1
|
Friuli M, Eramo B, Sepe C, Kiani M, Casolini P, Zuena AR. The endocannabinoid and paracannabinoid systems in natural reward processes: possible pharmacological targets? Physiol Behav 2025; 296:114929. [PMID: 40274041 DOI: 10.1016/j.physbeh.2025.114929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Natural rewards such as food, mating, and social interaction are essential for survival and species preservation, and their regulation involves a complex interplay of motivational, cognitive, and emotional processes. Over the past two decades, increasing attention has been directed toward the endocannabinoid system and its paracannabinoid counterpart as key modulators of these behaviors. This review aims to provide an integrated overview of the roles played by the endocannabinoid and paracannabinoid systems in regulating natural reward-driven behaviors, focusing on feeding, reproductive behavior, and social interaction. We highlight how the endocannabinoid system - mainly through CB1 receptor signaling - modulates central and peripheral circuits involved in energy homeostasis, reward processing, and emotional regulation. In parallel, we explore the role of paracannabinoids, such as oleoylethanolamide (OEA), palmitoylethanolamide (PEA), and stearoylethanolamide (SEA), which act primarily via non-cannabinoid receptors and contribute to the regulation of appetite, sexual motivation, and social behavior. Special attention is given to the relevance of these systems in the pathophysiology of obesity, eating disorders, sexual dysfunctions, and social impairments, as well as their potential as pharmacological targets. Overall, the evidence discussed supports a broader conceptualization of endocannabinoid and paracannabinoid signaling as pivotal regulators of natural rewards and opens new avenues for the development of targeted interventions for motivational and reward-related disorders.
Collapse
Affiliation(s)
- Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy.
| | - Barbara Eramo
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Christian Sepe
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Mitra Kiani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy; Department of Pharmacology & Experimental Therapeutics, School of Medicine, Boston University, Boston, MA 02118, USA
| | - Paola Casolini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Anna Rita Zuena
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
2
|
Radzicki D, McCann KE, Alexander GM, Dudek SM. Hippocampal area CA2 activity supports social investigation following an acute social stress. Mol Psychiatry 2025; 30:2284-2296. [PMID: 39548322 DOI: 10.1038/s41380-024-02834-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Neuronal activity in the hippocampus is critical for many types of memory acquisition and retrieval and influences an animal's response to stress. Moreover, the molecularly distinct principal neurons of hippocampal area CA2 are required for social recognition memory and aggression in mice. To interrogate the effects of stress on CA2-dependent behaviors, we chemogenetically manipulated neuronal activity in vivo during an acute, socially derived stressor and tested whether memory for the defeat was influenced. One day after an acute social defeat (aSD), defeated mice spent significantly less time investigating another mouse when compared to non-defeated control mice. We found that this avoidant phenotype persisted for up to one month following a single defeat encounter. When CA2 pyramidal neuron activity was inhibited with Gi-DREADD receptors during the defeat, subject mice exhibited a significantly higher amount of social avoidance one day later when compared to defeated littermates not expressing DREADDs. Moreover, CA2 inhibition during defeat caused a reduction in submissive defense behaviors in response to aggression. In vitro electrophysiology and tracing experiments revealed a circuit wherein CA2 neurons connect to caudal CA1 projection neurons that, in turn, project to corticolimbic regions including the anterior cingulate cortex. Finally, socially avoidant, defeated mice exhibited significant reductions in cFos expression in caudal hippocampal and limbic brain areas during a social investigation task 24 h after aSD. Taken together, these results indicate that CA2 neuronal activity is required to support behavioral resilience following an acute social stressor and that submissive defensive behavior during the defeat (vs. fleeing) is a predictor of future resilience to social stress. Furthermore, CA2 preferentially targets a population of caudal CA1 projection neurons that contact cortical brain regions where activity is modulated by an acute social stressor.
Collapse
Affiliation(s)
- Daniel Radzicki
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, 27713, USA
| | - Katharine E McCann
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, 27713, USA
- Neuroscience Undergraduate Program and School of Psychology, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, 27713, USA
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institute of Health, Research Triangle Park, NC, 27713, USA.
| |
Collapse
|
3
|
Alemán-Andrade P, Witter MP, Tsutsui KI, Ohara S. Dorsal-Caudal and Ventral Hippocampi Target Different Cell Populations in the Medial Frontal Cortex in Rodents. J Neurosci 2025; 45:e0217252025. [PMID: 40204437 PMCID: PMC12121713 DOI: 10.1523/jneurosci.0217-25.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025] Open
Abstract
Direct projections from the ventral hippocampus (vHPC) to the medial frontal cortex (MFC) play crucial roles in memory and emotional regulation. Using anterograde transsynaptic tracing and ex vivo electrophysiology in male mice, we document a previously unexplored pathway that parallels the established vHPC-MFC connectivity. This pathway connects the dorsal-caudal hippocampus (dcHPC) to specific subregions of the ventral MFC (vMFC), in particular the dorsal peduncular cortex. Notably, this pathway exerts a strong inhibitory influence on vMFC by targeting a substantial proportion of inhibitory neurons. Retrograde transsynaptic tracing in male rats indicated that vMFC subregions project disynaptically back to vHPC. These results, altogether, suggest the existence of a remarkable functional circuit connecting distinct functional areas: the cognition-related dcHPC with the emotion-related vMFC and vHPC. These findings further provide valuable insights in the cognitive and emotional abnormalities associated with the HPC-MFC connectivity in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Paola Alemán-Andrade
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8577, Japan
| | - Menno P Witter
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8577, Japan
- Kavli institute for Systems Neuroscience, NTNU Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Ken-Ichiro Tsutsui
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8577, Japan
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai 980-8577, Japan
| | - Shinya Ohara
- Laboratory of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai 980-8577, Japan
| |
Collapse
|
4
|
Dwortz MF, Curley JP. Capturing dynamic neuronal responses to dominant and subordinate social hierarchy members with catFISH. Neuroscience 2025:S0306-4522(25)00387-2. [PMID: 40414523 DOI: 10.1016/j.neuroscience.2025.05.025] [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: 12/19/2024] [Revised: 03/31/2025] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
Dominance hierarchies are key to social organization in group-living species, requiring individuals to recognize their own and others' ranks. This is particularly complex for mid-ranking animals, who navigate interactions with higher- and lower-ranking individuals. Using in situ hybridization, we examined how mid-ranked mice's brains respond to dominant and subordinate stimuli by labeling activity-induced immediate early genes and neuronal markers. We show that distinct neuronal populations in the amygdala and hippocampus respond differentially across social contexts. In the basolateral amygdala and dorsal endopiriform, glutamatergic Slc17a7+ neurons, particularly dopamine-receptive Slc17a7+Drd1+ neurons, show elevated IEG expression in response to social stimuli, with a higher response to dominant over subordinate animals. Similar response patterns are observed among GABAergic Slc32a1+Oxtr+ neurons in the medial amygdala. We also identified distinct neural ensembles selectively active in response to dominant and subordinate animals by examining cell reactivation over repeated stimulus presentations. We find a higher degree of reactivation among Slc17a7+Oxtr+ ensembles in the endopiriform when the same individual was presented twice in succession. A similar pattern was observed among Oxtr+ neurons in the dentate gyrus hilus, while the inverse was observed among Slc17a7+Avrp1b+Oxtr+ neurons in distal CA2CA3, suggesting distinct encoding or recollection mechanisms across hippocampal subregions. We also highlight methodological advances showing that IEG responses are shaped by stimulus duration and the identity of the IEG and timepoint at which expression is measured. This work lays the foundation for further precise, cell type-resolved investigation into how the brain processes social information.
