29/2021] [Indexed: 11/21/2022] Open
Abstract
Inhibitory microcircuits play an essential role in regulating cortical responses to sensory stimuli. Interneurons that inhibit dendritic or somatic integration act as gatekeepers for neural activity, synaptic plasticity, and the formation of sensory representations. Conversely, interneurons that selectively inhibit other interneurons can open gates through disinhibition. In the anterior piriform cortex, relief of inhibition permits associative LTP of excitatory synapses between pyramidal neurons. However, the interneurons and circuits mediating disinhibition have not been elucidated. In this study, we use an optogenetic approach in mice of both sexes to identify the inhibitory interneurons and disinhibitory circuits that regulate LTP. We focused on three prominent interneuron classes: somatostatin (SST), parvalbumin (PV), and vasoactive intestinal polypeptide (VIP) interneurons. We find that LTP is gated by the inactivation SST or PV interneurons and by the activation of VIP interneurons. Further, VIP interneurons strongly inhibit putative SST cells during LTP induction but only weakly inhibit PV interneurons. Together, these findings suggest that VIP interneurons mediate a disinhibitory circuit that gates synaptic plasticity during the formation of olfactory representations.SIGNIFICANCE STATEMENT Inhibitory interneurons stabilize neural activity during sensory processing. However, inhibition must also be modulated to allow sensory experience shape neural responses. In olfactory cortex, inhibition regulates activity-dependent increases in excitatory synaptic strength that accompany odor learning. We identify two inhibitory interneuron classes that act as gatekeepers preventing excitatory enhancement. We demonstrate that driving a third class of interneurons inhibits the gatekeepers and opens the gate for excitatory enhancement. All three inhibitory neuron classes comprise disinhibitory microcircuit motifs found throughout the cortex. Our findings suggest that a common disinhibitory microcircuit promotes changes in synaptic strength during sensory processing and learning.
Collapse
Affiliation(s)
- Martha Canto-Bustos
- Department of Neuroscience
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - F Kathryn Friason
- Department of Neuroscience
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | | | - Anne-Marie M Oswald
- Department of Neuroscience
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
- Department of Neurobiology
- Neuroscience Institute, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
23
|
TrkA-cholinergic signaling modulates fear encoding and extinction learning in PTSD-like behavior. Transl Psychiatry 2022; 12:111. [PMID: 35301275 PMCID: PMC8931170 DOI: 10.1038/s41398-022-01869-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/19/2022] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
Recent studies have suggested that the use of cognitive enhancers as adjuncts to exposure-based therapy in individuals suffering from post-traumatic stress disorder (PTSD) may be beneficial. Brain cholinergic signaling through basal forebrain projections to the hippocampus is an established pathway mediating fear response and cognitive flexibility. Here we employed a genetic strategy to enhance cholinergic activity through increased signaling of the NGF receptor TrkA. This strategy leads to increased levels of the marker of cholinergic activation, acetylcholine synthesizing enzyme choline acetyltransferase, in forebrain cholinergic regions and their projection areas such as the hippocampus. Mice with increased cholinergic activity do not display any neurobehavioral abnormalities except a selective attenuation of fear response and lower fear expression in extinction trials. Reduction in fear response is rescued by the GABA antagonist picrotoxin in mutant mice, and, in wild-type mice, is mimicked by the GABA agonist midazolam suggesting that GABA can modulate cholinergic functions on fear circuitries. Importantly, mutant mice also show a reduction in fear processing under stress conditions in a single prolonged stress (SPS) model of PTSD-like behavior, and augmentation of cholinergic signaling by the drug donepezil in wild-type mice promotes extinction learning in a similar SPS model of PTSD-like behavior. Donepezil is already in clinical use for the treatment of dementia suggesting a new translational application of this drug for improving exposure-based psychotherapy in PTSD patients.
Collapse
|
24
|
Oberländer K, Witte V, Mallien AS, Gass P, Bengtson CP, Bading H. Dysregulation of Npas4 and Inhba expression and an altered excitation-inhibition balance are associated with cognitive deficits in DBA/2 mice. Learn Mem 2022; 29:55-70. [PMID: 35042829 PMCID: PMC8774195 DOI: 10.1101/lm.053527.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/28/2021] [Indexed: 02/03/2023]
Abstract
Differences in the learning associated transcriptional profiles between mouse strains with distinct learning abilities could provide insight into the molecular basis of learning and memory. The inbred mouse strain DBA/2 shows deficits in hippocampus-dependent memory, yet the transcriptional responses to learning and the underlying mechanisms of the impairments are unknown. Comparing DBA/2J mice with the reference standard C57BL/6N mouse strain we verify an enhanced susceptibility to kainic acid induced seizures, confirm impairments in hippocampus-dependent spatial memory tasks and uncover additional behavioral abnormalities including deficits in hippocampus-independent learning. Surprisingly, we found no broad dysfunction of the DBA/2J strain in immediate early gene (IEG) activation but instead report brain region-specific and gene-specific alterations. The learning-associated IEGs Arc, c-Fos, and Nr4a1 showed no DBA/2J deficits in basal or synaptic activity induced gene expression in hippocampal or cortical primary neuronal cultures or in the CA1, CA3, or retrosplenial cortex following spatial object recognition (SOR) training in vivo. However, the parietal cortex showed reduced and the dentate gyrus showed enhanced SOR-evoked induction of most IEGs. All DBA/2J hippocampal regions exhibited elevated basal expression of inhibin β A (Inhba) and a learning-associated superinduction of the transcription factor neuronal Per-Arnt-Sim domain protein 4 (Npas4) known to regulate the synaptic excitation-inhibition balance. In line with this, CA1 pyramidal neurons of DBA/2J mice showed fewer inhibitory and more excitatory miniature postsynaptic currents but no alteration in most other electrophysiological properties or gross dendritic morphology. The dysregulation of Npas4 and Inhba expression and synaptic connectivity may underlie the cognitive deficits and increased susceptibility to seizures of DBA/2J mice.