Collapse
Affiliation(s)
- Madeleine F Dwortz
- Interdisciplinary Neuroscience Program, The University of Texas at Austin, Austin, TX 78712, United States; Department of Psychology, The University of Texas at Austin, Austin, TX 78712, United States; Department of Psychiatry and Behavioral Sciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - James P Curley
- Interdisciplinary Neuroscience Program, The University of Texas at Austin, Austin, TX 78712, United States; Department of Psychology, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
5
|
Zorab JM, Li H, Awasthi R, Schinasi A, Cho Y, O'Loughlin T, Wu X. Serotonin and neurotensin inputs in the vCA1 dictate opposing social valence. Nature 2025:10.1038/s41586-025-08809-2. [PMID: 40307550 DOI: 10.1038/s41586-025-08809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/19/2025] [Indexed: 05/02/2025]
Abstract
The ability to evaluate valence of a social agent based on social experience is essential for an animal's survival in its social group1. Although hippocampal circuits have been implicated in distinguishing novel and familiar conspecifics2-7, it remains unclear how social valence is constructed on the basis of social history and what mechanisms underlie the heightened valence versatility in dynamic relationships. Here we demonstrate that the ventral (v)CA1 integrates serotonin (5-HT) inputs from the dorsal raphe and neurotensin inputs from the paraventricular nucleus of the thalamus (PVT) to determine positive or negative valence of conspecific representations. Specifically, during an appetitive social interaction 5-HT is released into the vCA1 and disinhibits pyramidal neurons through 5-HT1B receptors, whereas neurotensin is released during an aversive social interaction and potentiates vCA1 neurons directly through NTR1s. Optogenetic silencing of dorsal raphe 5-HT and PVT neurotensin inputs into the vCA1 impairs positive and negative social valence, respectively, and excitation flexibly switches valence assignment. These results show how aversive and rewarding social experiences are linked to conspecific identity through converging dorsal raphe 5-HT and PVT neurotensin signals in the vCA1 that instruct opposing valence, and represent a synaptic switch for flexible social valence computation.
Collapse
Affiliation(s)
- Julia M Zorab
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Huanhuan Li
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Richa Awasthi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Schinasi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yoonjeong Cho
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas O'Loughlin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaoting Wu
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Michael A, Onisiforou A, Georgiou P, Koumas M, Powels C, Mammadov E, Georgiou AN, Zanos P. (2R,6R)-hydroxynorketamine prevents opioid abstinence-related negative affect and stress-induced reinstatement in mice. Br J Pharmacol 2025. [PMID: 40155780 DOI: 10.1111/bph.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 02/05/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND AND PURPOSE Opioid use disorder (OUD) is a pressing public health concern marked by frequent relapse during periods of abstinence, perpetuated by negative affective states. Classical antidepressants or the currently prescribed opioid pharmacotherapies have limited efficacy to reverse the negative affect or prevent relapse. EXPERIMENTAL APPROACH Using mouse models, we investigated the effects of ketamine's metabolite (2R,6R)-hydroxynorketamine (HNK) on reversing conditioning to sub-effective doses of morphine in stress-susceptible mice, preventing conditioned-place aversion and alleviating acute somatic abstinence symptoms in opioid-dependent mice. Additionally, we evaluated its effects on anhedonia, anxiety-like behaviours and cognitive impairment during protracted opioid abstinence, while mechanistic studies examined cortical EEG oscillations and synaptic plasticity markers. KEY RESULTS (2R,6R)-HNK reversed conditioning to sub-effective doses of morphine in stress-susceptible mice and prevented conditioned-place aversion and acute somatic abstinence symptoms in opioid-dependent mice. In addition, (2R,6R)-HNK reversed anhedonia, anxiety-like behaviours and cognitive impairment emerging during protracted opioid abstinence plausibly via a restoration of impaired cortical high-frequency EEG oscillations, through a GluN2A-NMDA receptor-dependent mechanism. Notably, (2R,6R)-HNK facilitated the extinction of opioid conditioning, prevented stress-induced reinstatement of opioid-seeking behaviours and reduced the propensity for enhanced morphine self-consumption in mice previously exposed to opioids. CONCLUSIONS AND IMPLICATIONS These findings emphasize the therapeutic potential of (2R,6R)-HNK, which is currently in Phase II clinical trials, in addressing stress-related opioid responses. Reducing the time and cost required for development of new medications for the treatment of OUDs via drug repurposing is critical due to the opioid crisis we currently face.
Collapse
Affiliation(s)
- Andria Michael
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Anna Onisiforou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Morfeas Koumas
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chris Powels
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elmar Mammadov
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Andrea N Georgiou
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Center for Applied Neuroscience (CAN), University of Cyprus, Nicosia, Cyprus
- Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Di Re J, Koff L, Avchalumov Y, Singh AK, Baumgartner TJ, Marosi M, Matz LM, Hallberg LM, Ameredes BT, Seeley EH, Buffington SA, Green TA, Laezza F. Environmental exposure to common pesticide induces synaptic deficit and social memory impairment driven by neurodevelopmental vulnerability of hippocampal parvalbumin interneurons. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136893. [PMID: 39706027 PMCID: PMC11970102 DOI: 10.1016/j.jhazmat.2024.136893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Environmental exposure to pesticides at levels deemed safe by regulatory agencies has been linked to increased risk for neurodevelopmental disorders. Yet, the mechanisms linking exposure to these disorders remain unclear. Here, we show that maternal exposure to the pesticide deltamethrin (DM) at the no observed adverse effect level (NOAEL) disrupts long-term potentiation (LTP) in the hippocampus of adult male offspring three months after exposure, a phenotype absent in female offspring. Clonazepam, a GABAa receptor agonist, rescued this deficit, indicating impaired hippocampal GABAergic signaling. Recordings from CA1 pyramidal neurons, complemented by MALDI mass spectrometry imaging, showed an imbalance in excitatory/inhibitory tone. Using a combination of parvalbumin (PV)-Cre transgenic mice and hippocampal injection of designer receptors exclusively activated by designer drugs (DREADDs), we show that developmental DM exposure reduces hippocampal PV interneuron intrinsic firing. DREADD activation rescued both PV interneuron firing and LTP deficits. Complementary behavioral experiments revealed a deficit in social memory, a behavior relevant to autism spectrum disorder (ASD) symptomatology, which was restored by DREADD activation. Overall, these results establish a novel mechanistic link between maternal exposure to DM at the NOAEL and known cellular, circuital, and behavioral vulnerabilities, indicating it is a potential driver in the exposome of ASD.
Collapse
Affiliation(s)
- Jessica Di Re
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; NIEHS Environmental Toxicology Training Program, University of Texas Medical Branch, USA
| | - Leandra Koff
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yosef Avchalumov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Timothy J Baumgartner
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mate Marosi
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Lisa M Matz
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lance M Hallberg
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Inhalation Toxicology Core, University of Texas Medical Branch, USA
| | - Bill T Ameredes
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Inhalation Toxicology Core, University of Texas Medical Branch, USA
| | - Erin H Seeley
- Department of Chemistry, University of Texas, Austin, TX 78712, USA
| | - Shelly A Buffington
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA; Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
8
|
Grieco F, Balla A, Larrieu T, Toni N. Natural variations of adolescent neurogenesis and anxiety predict the hierarchical status of adult inbred mice. EMBO Rep 2025; 26:1440-1456. [PMID: 39849205 PMCID: PMC11933688 DOI: 10.1038/s44319-025-00367-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/25/2025] Open
Abstract
Hierarchy provides a survival advantage to social animals in challenging circumstances. In mice, social dominance is associated with trait anxiety which is regulated by adult hippocampal neurogenesis. Here, we test whether adolescent hippocampal neurogenesis may regulate social dominance behavior in adulthood. We observe that adolescent individuals with higher trait anxiety and lower levels of hippocampal neurogenesis prior to the formation of a new group become dominants, suggesting that baseline adolescent neurogenesis predicts hierarchical status. This phenotype persists beyond social hierarchy stabilization. Experimentally reducing neurogenesis prior to the stabilization of social hierarchy in group-housed adolescent males increases the probability of mice to become dominant and increases anxiety. Finally, when innate dominance is assessed in socially isolated and anxiety-matched animals, mice with impaired neurogenesis display a dominant status toward strangers. Together, these results indicate that adolescent neurogenesis predicts and regulates hierarchical and situational dominance behavior along with anxiety-related behavior. These results provide a framework to study the mechanisms underlying social hierarchy and the dysregulation of dominance behavior in psychiatric diseases related to anxiety.
Collapse
Affiliation(s)
- Fabio Grieco
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Prilly, Switzerland
| | - Atik Balla
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Prilly, Switzerland
| | - Thomas Larrieu
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Prilly, Switzerland.
| | - Nicolas Toni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Prilly, Switzerland.
| |
Collapse
|
9
|
Liu YC, Deng YC, Zhu ZT, Rao B, Shang HL, Wang LK, Li T, Wang YR, Wang JZ, Zhang QP, Gao Y, Xu HB. Oxytocin modulates inhibitory balance in the prelimbic cortex to support social memory consolidation during REM sleep. Theranostics 2025; 15:3257-3274. [PMID: 40093885 PMCID: PMC11905142 DOI: 10.7150/thno.109104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/26/2025] [Indexed: 03/19/2025] Open
Abstract
Rationale: The prelimbic cortex (PrL), enriched with oxytocin (OXT) receptors, plays a critical role in memory consolidation. However, the role of OXT in social memory consolidation within the PrL microcircuit remains poorly understood. Methods: To examine the role of OXT signaling in social memory consolidation, we used OXT biosensors and loss-of-function approaches, including tetanus toxin-mediated silencing of OXT neurons in the paraventricular nucleus (PVNOXT), optogenetic inhibition of the PVNOXT-PrL pathway during rapid-eye-movement (REM) sleep, and local administration of an OXT receptor antagonist in the PrL. In vivo molecular biosensors for vasoactive intestinal peptide (VIP), somatostatin, and presynaptic calcium imaging were employed to assess inhibitory signaling in the PrL microcircuit. Optogenetic activation of the PVNOXT-PrL pathway and intranasal OXT were used to evaluate resilience to chronic sleep deprivation-induced social memory deficits. Results: We identified that REM-sleep OXT release via the PVN to PrL pathway supports social memory consolidation. OXT signaling deficiency reduces the activity of VIP and parvalbumin (PV) neurons, thereby disrupting the inhibitory balance between somatic inhibition mediated by PV neurons and dendritic disinhibition mediated by VIP neurons in PrL microcircuits during REM sleep. Chronic sleep deprivation (SD) disrupts OXT release and inhibitory balance, leading to pyramidal neuron hyperactivity and social memory impairments. Notably, REM-sleep-specific activation of the PVNOXT-PrL pathway or intranasal OXT restores inhibitory balance and rescues social memory deficits in SD mice. Conclusion: Our results reveal how OXT modulates inhibitory balance in the PrL microcircuit to support social memory consolidation during REM sleep, suggesting potential therapeutic strategies for treating sleep-related memory disorders.
Collapse
Affiliation(s)
- Yan-chao Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yu-chen Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Zi-tao Zhu
- Second Clinical College, Wuhan University, Wuhan, 430071, China
| | - Bo Rao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Hong-lei Shang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Li-ke Wang
- Second Clinical College, Wuhan University, Wuhan, 430071, China
| | - Tao Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Ya-rong Wang
- Li-Yuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing-ping Zhang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yang Gao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Hai-bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Multimodal Medical Imaging Technology and Clinical Application, Wuhan, 430071, China
- Wuhan clinical research and development center of brain resuscitation and functional imaging, Wuhan, 430071, China
| |
Collapse
|
10
|
Illescas-Huerta E, Padilla-Coreano N. Opposing and segregated cortical circuits control winning and losing behaviors. Neuron 2025; 113:335-336. [PMID: 39914363 DOI: 10.1016/j.neuron.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 05/07/2025]
Abstract
In this issue of Neuron, Xin et al.1 reveal how the dorsomedial prefrontal cortex (dmPFC) orchestrates social dominance through subcortical pathways to the amygdala and brainstem. Using optogenetics and functional mapping, they identify opposing win- and lose-related circuits, uncovering a laminar organization driving competitive behavior in mice.
Collapse
|
11
|
Vantomme G, Devienne G, Hull JM, Huguenard JR. Reuniens thalamus recruits recurrent excitation in medial prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.31.596906. [PMID: 38854099 PMCID: PMC11160760 DOI: 10.1101/2024.05.31.596906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Medial prefrontal cortex (mPFC) and hippocampus are critical for memory retrieval, decision making and emotional regulation. While ventral CA1 (vCA1) shows direct and reciprocal connections with mPFC, dorsal CA1 (dCA1) forms indirect pathways to mPFC, notably via the thalamic Reuniens nucleus (Re). Neuroanatomical tracing has documented structural connectivity of this indirect pathway through Re however, its functional operation is largely unexplored. Here we used in vivo and in vitro electrophysiology along with optogenetics to address this question. Whole-cell patch-clamp recordings in acute mouse brain slices revealed both monosynaptic excitatory responses and disynaptic feedforward inhibition at Re-mPFC synapses. However, we also identified a novel prolonged excitation of mPFC by Re. These early monosynaptic and late recurrent components are in marked contrast to the primarily feedforward inhibition characteristic of thalamic inputs to neocortex. Local field potential recordings in mPFC brain slices revealed prolonged synaptic activity throughout all cortical lamina upon Re activation, with the late excitation enhanced by blockade of parvalbumin neurons and GABAARs. In vivo Neuropixels recordings in head-fixed awake mice revealed a similar prolonged excitation of mPFC units by Re activation. In summary, Re output produces recurrent feedforward excitation within mPFC suggesting a potent amplification system in the Re-mPFC network. This may facilitate amplification of dCA1->mPFC signals for which Re acts as the primary conduit, as there is little direct connectivity. In addition, the capacity of mPFC neurons to fire bursts of action potentials in response to Re input suggests that these synapses have a high gain. Significance statement The interactions between medial prefrontal cortex and hippocampus are crucial for memory formation and retrieval. Yet, it is still poorly understood how the functional connectivity of direct and indirect pathways underlies these functions. This research explores the synaptic connectivity of the indirect pathway through the Reuniens nucleus of the thalamus using electrophysiological recordings and optogenetic manipulations. The study found that Reuniens stimulation recruits recurrent and long-lasting activity in mPFC - a phenomenon not previously recorded. This recurrent activity might create a temporal window ideal for coincidence detection and be an underlying mechanism for memory formation and retrieval.