Collapse
Affiliation(s)
- Kristin Oberländer
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Victoria Witte
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Anne Stephanie Mallien
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, 68159 Mannheim, Germany
| | - C. Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Whitehurst LN, Subramoniam A, Krystal A, Prather AA. Links between the brain and body during sleep: implications for memory processing. Trends Neurosci 2022; 45:212-223. [PMID: 35074220 DOI: 10.1016/j.tins.2021.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
Sleep is intimately related to memory processes. The established view is that the transformation of experiences into long-term memories is linked to sleep-related CNS function. However, there is increasing evidence that the autonomic nervous system (ANS), long recognized to modulate cognition during waking, can impact memory processing during sleep. Here, we review human research that examines the role of autonomic activity and sleep in memory formation. We argue that autonomic activity during sleep may set the stage for the CNS dynamics associated with sleep and memory stability and integration. Further, we consider how the link between ANS activity and polysomnographic markers of sleep may help elucidate both healthy and pathological cognitive aging in humans.
Collapse
Affiliation(s)
| | | | - Andrew Krystal
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Aric A Prather
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
26
|
Tzilivaki A, Kastellakis G, Schmitz D, Poirazi P. GABAergic Interneurons with Nonlinear Dendrites: From Neuronal Computations to Memory Engrams. Neuroscience 2021; 489:34-43. [PMID: 34843894 DOI: 10.1016/j.neuroscience.2021.11.033] [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: 05/16/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
GABAergic interneurons (INs) are a highly diverse class of neurons in the mammalian brain with a critical role in orchestrating multiple cognitive functions and maintaining the balance of excitation/inhibition across neuronal circuitries. In this perspective, we discuss recent findings regarding the ability of some IN subtypes to integrate incoming inputs in nonlinear ways within their dendritic branches. These recently discovered features may endow the specific INs with advanced computing capabilities, whose breadth and functional contributions remain an open question. Along these lines, we discuss theoretical and experimental evidence regarding the potential role of nonlinear IN dendrites in advancing single neuron computations and contributing to memory formation.
Collapse
Affiliation(s)
- Alexandra Tzilivaki
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany; Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Foundation for Research and Technology Hellas, Institute of Molecular Biology and Biotechnology, Greece
| | - George Kastellakis
- Foundation for Research and Technology Hellas, Institute of Molecular Biology and Biotechnology, Greece
| | - Dietmar Schmitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany; Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Panayiota Poirazi
- Foundation for Research and Technology Hellas, Institute of Molecular Biology and Biotechnology, Greece.
| |
Collapse
|
27
|
Li HD, Li DN, Yang L, Long C. Deficiency of the CYLD Impairs Fear Memory of Mice and Disrupts Neuronal Activity and Synaptic Transmission in the Basolateral Amygdala. Front Cell Neurosci 2021; 15:740165. [PMID: 34602983 PMCID: PMC8485066 DOI: 10.3389/fncel.2021.740165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Fear learning and memory are crucial for animal survival. Abnormal fear memory is a hallmark of many neuropsychiatric disorders. Appropriate neuronal activation and excitability in the basolateral amygdala (BLA) are necessary for the formation of fear memory. The gene cylindromatosis (Cyld), which encodes a lysine-63 deubiquitinase, is expressed in several brain regions including the amygdala. The functions of the cylindromatosis protein (CYLD) in the regulation of the neuronal activity, neural circuits and fear memory, remain largely unknown, however. Here, we report that Cyld knockout impairs amygdala-dependent tone-cued fear memory. The number of c-Fos+ neurons responding to the tone-cued fear test was reduced in the BLA of Cyld–/– mice, suggesting that the absence of CYLD causes aberrant neuronal activation. We found that this aberrant neuronal activation in the BLA of Cyld–/– mice may relate to the decreased excitability of principal neurons. Another possibility of aberrant neuronal activation could be the impaired excitatory synaptic transmission in the BLA of Cyld–/– mice. Specifically, both the frequency of spontaneous excitatory postsynaptic currents and the amplitude of miniature excitatory postsynaptic currents in BLA principal neurons were decreased. In addition, Cyld mutation caused an increase in both the frequency of miniature inhibitory postsynaptic currents in principal neurons and the number of parvalbumin+ interneurons, consistent with excessive local circuit inhibition in the BLA of Cyld–/– mice. Taken together, these results suggest that CYLD deficiency disrupts the neuronal activity and synaptic transmission in the BLA of mice which may contribute to the impaired fear memory observed in Cyld–/– mice.
Collapse
Affiliation(s)
- Hui-Dong Li
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Dan-Ni Li
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
28
|
Jiménez-Balado J, Ycaza Herrera A, Igwe K, Klem L, Buyukturkoglu K, Irimia A, Chen L, Guo J, Brickman AM, Eich TS. Reduced Hippocampal GABA+ Is Associated With Poorer Episodic Memory in Healthy Older Women: A Pilot Study. Front Behav Neurosci 2021; 15:695416. [PMID: 34512283 PMCID: PMC8427754 DOI: 10.3389/fnbeh.2021.695416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/13/2021] [Indexed: 01/13/2023] Open
Abstract
Background: The current pilot study was designed to examine the association between hippocampal γ-aminobutyric acid (GABA) concentration and episodic memory in older individuals, as well as the impact of two major risk factors for Alzheimer’s disease (AD)—female sex and Apolipoprotein ε4 (ApoE ε4) genotype—on this relationship. Methods: Twenty healthy, community-dwelling individuals aged 50–71 (11 women) took part in the study. Episodic memory was evaluated using a Directed Forgetting task, and GABA+ was measured in the right hippocampus using a Mescher-Garwood point-resolved magnetic resonance spectroscopy (MRS) sequence. Multiple linear regression models were used to quantify the relationship between episodic memory, GABA+, ApoE ɛ4, and sex, controlling for age and education. Results: While GABA+ did not interact with ApoE ɛ4 carrier status to influence episodic memory (p = 0.757), the relationship between GABA+ and episodic memory was moderated by sex: lower GABA+ predicted worse memory in women such that, for each standard deviation decrease in GABA+ concentration, memory scores were reduced by 11% (p = 0.001). Conclusions: This pilot study suggests that sex, but not ApoE ɛ4 genotype, moderates the relationship between hippocampal GABA+ and episodic memory, such that women with lower GABA+ concentration show worse memory performance. These findings, which must be interpreted with caution given the small sample size, may serve as a starting point for larger studies using multimodal neuroimaging to understand the contributions of GABA metabolism to age-related memory decline.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Alexandra Ycaza Herrera
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Kay Igwe
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States.,Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | - Lynda Klem
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | | | - Andrei Irimia
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States.,Corwin D. Denney Research Center, Department of Biomedical Engineering, Andrew Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Liu Chen
- Department of Electrical Engineering, Columbia University, New York, NY, United States
| | - Jia Guo
- Department of Psychiatry, Columbia University, New York, NY, United States
| | - Adam M Brickman
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States.,Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States.,Gertrude H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States.,Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States.,Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
29
|
Guadagno A, Belliveau C, Mechawar N, Walker CD. Effects of Early Life Stress on the Developing Basolateral Amygdala-Prefrontal Cortex Circuit: The Emerging Role of Local Inhibition and Perineuronal Nets. Front Hum Neurosci 2021; 15:669120. [PMID: 34512291 PMCID: PMC8426628 DOI: 10.3389/fnhum.2021.669120] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
The links between early life stress (ELS) and the emergence of psychopathology such as increased anxiety and depression are now well established, although the specific neurobiological and developmental mechanisms that translate ELS into poor health outcomes are still unclear. The consequences of ELS are complex because they depend on the form and severity of early stress, duration, and age of exposure as well as co-occurrence with other forms of physical or psychological trauma. The long term effects of ELS on the corticolimbic circuit underlying emotional and social behavior are particularly salient because ELS occurs during critical developmental periods in the establishment of this circuit, its local balance of inhibition:excitation and its connections with other neuronal pathways. Using examples drawn from the human and rodent literature, we review some of the consequences of ELS on the development of the corticolimbic circuit and how it might impact fear regulation in a sex- and hemispheric-dependent manner in both humans and rodents. We explore the effects of ELS on local inhibitory neurons and the formation of perineuronal nets (PNNs) that terminate critical periods of plasticity and promote the formation of stable local networks. Overall, the bulk of ELS studies report transient and/or long lasting alterations in both glutamatergic circuits and local inhibitory interneurons (INs) and their associated PNNs. Since the activity of INs plays a key role in the maturation of cortical regions and the formation of local field potentials, alterations in these INs triggered by ELS might critically participate in the development of psychiatric disorders in adulthood, including impaired fear extinction and anxiety behavior.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Claudia Belliveau
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
30
|
Cummings KA, Lacagnina AF, Clem RL. GABAergic microcircuitry of fear memory encoding. Neurobiol Learn Mem 2021; 184:107504. [PMID: 34425220 DOI: 10.1016/j.nlm.2021.107504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 12/30/2022]
Abstract
The paradigm of fear conditioning is largely responsible for our current understanding of how memories are encoded at the cellular level. Its most fundamental underlying mechanism is considered to be plasticity of synaptic connections between excitatory projection neurons (PNs). However, recent studies suggest that while PNs execute critical memory functions, their activity at key stages of learning and recall is extensively orchestrated by a diverse array of GABAergic interneurons (INs). Here we review the contributions of genetically-defined INs to processing of threat-related stimuli in fear conditioning, with a particular focus on how synaptic interactions within interconnected networks of INs modulates PN activity through both inhibition and disinhibition. Furthermore, we discuss accumulating evidence that GABAergic microcircuits are an important locus for synaptic plasticity during fear learning and therefore a viable substrate for long-term memory. These findings suggest that further investigation of INs could unlock unique conceptual insights into the organization and function of fear memory networks.
Collapse
Affiliation(s)
- Kirstie A Cummings
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neurobiology, University of Alabama Birmingham School of Medicine, Birmingham, AL 35294, United States
| | - Anthony F Lacagnina
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Roger L Clem
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
31
|
Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:146-159. [PMID: 33573856 PMCID: PMC8292162 DOI: 10.1016/j.semcdb.2021.01.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
In this review, we focus on the potential role of the γ-aminobutyric acidergic (GABAergic) system in age-related episodic memory impairments in humans, with a particular focus on Alzheimer's disease (AD). Well-established animal models have shown that GABA plays a central role in regulating and synchronizing neuronal signaling in the hippocampus, a brain area critical for episodic memory that undergoes early and significant morphologic and functional changes in the course of AD. Neuroimaging research in humans has documented hyperactivity in the hippocampus and losses of resting state functional connectivity in the Default Mode Network, a network that itself prominently includes the hippocampus-presaging episodic memory decline in individuals at-risk for AD. Apolipoprotein ε4, the highest genetic risk factor for AD, is associated with GABAergic dysfunction in animal models, and episodic memory impairments in humans. In combination, these findings suggest that GABA may be the linchpin in a complex system of factors that eventually leads to the principal clinical hallmark of AD: episodic memory loss. Here, we will review the current state of literature supporting this hypothesis. First, we will focus on the molecular and cellular basis of the GABAergic system and its role in memory and cognition. Next, we report the evidence of GABA dysregulations in AD and normal aging, both in animal models and human studies. Finally, we outline a model of GABAergic dysfunction based on the results of functional neuroimaging studies in humans, which have shown hippocampal hyperactivity to episodic memory tasks concurrent with and even preceding AD diagnosis, along with factors that may modulate this association.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
32
|
Giorgi C, Marinelli S. Roles and Transcriptional Responses of Inhibitory Neurons in Learning and Memory. Front Mol Neurosci 2021; 14:689952. [PMID: 34211369 PMCID: PMC8239217 DOI: 10.3389/fnmol.2021.689952] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/18/2021] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence supports a model whereby memories are encoded by sparse ensembles of neurons called engrams, activated during memory encoding and reactivated upon recall. An engram consists of a network of cells that undergo long-lasting modifications of their transcriptional programs and connectivity. Ground-breaking advancements in this field have been made possible by the creative exploitation of the characteristic transcriptional responses of neurons to activity, allowing both engram labeling and manipulation. Nevertheless, numerous aspects of engram cell-type composition and function remain to be addressed. As recent transcriptomic studies have revealed, memory encoding induces persistent transcriptional and functional changes in a plethora of neuronal subtypes and non-neuronal cells, including glutamatergic excitatory neurons, GABAergic inhibitory neurons, and glia cells. Dissecting the contribution of these different cellular classes to memory engram formation and activity is quite a challenging yet essential endeavor. In this review, we focus on the role played by the GABAergic inhibitory component of the engram through two complementary lenses. On one hand, we report on available physiological evidence addressing the involvement of inhibitory neurons to different stages of memory formation, consolidation, storage and recall. On the other, we capitalize on a growing number of transcriptomic studies that profile the transcriptional response of inhibitory neurons to activity, revealing important clues on their potential involvement in learning and memory processes. The picture that emerges suggests that inhibitory neurons are an essential component of the engram, likely involved in engram allocation, in tuning engram excitation and in storing the memory trace.