Collapse
Affiliation(s)
- Gil Vantomme
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Gabrielle Devienne
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jacob M Hull
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| |
Collapse
|
12
|
Tian X, Russo SJ, Li L. Behavioral Animal Models and Neural-Circuit Framework of Depressive Disorder. Neurosci Bull 2025; 41:272-288. [PMID: 39120643 PMCID: PMC11794861 DOI: 10.1007/s12264-024-01270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Depressive disorder is a chronic, recurring, and potentially life-endangering neuropsychiatric disease. According to a report by the World Health Organization, the global population suffering from depression is experiencing a significant annual increase. Despite its prevalence and considerable impact on people, little is known about its pathogenesis. One major reason is the scarcity of reliable animal models due to the absence of consensus on the pathology and etiology of depression. Furthermore, the neural circuit mechanism of depression induced by various factors is particularly complex. Considering the variability in depressive behavior patterns and neurobiological mechanisms among different animal models of depression, a comparison between the neural circuits of depression induced by various factors is essential for its treatment. In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression, aiming to provide a theoretical basis for depression prevention.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Long Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
13
|
Crist RC, Chehimi SN, Divakaran SS, Montague MJ, Tremblay S, Snyder-Mackler N, Bohlen MO, Chiou KL, Zintel TM, Platt ML, Juul H, Silvestri G, Hayes MR, Kolson DL, Reiner BC. SIV infection induces alterations in gene expression and loss of interneurons in Rhesus Macaque frontal cortex during early systemic infection. Transl Psychiatry 2025; 15:38. [PMID: 39890796 PMCID: PMC11785960 DOI: 10.1038/s41398-025-03261-2] [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: 10/15/2023] [Revised: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025] Open
Abstract
Understanding the neurobiological mechanisms underlying HIV-associated neurocognitive decline in people living with HIV is frequently complicated by an inability to analyze changes across the course of the infection and frequent presence of comorbid psychiatric and substance use disorders. Preclinical non-human primate simian immunodeficiency virus (SIV) models help address these shortcomings. However, SIV studies frequently target protracted endpoints, limiting our understanding of the neuromolecular alterations during the early post-infection window. To begin to address this knowledge gap, we utilized single nuclei transcriptomics to examine frontal cortex samples of rhesus macaques 10- and 20-days post-SIV infection, compared to non-infected controls. We identify and validated a decrease in inhibitory neurons during the early post infection window, representing a potential substrate of longer-term injury and neurocognitive impairment in people living with HIV. Differential expression identified alterations in cellular subtype gene expression that persisted over the 20-day time course and short-lived differences only detected at 10-days post-SIV infection. In silico predicted regulatory mechanisms and dysregulated neural signaling pathways are presented. Analysis of cell-cell interaction networks identify altered signal pathways in the frontal cortex that may represent regional alterations in cell-cell communications. In total, these results identify cell type-specific molecular mechanisms putatively capable of underlying long-term neurocognitive alterations in persons living with HIV.
Collapse
Affiliation(s)
- Richard C Crist
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samar N Chehimi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saurabh S Divakaran
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
| | - Sébastien Tremblay
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry & Neuroscience, Université Laval, Québec, QC, Canada
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Martin O Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Trish M Zintel
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, USA
- Marketing Department, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Halvor Juul
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guido Silvestri
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Druid Hills, GA, USA
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis L Kolson
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Shirokova OM, Kuzmina DM, Zaborskaya OG, Shchelchkova NA, Kozliaeva EV, Korotchenko SA, Pershin VI, Vasilchikov PI, Mukhina IV. The Long-Term Effects of Chronic Unpredictable Mild Stress Experienced During Adolescence Could Vary Depending on Biological Sex. Int J Mol Sci 2025; 26:1251. [PMID: 39941015 PMCID: PMC11818548 DOI: 10.3390/ijms26031251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Sex differences in the neurobiology of responses to chronic stress have been widely discussed but remain poorly understood. We found that chronic unpredictable mild stress (CUMS) experienced during adolescence induced different behavioral patterns in adult males and females. Immunohistochemical analysis of the CA1 field of the dorsal and ventral hippocampus revealed no quantitative or morphological changes in astrocytes in the long term after CUMS. Real-time PCR analysis showed no increase in the expression level of SigmaR1 after CUMS relative to individual housekeeping genes. Analysis of mouse cerebral cortex homogenates showed that IL-1β levels only decreased after CUMS in males. However, the SigmaR1 levels were significantly higher in the CUMS groups than in the control groups in both sexes. It can be concluded that biological sex and age influence the response to CUMS, although not in all cases. Further studies are needed to understand the effects of chronic stress on males and females. This is important because men and women have different risks for stress and mental disorders.
Collapse
Affiliation(s)
- Olesya M. Shirokova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Daria M. Kuzmina
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Olga G. Zaborskaya
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Natalia A. Shchelchkova
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarina Ave., 602022 Nizhny Novgorod, Russia;
- Scientific Center of Genetics and Life Sciences, Sirius University of Science and Technology, Sirius Federal Territory, 354340 Krasnodar, Russia
| | - Elizaveta V. Kozliaeva
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Svetlana A. Korotchenko
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Vladimir I. Pershin
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| | - Petr I. Vasilchikov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarina Ave., 602022 Nizhny Novgorod, Russia;
| | - Irina V. Mukhina
- Federal State Budgetary Educational Institution of Higher Education «Privolzhsky Research Medical University» of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.M.K.); (O.G.Z.); (V.I.P.); (I.V.M.)
| |
Collapse
|
15
|
Li N, He H, Xu C. Mesoscopic connectome enters the new age of single-neuron projectome. Clin Transl Med 2025; 15:e70155. [PMID: 39737752 DOI: 10.1002/ctm2.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Affiliation(s)
- Ning Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Hua He
- Department of Neurosurgery, Third Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Chun Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Dwortz MF, Curley JP. Capturing Dynamic Neuronal Responses to Dominant and Subordinate Social Hierarchy Members with catFISH. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629477. [PMID: 39763757 PMCID: PMC11702762 DOI: 10.1101/2024.12.19.629477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Dominance hierarchies are key to social organization in group-living species, requiring individuals to recognize their own and others' ranks. This is particularly complex for intermediate-ranking animals, who navigate interactions with higher- and lower-ranking individuals. Using in situ hybridization, we examined how the brains of intermediate-ranked mice in hierarchies respond to dominant and subordinate stimuli by labeling activity-induced immediate early genes and neuronal markers. We show that distinct neuronal populations in the amygdala and hippocampus respond differentially across social contexts. In the basolateral amygdala, glutamatergic Slc17a7+ neurons, particularly dopamine-receptive Slc17a7+Drd1+ neurons, show elevated IEG expression in response to social stimuli, with a higher response to dominant over subordinate animals. Similar patterns are observed among Slc17a7+Oxtr+ neurons in the dorsal endopiriform nucleus and GABAergic Slc32a+ neurons in the medial amygdala. We also identified distinct neural ensembles selectively active in response to dominant and subordinate hierarchy members. We find a higher degree of reactivation among Slc17a7+Oxtr+ ensembles in the dorsal endopiriform nucleus in animals repeatedly presented with the same hierarchy member, as opposed to those presented with a dominant and subordinate member. We observe a similar pattern among Oxtr+ neurons in the dentate gyrus hilus, while the inverse is observed among Slc17a7+ Avrp1b+Oxtr+ neurons in the distal CA2CA3 region. Collectively, our findings reveal how social context is associated with activity changes in social, olfactory, and memory systems in the brain at the neuronal cell type level. This work lays the foundation for further precise cell-type investigation into how the brain processes social information.
Collapse
|
17
|
Shivakumar AB, Mehak SF, Jijimon F, Gangadharan G. Extrahippocampal Contributions to Social Memory: The Role of Septal Nuclei. Biol Psychiatry 2024; 96:835-847. [PMID: 38718881 DOI: 10.1016/j.biopsych.2024.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 06/16/2024]
Abstract
Social memory, the ability to recognize and remember individuals within a social group, is crucial for social interactions and relationships. Deficits in social memory have been linked to several neuropsychiatric and neurodegenerative disorders. The hippocampus, especially the circuit that links dorsal CA2 and ventral CA1 neurons, is considered a neural substrate for social memory formation. Recent studies have provided compelling evidence of extrahippocampal contributions to social memory. The septal nuclei, including the medial and lateral septum, make up a basal forebrain region that shares bidirectional neuronal connections with the hippocampus and has recently been identified as critical for social memory. The focus of our review is the neural circuit mechanisms that underlie social memory, with a special emphasis on the septum. We also discuss the social memory dysfunction associated with neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Apoorva Bettagere Shivakumar
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sonam Fathima Mehak
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Feyba Jijimon
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Gireesh Gangadharan
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
18
|
Duarte JM, Nguyen R, Kyprou M, Li K, Milentijevic A, Cerquetella C, Forro T, Ciocchi S. Hippocampal contextualization of social rewards in mice. Nat Commun 2024; 15:9493. [PMID: 39489746 PMCID: PMC11532361 DOI: 10.1038/s41467-024-53866-2] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Acquiring and exploiting memories of rewarding experiences is critical for survival. The spatial environment in which a rewarding stimulus is encountered regulates memory retrieval. The ventral hippocampus (vH) has been implicated in contextual memories involving rewarding stimuli such as food, social cues or drugs. Yet, the neuronal representations and circuits underlying contextual memories of socially rewarding stimuli are poorly understood. Here, using in vivo electrophysiological recordings, in vivo one-photon calcium imaging, and optogenetics during a social reward contextual conditioning paradigm in male mice, we show that vH neurons discriminate between contexts with neutral or acquired social reward value. The formation of context-discriminating vH neurons following learning was contingent upon the presence of unconditioned stimuli. Moreover, vH neurons showed distinct contextual representations during the retrieval of social reward compared to fear contextual memories. Finally, optogenetic inhibition of locus coeruleus (LC) projections in the vH selectively disrupted social reward contextual memory by impairing vH contextual representations. Collectively, our findings reveal that the vH integrates contextual and social reward information, with memory encoding of these representations supported by input from the LC.
Collapse
Affiliation(s)
- Joana Mendes Duarte
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Robin Nguyen
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Jerome L. Greene Science Center, Columbia University, New York, NY, USA
| | - Marios Kyprou
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Kaizhen Li
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Anastasija Milentijevic
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Carlo Cerquetella
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Thomas Forro
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | - Stéphane Ciocchi
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Kassraian P, Bigler SK, Gilly Suarez DM, Shrotri N, Barnett A, Lee HJ, Young WS, Siegelbaum SA. The hippocampal CA2 region discriminates social threat from social safety. Nat Neurosci 2024; 27:2193-2206. [PMID: 39406949 DOI: 10.1038/s41593-024-01771-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/23/2024] [Indexed: 11/07/2024]
Abstract
The dorsal cornu ammonis 2 (dCA2) region of the hippocampus enables the discrimination of novel from familiar conspecifics. However, the neural bases for more complex social-spatial episodic memories are unknown. Here we report that the spatial and social contents of an aversive social experience require distinct hippocampal regions. While dorsal CA1 (dCA1) pyramidal neurons mediate the memory of an aversive location, dCA2 pyramidal neurons enable the discrimination of threat-associated (CS+) from safety-associated (CS-) conspecifics in both female and male mice. Silencing dCA2 during encoding or recall trials disrupted social fear discrimination memory, resulting in fear responses toward both the CS+ and CS- mice. Calcium imaging revealed that the aversive experience strengthened and stabilized dCA2 representations of both the CS+ and CS- mice, with the incorporation of an abstract representation of social valence into representations of social identity. Thus, dCA2 contributes to both social novelty detection and the adaptive discrimination of threat-associated from safety-associated individuals during an aversive social episodic experience.
Collapse
Affiliation(s)
- Pegah Kassraian
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA.
| | - Shivani K Bigler
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA
| | - Diana M Gilly Suarez
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA
| | - Neilesh Shrotri
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA
| | - Anastasia Barnett
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA
| | - Heon-Jin Lee
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - W Scott Young
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Steven A Siegelbaum
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York City, NY, USA
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York City, NY, USA
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York City, NY, USA
- Kavli Institute for Brain Science, Columbia University, New York City, NY, USA
| |
Collapse
|
20
|
Dunacka J, Świątek G, Wrona D. High Behavioral Reactivity to Novelty as a Susceptibility Factor for Memory and Anxiety Disorders in Streptozotocin-Induced Neuroinflammation as a Rat Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:11562. [PMID: 39519114 PMCID: PMC11546707 DOI: 10.3390/ijms252111562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Individual differences in responsiveness to environmental factors, including stress reactivity and anxiety levels, which differ between high (HR) and low (LR) responders to novelty, might be risk factors for development of memory and anxiety disorders in sporadic Alzheimer's disease (sAD). In the present study, we investigated whether behavioral characteristics of the HR and LR rats, influence the progression of sAD (neuroinflammation, β-amyloid peptide, behavioral activity related to memory (Morris water maze) and anxiety (elevated plus maze, white and illuminated open field test) in streptozotocin (STZ)-induced neuroinflammation as a model of early pathophysiological alterations in sAD. Early (45 days) in disease progression, there was a more severe impairment of reference memory and higher levels of anxiety in HRs compared with LRs. Behavioral depression in HRs was associated with higher expression of β-amyloid deposits, particularly in the NAcS, and activation of microglia (CD68+ cells) in the hypothalamus, as opposed to less inflammation in the hippocampus, particularly in CA1, compared with LRs in late (90 days) sAD progression. Our findings suggest that rats with higher behavioral activity and increased responsivity to stressors show more rapid progression of disease and anxiety disorders compared with low responders to novelty in the STZ-induced sAD model.
Collapse
Affiliation(s)
| | | | - Danuta Wrona
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, 59 Wita Stwosza Str., 80-308 Gdansk, Poland; (J.D.); (G.Ś.)
| |
Collapse
|
21
|
Bai H, Zuo X, Zhao C, Zhang S, Feng X. Non-nutritive Sweetener Aspartame Disrupts Circadian Behavior and Causes Memory Impairment in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23478-23492. [PMID: 39382230 DOI: 10.1021/acs.jafc.4c05394] [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: 10/10/2024]
Abstract
As a non-nutritive sweetener, aspartame is widely used in everyday life. However, its safety is highly controversial, especially its effects on neurobehavior. We evaluated the effects of chronic daily oral administration of aspartame-containing drinking water (at doses equivalent to 7-28% of the FDA-recommended human DIV) on memory and rhythm behaviors in mice and further investigated changes at the molecular level in the brains. Our results demonstrated that mice exposed to aspartame exhibited memory impairment. Disorders of hippocampal neurotransmitter metabolism and pathological damage may be responsible for the aspartame-induced memory impairment via inhibition of the BDNF/TrkB pathway. Furthermore, our findings suggested that disturbed clock gene expression in the hypothalamus after aspartame exposure led to altered rest-activity behavior, and this disruption of the circadian rhythm may exacerbate memory impairment. This study highlights the negative neurobehavioral effects of aspartame and provides valuable insights into its rational and safe use.
Collapse
Affiliation(s)
- Huijuan Bai
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xiang Zuo
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Chengtian Zhao
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Shuhui Zhang
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xizeng Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| |
Collapse
|
22
|
DeVuono MV, Nashed MG, Sarikahya MH, Kocsis A, Lee K, Vanin SR, Hudson R, Lonnee EP, Rushlow WJ, Hardy DB, Laviolette SR. Prenatal tetrahydrocannabinol and cannabidiol exposure produce sex-specific pathophysiological phenotypes in the adolescent prefrontal cortex and hippocampus. Neurobiol Dis 2024; 199:106588. [PMID: 38960101 DOI: 10.1016/j.nbd.2024.106588] [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: 06/05/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024] Open
Abstract
Clinical and preclinical evidence has demonstrated an increased risk for neuropsychiatric disorders following prenatal cannabinoid exposure. However, given the phytochemical complexity of cannabis, there is a need to understand how specific components of cannabis may contribute to these neurodevelopmental risks later in life. To investigate this, a rat model of prenatal cannabinoid exposure was utilized to examine the impacts of specific cannabis constituents (Δ9-tetrahydrocannabinol [THC]; cannabidiol [CBD]) alone and in combination on future neuropsychiatric liability in male and female offspring. Prenatal THC and CBD exposure were associated with low birth weight. At adolescence, offspring displayed sex-specific behavioural changes in anxiety, temporal order and social cognition, and sensorimotor gating. These phenotypes were associated with sex and treatment-specific neuronal and gene transcriptional alterations in the prefrontal cortex, and ventral hippocampus, regions where the endocannabinoid system is implicated in affective and cognitive development. Electrophysiology and RT-qPCR analysis in these regions implicated dysregulation of the endocannabinoid system and balance of excitatory and inhibitory signalling in the developmental consequences of prenatal cannabinoids. These findings reveal critical insights into how specific cannabinoids can differentially impact the developing fetal brains of males and females to enhance subsequent neuropsychiatric risk.