Collapse
Affiliation(s)
- Corinna Giorgi
- CNR, Institute of Molecular Biology and Pathology, Rome, Italy.,European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Silvia Marinelli
- European Brain Research Institute (EBRI), Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
33
|
McDonald AJ, Mascagni F. Specific neuronal subpopulations in the rat basolateral amygdala express high levels of nonphosphorylated neurofilaments. J Comp Neurol 2021; 529:3292-3312. [PMID: 33960421 DOI: 10.1002/cne.25169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/25/2022]
Abstract
Cortical pyramidal neurons (PNs) containing nonphosphorylated neurofilaments (NNFs) localized with the SMI-32 monoclonal antibody have been shown to be especially vulnerable to degeneration in Alzheimer's disease (AD). The present investigation is the first to study the expression of SMI-32+ NNFs in neurons of the basolateral nuclear complex of the amygdala (BNC), which contains cortex-like PNs and nonpyramidal neurons (NPNs). We observed that PNs in the rat basolateral nucleus (BL), but not in the lateral (LAT) or basomedial (BM) nuclei, have significant levels of SMI-32-ir in their somata with antibody diluents that did not contain Triton X-100, but staining in these cells was greatly attenuated when the antibody diluent contained 0.3% Triton. Using Triton-containing diluents, we found that all SMI-32+ neurons in all three of the BNC nuclei were NPNs. Using a dual-labeling immunoperoxidase technique, we demonstrated that most of these SMI-32+ NPNs were parvalbumin-positive (PV+) or somatostatin-positive NPNs but not vasoactive intestinal peptide-positive or neuropeptide Y-positive NPNs. Using a technique that combines retrograde tracing with SMI-32 immunohistochemistry using intermediate levels of Triton in the diluent, we found that all BNC neurons projecting to the mediodorsal thalamic nucleus (MD) were large NPNs, and most were SMI-32+. In contrast, BNC neurons projecting to the ventral striatum or cerebral cortex were PNs that expressed low levels of SMI-32 immunoreactivity (SMI-32-ir) in the BL, and no SMI-32-ir in the LAT or BM. These data suggest that the main neuronal subpopulations in the BNC that degenerate in AD may be PV+ and MD-projecting NPNs.
Collapse
Affiliation(s)
- Alexander Joseph McDonald
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Franco Mascagni
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| |
Collapse
|
34
|
Apical intercalated cell cluster: A distinct sensory regulator in the amygdala. Cell Rep 2021; 35:109151. [PMID: 34010641 DOI: 10.1016/j.celrep.2021.109151] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 02/20/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
GABAergic neurons regulate different aspects of information processing in the amygdala. Among these are clusters of intercalated cells (ITCs), which have been implicated in fear-related behaviors. Although a few of the ITC clusters have been studied, the functional role of apical ITCs (apITCs) is unknown. Here, we combine monosynaptic rabies tracing with optogenetics and demonstrate that apITCs receive synaptic input from medial geniculate nucleus (MGm), posterior intralaminar nucleus (PIN), and medial dorsal nucleus of the thalamus and from a diverse range of cortical areas including temporal association, entorhinal, insular, piriform, and somatosensory cortex. Upon fear learning, PIN/MGm inputs are strengthened, indicative of their involvement in fear behaviors. 3-D reconstruction of apITCs reveals local arborization and innervation of the dorsal striatum and lateral amygdala. We further show that apITCs provide sensory feedforward inhibition to LA principal cells, a putative mechanism for controlling plasticity during fear learning.
Collapse
|
35
|
Tucker LB, Fu AH, McCabe JT. Hippocampal-Dependent Cognitive Dysfunction following Repeated Diffuse Rotational Brain Injury in Male and Female Mice. J Neurotrauma 2021; 38:1585-1606. [PMID: 33622092 PMCID: PMC8126427 DOI: 10.1089/neu.2021.0025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cognitive dysfunction is a common, often long-term complaint following acquired traumatic brain injury (TBI). Cognitive deficits suggest dysfunction in hippocampal circuits. The goal of the studies described here is to phenotype in both male and female mice the hippocampal-dependent learning and memory deficits resulting from TBI sustained by the Closed-Head Impact Model of Engineered Rotational Acceleration (CHIMERA) device—a model that delivers both a contact–concussion injury as well as unrestrained rotational head movement. Mice sustained either sham procedures or four injuries (0.7 J, 24-h intervals). Spatial learning and memory skills assessed in the Morris water maze (MWM) approximately 3 weeks following injuries were significantly impaired by brain injuries; however, slower swimming speeds and poor performance on visible platform trials suggest that measurement of cognitive impairment with this test is confounded by injury-induced motor and/or visual impairments. A separate experiment confirmed hippocampal-dependent cognitive deficits with trace fear conditioning (TFC), a behavioral test less dependent on motor and visual function. Male mice had greater injury-induced deficits on both the MWM and TFC tests than female mice. Pathologically, the injury was characterized by white matter damage as observed by silver staining and glial fibrillary acidic protein (astrogliosis) in the optic tracts, with milder damage seen in the corpus callosum, and fimbria and brainstem (cerebral peduncles) of some animals. No changes in the density of GABAergic parvalbumin-expressing cells in the hippocampus, amygdala, or parietal cortex were found. This experiment confirmed significant sexually dimorphic cognitive impairments following a repeated, diffuse brain injury.