Collapse
Affiliation(s)
- Marieka V DeVuono
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada.
| | - Mina G Nashed
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Andrea Kocsis
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Kendrick Lee
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Sebastian R Vanin
- Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Roger Hudson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Eryn P Lonnee
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Psychiatry, University of Western Ontario, London, ON N6A 3K7, Canada
| | - Daniel B Hardy
- Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Physiology & Pharmacology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Obstetrics & Gynecology, University of Western Ontario, London, ON N6A 3K7, Canada; Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), Lawson Health Research Institute, St. Joseph's Health Care, London, ON N6C 2R5, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; Dept of Anatomy & Cell Biology, University of Western Ontario, London, ON N6A 3K7, Canada; Dept of Psychiatry, University of Western Ontario, London, ON N6A 3K7, Canada; Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), Lawson Health Research Institute, St. Joseph's Health Care, London, ON N6C 2R5, Canada
| |
Collapse
|
23
|
Puri TA, Lieblich SE, Ibrahim M, Galea LAM. Pregnancy history and estradiol influence spatial memory, hippocampal plasticity, and inflammation in middle-aged rats. Horm Behav 2024; 165:105616. [PMID: 39168073 DOI: 10.1016/j.yhbeh.2024.105616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Pregnancy and motherhood can have long-term effects on cognition and brain aging in both humans and rodents. Estrogens are related to cognitive function and neuroplasticity. Estrogens can improve cognition in postmenopausal women, but the evidence is mixed, partly due to differences in age of initiation, type of menopause, dose, formulation and route of administration. Additionally, past pregnancy influences brain aging and cognition as a younger age of first pregnancy in humans is associated with poorer aging outcomes. However, few animal studies have examined specific features of pregnancy history or the possible mechanisms underlying these changes. We examined whether maternal age at first pregnancy and estradiol differentially affected hippocampal neuroplasticity, inflammation, spatial reference cognition, and immediate early gene activation in response to spatial memory retrieval in middle-age. Thirteen-month-old rats (who were nulliparous (never mothered) or previously primiparous (had a litter) at three or seven months) received daily injections of estradiol (or vehicle) for sixteen days and were tested on the Morris Water Maze. An older age of first pregnancy was associated with impaired spatial memory but improved performance on reversal training, and increased number of new neurons in the ventral hippocampus. Estradiol decreased activation of new neurons in the dorsal hippocampus, regardless of parity history. Estradiol also decreased the production of anti-inflammatory cytokines based on age of first pregnancy. This work suggests that estradiol affects neuroplasticity and neuroinflammation in middle age, and that age of first pregnancy can have long lasting effects on hippocampus structure and function.
Collapse
Affiliation(s)
- Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Muna Ibrahim
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Center for Brain Health, University of British Columbia, Vancouver, BC, Canada; Center for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Medeiros D, Polepalli L, Li W, Pozzo-Miller L. Altered activity of mPFC pyramidal neurons and parvalbumin-expressing interneurons during social interactions in a Mecp2 mouse model for Rett syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606882. [PMID: 39149275 PMCID: PMC11326302 DOI: 10.1101/2024.08.06.606882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Social memory impairments in Mecp2 knockout (KO) mice result from altered neuronal activity in the monosynaptic projection from the ventral hippocampus (vHIP) to the medial prefrontal cortex (mPFC). The hippocampal network is hyperactive in this model for Rett syndrome, and such atypically heightened neuronal activity propagates to the mPFC through this monosynaptic projection, resulting in altered mPFC network activity and social memory deficits. However, the underlying mechanism of cellular dysfunction within this projection between vHIP pyramidal neurons (PYR) and mPFC PYRs and parvalbumin interneurons (PV-IN) resulting in social memory impairments in Mecp2 KO mice has yet to be elucidated. We confirmed social memory (but not sociability) deficits in Mecp2 KO mice using a new 4-chamber social memory arena, designed to minimize the impact of the tethering to optical fibers required for simultaneous in vivo fiber photometry of Ca2+-sensor signals during social interactions. mPFC PYRs of wildtype (WT) mice showed increases in Ca2+ signal amplitude during explorations of a novel toy mouse and interactions with both familiar and novel mice, while PYRs of Mecp2 KO mice showed smaller Ca2+ signals during interactions only with live mice. On the other hand, mPFC PV-INs of Mecp2 KO mice showed larger Ca2+ signals during interactions with a familiar cage-mate compared to those signals in PYRs, a difference absent in the WT mice. These observations suggest atypically heightened inhibition and impaired excitation in the mPFC network of Mecp2 KO mice during social interactions, potentially driving their deficit in social memory.
Collapse
Affiliation(s)
- Destynie Medeiros
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Likhitha Polepalli
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei Li
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
25
|
Wang D, Zhao D, Wang W, Hu F, Cui M, Liu J, Meng F, Liu C, Qiu C, Liu D, Xu Z, Wang Y, Zhang Y, Li W, Li C. How do lateral septum projections to the ventral CA1 influence sociability? Neural Regen Res 2024; 19:1789-1801. [PMID: 38103246 PMCID: PMC10960288 DOI: 10.4103/1673-5374.389304] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/10/2023] [Accepted: 08/02/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00033/figure1/v/2023-12-16T180322Z/r/image-tiff Social dysfunction is a risk factor for several neuropsychiatric illnesses. Previous studies have shown that the lateral septum (LS)-related pathway plays a critical role in mediating social behaviors. However, the role of the connections between the LS and its downstream brain regions in social behaviors remains unclear. In this study, we conducted a three-chamber test using electrophysiological and chemogenetic approaches in mice to determine how LS projections to ventral CA1 (vCA1) influence sociability. Our results showed that gamma-aminobutyric acid (GABA)-ergic neurons were activated following social experience, and that social behaviors were enhanced by chemogenetic modulation of these neurons. Moreover, LS GABAergic neurons extended their functional neural connections via vCA1 glutamatergic pyramidal neurons, and regulating LSGABA→vCA1Glu neural projections affected social behaviors, which were impeded by suppressing LS-projecting vCA1 neuronal activity or inhibiting GABAA receptors in vCA1. These findings support the hypothesis that LS inputs to the vCA1 can control social preferences and social novelty behaviors. These findings provide new insights regarding the neural circuits that regulate sociability.
Collapse
Affiliation(s)
- Dan Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Di Zhao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Wentao Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fengai Hu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Minghu Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Jing Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Fantao Meng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Cuilan Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Changyun Qiu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Dunjiang Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Zhicheng Xu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yameng Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yu Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- College of Nursing, Binzhou Medical University, Binzhou, Shandong Province, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Chen Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| |
Collapse
|
26
|
Walker H, Frost NA. Distinct transcriptional programs define a heterogeneous neuronal ensemble for social interaction. iScience 2024; 27:110355. [PMID: 39045099 PMCID: PMC11263963 DOI: 10.1016/j.isci.2024.110355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/01/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Social interactions are encoded by the coordinated activity of heterogeneous cell types within distributed brain regions including the medial prefrontal cortex (mPFC). However, our understanding of the cell types which comprise the social ensemble has been limited by available mouse lines and reliance on single marker genes. We identified differentially active neuronal populations during social interactions by quantifying immediate-early gene (IEG) expression using snRNA-sequencing. These studies revealed that distinct prefrontal neuron populations composed of heterogeneous cell types are activated by social interaction. Evaluation of IEG expression within these recruited neuronal populations revealed cell-type and region-specific programs, suggesting that reliance on a single molecular marker is insufficient to quantify activation across all cell types. Our findings provide a comprehensive description of cell-type specific transcriptional programs invoked by social interactions and reveal insights into the neuronal populations which compose the social ensemble.