Collapse
Affiliation(s)
- Laura B Tucker
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda H Fu
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Center for Neuroscience and Regenerative Medicine, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Maturation of amygdala inputs regulate shifts in social and fear behaviors: A substrate for developmental effects of stress. Neurosci Biobehav Rev 2021; 125:11-25. [PMID: 33581221 DOI: 10.1016/j.neubiorev.2021.01.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 11/21/2022]
Abstract
Stress can negatively impact brain function and behaviors across the lifespan. However, stressors during adolescence have particularly harmful effects on brain maturation, and on fear and social behaviors that extend beyond adolescence. Throughout development, social behaviors are refined and the ability to suppress fear increases, both of which are dependent on amygdala activity. We review rodent literature focusing on developmental changes in social and fear behaviors, cortico-amygdala circuits underlying these changes, and how this circuitry is altered by stress. We first describe changes in fear and social behaviors from adolescence to adulthood and parallel developmental changes in cortico-amygdala circuitry. We propose a framework in which maturation of cortical inputs to the amygdala promote changes in social drive and fear regulation, and the particularly damaging effects of stress during adolescence may occur through lasting changes in this circuit. This framework may explain why anxiety and social pathologies commonly co-occur, adolescents are especially vulnerable to stressors impacting social and fear behaviors, and predisposed towards psychiatric disorders related to abnormal cortico-amygdala circuits.
Collapse
|
37
|
McDonald AJ. Immunohistochemical Identification of Interneuronal Subpopulations in the Basolateral Amygdala of the Rhesus Monkey (Macaca mulatta). Neuroscience 2021; 455:113-127. [PMID: 33359654 PMCID: PMC7855802 DOI: 10.1016/j.neuroscience.2020.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022]
Abstract
Inhibitory circuits in the basolateral nuclear complex of the amygdala (BNC) critical for controlling the acquisition, expression, and extinction of emotional responses are mediated by GABAergic interneurons (INs). Studies in rodents have demonstrated that separate IN subpopulations, identified by their expression of calcium-binding proteins and neuropeptides, play discrete roles in the intrinsic circuitry of the BNC. Far less is known about IN subpopulations in primates. In order to fill in this gap in our understanding of primate INs, the present investigation used dual-labeling immunohistochemistry for IN markers to identify subpopulations expressing cholecystokinin (CCK), calbindin (CB), calretinin (CR), and somatostatin (SOM) in somata and axon terminals in the monkey BNC. In general, colocalization patterns seen in somata and axon terminals were similar. It was found that there was virtually no colocalization of CB and CR, the two calcium-binding proteins investigated. Three subtypes of CCK-immunoreactive (CCK+) INs were identified on the basis of their expression of CR or CB: (1) CCK+/CR+; (2) CCK+/CB+); and (3) CCK+/CR-/CB-. Almost no colocalization of CCK with SOM was observed, but there was extensive colocalization of SOM and CB. CCK+, CR+, and CCK+/CR+ double-labeled axon terminals were seen surrounding pyramidal cell somata in basket-like plexuses, as well as in the neuropil. CB+, SOM+, and CB+/SOM+ terminals did not form baskets, suggesting that these IN subpopulations are mainly dendrite-targeting neurons. In general, the IN subpopulations in the monkey are not dissimilar to those seen in rodents but, unlike rodents, CB+ INs in the monkey are not basket cells.
Collapse
Affiliation(s)
- Alexander J McDonald
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA.
| |
Collapse
|
38
|
Simonetti M, Paldy E, Njoo C, Bali KK, Worzfeld T, Pitzer C, Kuner T, Offermanns S, Mauceri D, Kuner R. The impact of Semaphorin 4C/Plexin-B2 signaling on fear memory via remodeling of neuronal and synaptic morphology. Mol Psychiatry 2021; 26:1376-1398. [PMID: 31444474 PMCID: PMC7985029 DOI: 10.1038/s41380-019-0491-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/20/2019] [Accepted: 06/03/2019] [Indexed: 12/19/2022]
Abstract
Aberrant fear is a cornerstone of several psychiatric disorders. Consequently, there is large interest in elucidation of signaling mechanisms that link extracellular cues to changes in neuronal function and structure in brain pathways that are important in the generation and maintenance of fear memory and its behavioral expression. Members of the Plexin-B family of receptors for class 4 semaphorins play important roles in developmental plasticity of neurons, and their expression persists in some areas of the adult nervous system. Here, we aimed to elucidate the role of Semaphorin 4C (Sema4C) and its cognate receptor, Plexin-B2, in the expression of contextual and cued fear memory, setting a mechanistic focus on structural plasticity and exploration of contributing signaling pathways. We observed that Plexin-B2 and Sema4C are expressed in forebrain areas related to fear memory, such as the anterior cingulate cortex, amygdala and the hippocampus, and their expression is regulated by aversive stimuli that induce fear memory. By generating forebrain-specific Plexin-B2 knockout mice and analyzing fear-related behaviors, we demonstrate that Sema4C-PlexinB2 signaling plays a crucial functional role in the recent and remote recall of fear memory. Detailed neuronal morphological analyses revealed that Sema4C-PlexinB2 signaling largely mediates fear-induced structural plasticity by enhancing dendritic ramifications and modulating synaptic density in the adult hippocampus. Analyses on signaling-related mutant mice showed that these functions are mediated by PlexinB2-dependent RhoA activation. These results deliver important insights into the mechanistic understanding of maladaptive plasticity in fear circuits and have implications for novel therapeutic strategies against fear-related disorders.
Collapse
Affiliation(s)
- Manuela Simonetti
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Eszter Paldy
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Christian Njoo
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Kiran Kumar Bali
- grid.7700.00000 0001 2190 4373Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Thomas Worzfeld
- grid.10253.350000 0004 1936 9756Institute of Pharmacology, Marburg University, Karl-von-Frisch-Str. 1, 35043 Marburg, Germany ,grid.418032.c0000 0004 0491 220XDepartment of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Claudia Pitzer
- grid.7700.00000 0001 2190 4373Interdisciplinary Neurobehavioral Core, Heidelberg University, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Thomas Kuner
- grid.7700.00000 0001 2190 4373Anatomy and Cell Biology Institute, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Stefan Offermanns
- grid.418032.c0000 0004 0491 220XDepartment of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Daniela Mauceri
- grid.7700.00000 0001 2190 4373Department of Neurobiology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
39
|
Saha R, Kriebel M, Anunu R, Volkmer H, Richter-Levin G. Intra-amygdala metaplasticity modulation of fear extinction learning. Eur J Neurosci 2020; 55:2455-2463. [PMID: 33305403 DOI: 10.1111/ejn.15080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/22/2022]
Abstract
The amygdala is a key brain region involved in emotional memory formation. It is also responsible for memory modulation in other brain areas. Under extreme conditions, amygdala modulation may lead to the generation of abnormal plasticity and trauma-related psychopathologies. However, the amygdala itself is a dynamic brain region, which is amenable to long-term plasticity and is affected by emotional experiences. These alterations may modify the way the amygdala modulates activity and plasticity in other related brain regions, which in turn may alter the animal's response to subsequent challenges in what could be termed as "Behavioral metaplasticity."Because of the reciprocal interactions between the amygdala and other emotion processing regions, such as the medial prefrontal cortex (mPFC) or the hippocampus, experience-induced intra-amygdala metaplasticity could lead to alterations in mPFC-dependent or hippocampus-dependent behaviors. While initiated by alterations within the basolateral amygdala (BLA), such alterations in other brain regions may come to be independent of BLA modulation, thus establishing what may be termed "Trans-regional metaplasticity." In this article, we review evidence supporting the notions of intra-BLA metaplasticity and how this may develop into "Trans-regional metaplasticity." Future research is needed to understand how such dynamic metaplastic alterations contribute to developing psychopathologies, and how this knowledge may be translated into promoting novel interventions in psychopathologies associated with fear, stress, and trauma.
Collapse
Affiliation(s)
- Rinki Saha
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Martin Kriebel
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | - Rachel Anunu
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Hansjuergen Volkmer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Tübingen, Germany
| | - Gal Richter-Levin
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel.,Department of Psychology, University of Haifa, Haifa, Israel.,The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa, Israel
| |
Collapse
|
40
|
Desmarchelier MR. Clinical Psychopharmacology for the Exotic Animal Practitioner. Vet Clin North Am Exot Anim Pract 2020; 24:17-35. [PMID: 33189249 DOI: 10.1016/j.cvex.2020.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Literature regarding the clinical use of psychotropic drugs in exotic animals remains scarce. Psychotropic drugs acting on serotonin, dopamine, norepinephrine, and gamma-aminobutyric acid pathways work by decreasing fear and anxiety, reactivity, and hypervigilance, and by improving impulse control. They are indicated for some cases of aggression, self-mutilation, and compulsive and anxiety disorders, including feather-damaging behavior. Side effects are rarely seen when dosages are appropriately adjusted to the individual, starting with a low dose and slowly titrating to effect. Several drug interactions exist between psychotropic drugs and other classes. Psychotropic drugs cannot be used to replace appropriate environmental conditions in exotic animals. before "Side effects".
Collapse
Affiliation(s)
- Marion R Desmarchelier
- Department of Clinical Sciences, Faculté de médecine vétérinaire, Université de Montréal, 3200 rue Sicotte, Saint-Hyacinthe, Québec J2S 2M2, Canada.
| |
Collapse
|
41
|
Mattera A, Pagani M, Baldassarre G. A Computational Model Integrating Multiple Phenomena on Cued Fear Conditioning, Extinction, and Reinstatement. Front Syst Neurosci 2020; 14:569108. [PMID: 33132856 PMCID: PMC7550679 DOI: 10.3389/fnsys.2020.569108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/13/2020] [Indexed: 11/23/2022] Open
Abstract
Conditioning, extinction, and reinstatement are fundamental learning processes of animal adaptation, also strongly involved in human pathologies such as post-traumatic stress disorder, anxiety, depression, and dependencies. Cued fear conditioning, extinction, restatement, and systematic manipulations of the underlying brain amygdala and medial prefrontal cortex, represent key experimental paradigms to study such processes. Numerous empirical studies have revealed several aspects and the neural systems and plasticity underlying them, but at the moment we lack a comprehensive view. Here we propose a computational model based on firing rate leaky units that contributes to such integration by accounting for 25 different experiments on fear conditioning, extinction, and restatement, on the basis of a single neural architecture having a structure and plasticity grounded in known brain biology. This allows the model to furnish three novel contributions to understand these open issues: (a) the functioning of the central and lateral amygdala system supporting conditioning; (b) the role played by the endocannabinoids system in within- and between-session extinction; (c) the formation of three important types of neurons underlying fear processing, namely fear, extinction, and persistent neurons. The model integration of the results on fear conditioning goes substantially beyond what was done in previous models.
Collapse
Affiliation(s)
- Andrea Mattera
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Marco Pagani
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Gianluca Baldassarre
- Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
42
|
Neurotrophin signalling in amygdala-dependent cued fear learning. Cell Tissue Res 2020; 382:161-172. [PMID: 32845430 PMCID: PMC7529623 DOI: 10.1007/s00441-020-03260-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022]
Abstract
The amygdala is a central hub for fear learning assessed by Pavlovian fear conditioning. Indeed, the prevailing hypothesis that learning and memory are mediated by changes in synaptic strength was shown most convincingly at thalamic and cortical afferents to the lateral amygdala. The neurotrophin brain-derived neurotrophic factor (BDNF) is known to regulate synaptic plasticity and memory formation in many areas of the mammalian brain including the amygdala, where BDNF signalling via tropomyosin-related kinase B (TrkB) receptors is prominently involved in fear learning. This review updates the current understanding of BDNF/TrkB signalling in the amygdala related to fear learning and extinction. In addition, actions of proBDNF/p75NTR and NGF/TrkA as well as NT-3/TrkC signalling in the amygdala are introduced.