Collapse
Affiliation(s)
- Hailee Walker
- University of Utah, Department of Neurology, Salt Lake City, UT 84132, USA
| | - Nicholas A. Frost
- University of Utah, Department of Neurology, Salt Lake City, UT 84132, USA
| |
Collapse
|
27
|
Mack NR, Bouras NN, Gao WJ. Prefrontal Regulation of Social Behavior and Related Deficits: Insights From Rodent Studies. Biol Psychiatry 2024; 96:85-94. [PMID: 38490368 PMCID: PMC12064213 DOI: 10.1016/j.biopsych.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024]
Abstract
The prefrontal cortex (PFC) is well known as the executive center of the brain, combining internal states and goals to execute purposeful behavior, including social actions. With the advancement of tools for monitoring and manipulating neural activity in rodents, substantial progress has been made in understanding the specific cell types and neural circuits within the PFC that are essential for processing social cues and influencing social behaviors. Furthermore, combining these tools with translationally relevant behavioral paradigms has also provided novel insights into the PFC neural mechanisms that may contribute to social deficits in various psychiatric disorders. This review highlights findings from the past decade that have shed light on the PFC cell types and neural circuits that support social information processing and distinct aspects of social behavior, including social interactions, social memory, and social dominance. We also explore how the PFC contributes to social deficits in rodents induced by social isolation, social fear conditioning, and social status loss. These studies provide evidence that the PFC uses both overlapping and unique neural mechanisms to support distinct components of social cognition. Furthermore, specific PFC neural mechanisms drive social deficits induced by different contexts.
Collapse
Affiliation(s)
- Nancy R Mack
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| | - Nadia N Bouras
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
28
|
Bhardwaj SK, Nath M, Wong TP, Srivastava LK. Loss of dysbindin-1 in excitatory neurons in mice impacts NMDAR-dependent behaviors, neuronal morphology and synaptic transmission in the ventral hippocampus. Sci Rep 2024; 14:15239. [PMID: 38956130 PMCID: PMC11219769 DOI: 10.1038/s41598-024-65566-4] [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: 12/22/2023] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Dysbindin-1, a protein encoded by the schizophrenia susceptibility gene DTNBP1, is reduced in the hippocampus of schizophrenia patients. It is expressed in various cellular populations of the brain and implicated in dopaminergic and glutamatergic transmission. To investigate the impact of reduced dysbindin-1 in excitatory cells on hippocampal-associated behaviors and synaptic transmission, we developed a conditional knockout mouse model with deletion of dysbindin-1 gene in CaMKIIα expressing cells. We found that dysbindin-1 reduction in CaMKII expressing cells resulted in impaired spatial and social memories, and attenuation of the effects of glutamate N-methyl-d-asparate receptor (NMDAR) antagonist MK801 on locomotor activity and prepulse inhibition of startle (PPI). Dysbindin-1 deficiency in CaMKII expressing cells also resulted in reduced protein levels of NMDAR subunit GluN1 and GluN2B. These changes were associated with increased expression of immature dendritic spines in basiliar dendrites and abnormalities in excitatory synaptic transmission in the ventral hippocampus. These results highlight the functional relevance of dysbindin-1 in excitatory cells and its implication in schizophrenia-related pathologies.
Collapse
Affiliation(s)
- Sanjeev K Bhardwaj
- Douglas Hospital Research Centre, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada.
| | - Moushumi Nath
- Douglas Hospital Research Centre, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Lalit K Srivastava
- Douglas Hospital Research Centre, Douglas Mental Health University Institute, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Integrated Programme in Neuroscience, McGill University, Montreal, QC, Canada.
| |
Collapse
|
29
|
Chung M, Imanaka K, Huang Z, Watarai A, Wang MY, Tao K, Ejima H, Aida T, Feng G, Okuyama T. Conditional knockout of Shank3 in the ventral CA1 by quantitative in vivo genome-editing impairs social memory in mice. Nat Commun 2024; 15:4531. [PMID: 38866749 PMCID: PMC11169449 DOI: 10.1038/s41467-024-48430-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/26/2024] [Indexed: 06/14/2024] Open
Abstract
Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.
Collapse
Affiliation(s)
- Myung Chung
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Katsutoshi Imanaka
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ziyan Huang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiyuki Watarai
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Mu-Yun Wang
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kentaro Tao
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Ejima
- Department of Materials Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tomomi Aida
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Teruhiro Okuyama
- Laboratory of Behavioral Neuroscience, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
30
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau C, Robinson H, Phillips ML, Yang T, Longo FM, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. Dis Model Mech 2024; 17:dmm050612. [PMID: 38785269 PMCID: PMC11139040 DOI: 10.1242/dmm.050612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/06/2024] [Indexed: 05/25/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.
Collapse
Affiliation(s)
- Destynie Medeiros
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen Ayala-Baylon
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Egido-Betancourt
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Chapleau
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Holly Robinson
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary L. Phillips
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Li
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
31
|
Bhandari K, Kanodia H, Donato F, Caroni P. Selective vulnerability of the ventral hippocampus-prelimbic cortex axis parvalbumin interneuron network underlies learning deficits of fragile X mice. Cell Rep 2024; 43:114124. [PMID: 38630591 DOI: 10.1016/j.celrep.2024.114124] [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: 10/06/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
High-penetrance mutations affecting mental health can involve genes ubiquitously expressed in the brain. Whether the specific patterns of dysfunctions result from ubiquitous circuit deficits or might reflect selective vulnerabilities of targetable subnetworks has remained unclear. Here, we determine how loss of ubiquitously expressed fragile X mental retardation protein (FMRP), the cause of fragile X syndrome, affects brain networks in Fmr1y/- mice. We find that in wild-type mice, area-specific knockout of FMRP in the adult mimics behavioral consequences of area-specific silencing. By contrast, the functional axis linking the ventral hippocampus (vH) to the prelimbic cortex (PreL) is selectively affected in constitutive Fmr1y/- mice. A chronic alteration in late-born parvalbumin interneuron networks across the vH-PreL axis rescued by VIP signaling specifically accounts for deficits in vH-PreL theta-band network coherence, ensemble assembly, and learning functions of Fmr1y/- mice. Therefore, vH-PreL axis function exhibits a selective vulnerability to loss of FMRP in the vH or PreL, leading to learning and memory dysfunctions in fragile X mice.