Collapse
|
43
|
Abstract
Brain-wide circuits that coordinate affective and social behaviours intersect in the amygdala. Consequently, amygdala lesions cause a heterogeneous array of social and non-social deficits. Social behaviours are not localized to subdivisions of the amygdala even though the inputs and outputs that carry social signals are anatomically restricted to distinct subnuclear regions. This observation may be explained by the multidimensional response properties of the component neurons. Indeed, the multitudes of circuits that converge in the amygdala enlist the same subset of neurons into different ensembles that combine social and non-social elements into high-dimensional representations. These representations may enable flexible, context-dependent social decisions. As such, multidimensional processing may operate in parallel with subcircuits of genetically identical neurons that serve specialized and functionally dissociable functions. When combined, the activity of specialized circuits may grant specificity to social behaviours, whereas multidimensional processing facilitates the flexibility and nuance needed for complex social behaviour.
Collapse
|
44
|
Manzano Nieves G, Bravo M, Baskoylu S, Bath KG. Early life adversity decreases pre-adolescent fear expression by accelerating amygdala PV cell development. eLife 2020; 9:55263. [PMID: 32692310 PMCID: PMC7413666 DOI: 10.7554/elife.55263] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Early life adversity (ELA) is associated with increased risk for stress-related disorders later in life. The link between ELA and risk for psychopathology is well established but the developmental mechanisms remain unclear. Using a mouse model of resource insecurity, limited bedding (LB), we tested the effects of LB on the development of fear learning and neuronal structures involved in emotional regulation, the medial prefrontal cortex (mPFC) and basolateral amygdala (BLA). LB delayed the ability of peri-weanling (21 days old) mice to express, but not form, an auditory conditioned fear memory. LB accelerated the developmental emergence of parvalbumin (PV)-positive cells in the BLA and increased anatomical connections between PL and BLA. Fear expression in LB mice was rescued through optogenetic inactivation of PV-positive cells in the BLA. The current results provide a model of transiently blunted emotional reactivity in early development, with latent fear-associated memories emerging later in adolescence.
Collapse
Affiliation(s)
| | - Marilyn Bravo
- Department of Neuroscience, Brown University, Providence, United States
| | - Saba Baskoylu
- Department of Neuroscience, Brown University, Providence, United States
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, United States
| |
Collapse
|
45
|
Abstract
Cortical interneurons display striking differences in shape, physiology, and other attributes, challenging us to appropriately classify them. We previously suggested that interneuron types should be defined by their role in cortical processing. Here, we revisit the question of how to codify their diversity based upon their division of labor and function as controllers of cortical information flow. We suggest that developmental trajectories provide a guide for appreciating interneuron diversity and argue that subtype identity is generated using a configurational (rather than combinatorial) code of transcription factors that produce attractor states in the underlying gene regulatory network. We present our updated three-stage model for interneuron specification: an initial cardinal step, allocating interneurons into a few major classes, followed by definitive refinement, creating subclasses upon settling within the cortex, and lastly, state determination, reflecting the incorporation of interneurons into functional circuit ensembles. We close by discussing findings indicating that major interneuron classes are both evolutionarily ancient and conserved. We propose that the complexity of cortical circuits is generated by phylogenetically old interneuron types, complemented by an evolutionary increase in principal neuron diversity. This suggests that a natural neurobiological definition of interneuron types might be derived from a match between their developmental origin and computational function.
Collapse
Affiliation(s)
- Gord Fishell
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA;
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02142, USA
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Adam Kepecs
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri 63130, USA;
| |
Collapse
|
46
|
Luo L, Sun T, Yang L, Liu A, Liu QQ, Tian QQ, Wang Y, Zhao MG, Yang Q. Scopoletin ameliorates anxiety-like behaviors in complete Freund's adjuvant-induced mouse model. Mol Brain 2020; 13:15. [PMID: 32019580 PMCID: PMC7001522 DOI: 10.1186/s13041-020-0560-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/23/2020] [Indexed: 12/20/2022] Open
Abstract
Anxiety disorder is highly prevalent worldwide and represents a chronic and functionally disabling condition, with high levels of psychological stress characterized by cognitive and physiological symptoms. Scopoletin (SP), a main active compound in Angelica dahurica, is traditionally used for the treatment of headache, rhinitis, pain, and other conditions. Here, we evaluated the effects of SP in a mouse model of complete Freund’s adjuvant (CFA)-induced chronic inflammation anxiety. SP (2.0, 10.0, 50.0 mg/kg) administration for 2 weeks dose-dependently ameliorated CFA-induced anxiety-like behaviors in the open field test and elevated plus maze test. Moreover, we found that SP treatment inhibited microglia activation and decreased both peripheral and central IL-1β, IL-6, and TNF-α levels in a dose-dependent manner. Additionally, the imbalance in excitatory/inhibitory receptors and neurotransmitters in the basolateral nucleus after CFA injection was also modulated by SP administration. Our findings indicate that the inhibition of the nuclear factor-kappa B and mitogen-activated protein kinase signaling pathways involving anti-inflammatory activities and regulation of the excitatory/inhibitory balance can be attributed to the anxiolytic effects of SP. Moreover, our molecular docking analyses show that SP also has good affinity for gamma-aminobutyric acid (GABA) transaminase and GABAA receptors. Therefore, these results suggest that SP could be a candidate compound for anxiolytic therapy and for use as a structural base for developing new drugs.
Collapse
Affiliation(s)
- Li Luo
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Ting Sun
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - An Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Qing-Qing Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Qin-Qin Tian
- Department of Chemistry, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yan Wang
- Department of Gastroenterology and Endoscopy Center, No. 986 Hospital, Fourth Military Medical University, Xi'an, 710054, China
| | - Ming-Gao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Qi Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
47
|
Abstract
Fear is a response to impending threat that prepares a subject to make appropriate defensive responses, whether to freeze, fight, or flee to safety. The neural circuits that underpin how subjects learn about cues that signal threat, and make defensive responses, have been studied using Pavlovian fear conditioning in laboratory rodents as well as humans. These studies have established the amygdala as a key player in the circuits that process fear and led to a model where fear learning results from long-term potentiation of inputs that convey information about the conditioned stimulus to the amygdala. In this review, we describe the circuits in the basolateral amygdala that mediate fear learning and its expression as the conditioned response. We argue that while the evidence linking synaptic plasticity in the basolateral amygdala to fear learning is strong, there is still no mechanism that fully explains the changes that underpin fear conditioning.
Collapse
Affiliation(s)
- Yajie Sun
- Queensland Brain Institute, University of Queensland, Queensland, Australia
| | - Helen Gooch
- Queensland Brain Institute, University of Queensland, Queensland, Australia
| | - Pankaj Sah
- Queensland Brain Institute, University of Queensland, Queensland, Australia.,Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
48
|
Polepalli JS, Gooch H, Sah P. Diversity of interneurons in the lateral and basal amygdala. NPJ SCIENCE OF LEARNING 2020; 5:10. [PMID: 32802405 PMCID: PMC7400739 DOI: 10.1038/s41539-020-0071-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/29/2020] [Indexed: 05/06/2023]
Abstract
The basolateral amygdala (BLA) is a temporal lobe structure that contributes to a host of behaviors. In particular, it is a central player in learning about aversive events and thus assigning emotional valence to sensory events. It is a cortical-like structure and contains glutamatergic pyramidal neurons and GABAergic interneurons. It is divided into the lateral (LA) and basal (BA) nuclei that have distinct cell types and connections. Interneurons in the BLA are a heterogenous population, some of which have been implicated in specific functional roles. Here we use optogenetics and slice electrophysiology to investigate the innervation, postsynaptic receptor stoichiometry, and plasticity of excitatory inputs onto interneurons within the BLA. Interneurons were divided into six groups based on their discharge properties, each of which received input from the auditory thalamus (AT) and auditory cortex (AC). Auditory innervation was concentrated in the LA, and optogenetic stimulation evoked robust synaptic responses in nearly all interneurons, drove many cells to threshold, and evoked disynaptic inhibition in most interneurons. Auditory input to the BA was sparse, innervated fewer interneurons, and evoked smaller synaptic responses. Biophysically, the subunit composition and distribution of AMPAR and NMDAR also differed between the two nuclei, with fewer BA IN expressing calcium permeable AMPAR, and a higher proportion expressing GluN2B-containing NMDAR. Finally, unlike LA interneurons, LTP could not be induced in the BA. These findings show that interneurons in the LA and BA are physiologically distinct populations and suggest they may have differing roles during associative learning.
Collapse
Affiliation(s)
- Jai S. Polepalli
- Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072 Australia
- Department of Anatomy, Yong Yoo Lin School of Medicine, National University of Singapore, Singapore, 117594 Singapore
| | - Helen Gooch
- Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072 Australia
| | - Pankaj Sah
- Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072 Australia
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Nanshan District, Shenzhen, Guangdong Province P.R. China
| |
Collapse
|
49
|
Nash B, Festa L, Lin C, Meucci O. Opioid and chemokine regulation of cortical synaptodendritic damage in HIV-associated neurocognitive disorders. Brain Res 2019; 1723:146409. [PMID: 31465771 PMCID: PMC6766413 DOI: 10.1016/j.brainres.2019.146409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/20/2019] [Accepted: 08/25/2019] [Indexed: 01/17/2023]
Abstract
Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) persist despite effective antiretroviral therapies (ART). Evidence suggests that modern HAND is driven by subtle synaptodendritic damage in select brain regions, as ART-treated patients do not display overt neuronal death in postmortem brain studies. HAND symptoms are also aggravated by drug abuse, particularly with injection opioids. Opioid use produces region-specific synaptodendritic damage in similar brain regions, suggesting a convergent mechanism that may enhance HAND progression in opioid-using patients. Importantly, studies indicate that synaptodendritic damage and cognitive impairment in HAND may be reversible. Activation of the homeostatic chemokine receptor CXCR4 by its natural ligand CXCL12 positively regulates neuronal survival and dendritic spine density in cortical neurons, reducing functional deficits. However, the molecular mechanisms that underlie CXCR4, as well as opioid-mediated regulation of dendritic spines are not completely defined. Here, we will consolidate studies that describe the region-specific synaptodendritic damage in the cerebral cortex of patients and animal models of HAND, describe the pathways by which opioids may contribute to cortical synaptodendritic damage, and discuss the prospects of using the CXCR4 signaling pathway to identify new approaches to reverse dendritic spine deficits. Additionally, we will discuss novel research questions that have emerged from recent studies of CXCR4 and µ-opioid actions in the cortex. Understanding the pathways that underlie synaptodendritic damage and rescue are necessary for developing novel, effective therapeutics for this growing patient population.
Collapse
Affiliation(s)
- Bradley Nash
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Lindsay Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Chihyang Lin
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA; Department of Microbiology and Immunology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA.
| |
Collapse
|
50
|
Inhibitory microcircuits for top-down plasticity of sensory representations. Nat Commun 2019; 10:5055. [PMID: 31699994 PMCID: PMC6838080 DOI: 10.1038/s41467-019-12972-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 10/11/2019] [Indexed: 01/06/2023] Open
Abstract
Rewards influence plasticity of early sensory representations, but the underlying changes in circuitry are unclear. Recent experimental findings suggest that inhibitory circuits regulate learning. In addition, inhibitory neurons are highly modulated by diverse long-range inputs, including reward signals. We, therefore, hypothesise that inhibitory plasticity plays a major role in adjusting stimulus representations. We investigate how top-down modulation by rewards interacts with local plasticity to induce long-lasting changes in circuitry. Using a computational model of layer 2/3 primary visual cortex, we demonstrate how interneuron circuits can store information about rewarded stimuli to instruct long-term changes in excitatory connectivity in the absence of further reward. In our model, stimulus-tuned somatostatin-positive interneurons develop strong connections to parvalbumin-positive interneurons during reward such that they selectively disinhibit the pyramidal layer henceforth. This triggers excitatory plasticity, leading to increased stimulus representation. We make specific testable predictions and show that this two-stage model allows for translation invariance of the learned representation. Rewards can improve stimulus processing in early sensory areas but the underlying neural circuit mechanisms are unknown. Here, the authors build a computational model of layer 2/3 primary visual cortex and suggest that plastic inhibitory circuits change first and then increase excitatory representations beyond the presence of rewards.
Collapse
|