Collapse
Affiliation(s)
- Komal Bhandari
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Harsh Kanodia
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Pico Caroni
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| |
Collapse
|
32
|
Mercier O, Quilichini PP, Magalon K, Gil F, Ghestem A, Richard F, Boudier T, Cayre M, Durbec P. Transient demyelination causes long-term cognitive impairment, myelin alteration and network synchrony defects. Glia 2024; 72:960-981. [PMID: 38363046 DOI: 10.1002/glia.24513] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
In the adult brain, activity-dependent myelin plasticity is required for proper learning and memory consolidation. Myelin loss, alteration, or even subtle structural modifications can therefore compromise the network activity, leading to functional impairment. In multiple sclerosis, spontaneous myelin repair process is possible, but it is heterogeneous among patients, sometimes leading to functional recovery, often more visible at the motor level than at the cognitive level. In cuprizone-treated mouse model, massive brain demyelination is followed by spontaneous and robust remyelination. However, reformed myelin, although functional, may not exhibit the same morphological characteristics as developmental myelin, which can have an impact on the activity of neural networks. In this context, we used the cuprizone-treated mouse model to analyze the structural, functional, and cognitive long-term effects of transient demyelination. Our results show that an episode of demyelination induces despite remyelination long-term cognitive impairment, such as deficits in spatial working memory, social memory, cognitive flexibility, and hyperactivity. These deficits were associated with a reduction in myelin content in the medial prefrontal cortex (mPFC) and hippocampus (HPC), as well as structural myelin modifications, suggesting that the remyelination process may be imperfect in these structures. In vivo electrophysiological recordings showed that the demyelination episode altered the synchronization of HPC-mPFC activity, which is crucial for memory processes. Altogether, our data indicate that the myelin repair process following transient demyelination does not allow the complete recovery of the initial myelin properties in cortical structures. These subtle modifications alter network features, leading to prolonged cognitive deficits in mice.
Collapse
Affiliation(s)
- Océane Mercier
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale P Quilichini
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Karine Magalon
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Florian Gil
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Antoine Ghestem
- U1106 after INS, Aix Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Fabrice Richard
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Thomas Boudier
- Aix Marseille Univ, Turing Centre for Living Systems, Marseille, France
| | - Myriam Cayre
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | - Pascale Durbec
- UMR7288 after IBDM, Aix Marseille Univ, CNRS, IBDM, Marseille, France
| |
Collapse
|
33
|
Zhang M, Li X, Zhuo S, Yang M, Yu Z. Enriched Environment Enhances Sociability Through the Promotion of ESyt1-Related Synaptic Formation in the Medial Prefrontal Cortex. Mol Neurobiol 2024; 61:3019-3030. [PMID: 37964089 DOI: 10.1007/s12035-023-03742-9] [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: 08/21/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
Sociability stands as a crucial factor in the evolutionary success of all mammalian species. Notably, enriched environment (EE) housing has been shown to enhance sociability in mice. However, the precise underlying molecular mechanism remains elusive. In this study, we established an EE paradigm, housing mice for a 14-day period. Both enhanced sociability and an increased spine density in the medial prefrontal cortex (mPFC) of mice subjected to EE were detected. To elucidate the potential molecular pathway, we conducted high-performance liquid chromatography tandem mass spectrometry (HPLC-MS) analysis of the entire mPFC from both EE and home-caged (HC) housed mice. Our analysis identified 16 upregulated and 20 downregulated proteins in the EE group. Among them, Extended Synaptotagmin 1 (ESyt1), an activity-dependent endoplasmic reticulum (ER)-plasma membrane (PM) tethering protein associated with synaptic function and growth, emerged as a potentially key player in the increased synapse formation and enhanced sociability observed in EE-housed mice. Further investigation, involving the knockdown of ESyt1 expression via sh ESyt1 lentivirus in the mPFC, revealed that ESyt1 is crucial for increased spine density of mPFC and enhanced sociability of mice in an enriched environment but not in normal condition. Overall, our findings uncover a novel mechanistic insight into the positive influence of environmental enrichment on social behavior via ESyt1-mediated pathways.
Collapse
Affiliation(s)
- Meiying Zhang
- Translational Medicine Immunology Laboratory, Clinical Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Xianghe Li
- Queen Mary School of Nanchang University, Nanchang, 330031, Jiangxi Province, China
| | - Shitu Zhuo
- Translational Medicine Immunology Laboratory, Clinical Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Meili Yang
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| | - Zheng Yu
- Translational Medicine Immunology Laboratory, Clinical Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
34
|
Tudi A, Yao M, Tang F, Zhou J, Li A, Gong H, Jiang T, Li X. Subregion preference in the long-range connectome of pyramidal neurons in the medial prefrontal cortex. BMC Biol 2024; 22:95. [PMID: 38679719 PMCID: PMC11057135 DOI: 10.1186/s12915-024-01880-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is involved in complex functions containing multiple types of neurons in distinct subregions with preferential roles. The pyramidal neurons had wide-range projections to cortical and subcortical regions with subregional preferences. Using a combination of viral tracing and fluorescence micro-optical sectioning tomography (fMOST) in transgenic mice, we systematically dissected the whole-brain connectomes of intratelencephalic (IT) and pyramidal tract (PT) neurons in four mPFC subregions. RESULTS IT and PT neurons of the same subregion projected to different target areas while receiving inputs from similar upstream regions with quantitative differences. IT and PT neurons all project to the amygdala and basal forebrain, but their axons target different subregions. Compared to subregions in the prelimbic area (PL) which have more connections with sensorimotor-related regions, the infralimbic area (ILA) has stronger connections with limbic regions. The connection pattern of the mPFC subregions along the anterior-posterior axis showed a corresponding topological pattern with the isocortex and amygdala but an opposite orientation correspondence with the thalamus. CONCLUSIONS By using transgenic mice and fMOST imaging, we obtained the subregional preference whole-brain connectomes of IT and pyramidal tract PT neurons in the mPFC four subregions. These results provide a comprehensive resource for directing research into the complex functions of the mPFC by offering anatomical dissections of the different subregions.
Collapse
Affiliation(s)
- Ayizuohere Tudi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Yao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Feifang Tang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Jiandong Zhou
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.
| | - Xiangning Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China.
| |
Collapse
|
35
|
Bakoyiannis I, Ducourneau EG, N'diaye M, Fermigier A, Ducroix-Crepy C, Bosch-Bouju C, Coutureau E, Trifilieff P, Ferreira G. Obesogenic diet induces circuit-specific memory deficits in mice. eLife 2024; 13:e80388. [PMID: 38436653 PMCID: PMC10911750 DOI: 10.7554/elife.80388] [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: 05/18/2022] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Obesity is associated with neurocognitive dysfunction, including memory deficits. This is particularly worrisome when obesity occurs during adolescence, a maturational period for brain structures critical for cognition. In rodent models, we recently reported that memory impairments induced by obesogenic high-fat diet (HFD) intake during the periadolescent period can be reversed by chemogenetic manipulation of the ventral hippocampus (vHPC). Here, we used an intersectional viral approach in HFD-fed male mice to chemogenetically inactivate specific vHPC efferent pathways to nucleus accumbens (NAc) or medial prefrontal cortex (mPFC) during memory tasks. We first demonstrated that HFD enhanced activation of both pathways after training and that our chemogenetic approach was effective in normalizing this activation. Inactivation of the vHPC-NAc pathway rescued HFD-induced deficits in recognition but not location memory. Conversely, inactivation of the vHPC-mPFC pathway restored location but not recognition memory impairments produced by HFD. Either pathway manipulation did not affect exploration or anxiety-like behaviour. These findings suggest that HFD intake throughout adolescence impairs different types of memory through overactivation of specific hippocampal efferent pathways and that targeting these overactive pathways has therapeutic potential.
Collapse
Affiliation(s)
- Ioannis Bakoyiannis
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Eva Gunnel Ducourneau
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Mateo N'diaye
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Alice Fermigier
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Celine Ducroix-Crepy
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Clementine Bosch-Bouju
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | | | - Pierre Trifilieff
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077BordeauxFrance
| |
Collapse
|
36
|
Leontiadis LJ, Felemegkas P, Trompoukis G, Tsotsokou G, Miliou A, Karagianni E, Rigas P, Papatheodoropoulos C. Septotemporal Variation of Information Processing in the Hippocampus of Fmr1 KO Rat. Dev Neurosci 2024; 46:353-364. [PMID: 38368859 PMCID: PMC11614420 DOI: 10.1159/000537879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/14/2024] [Indexed: 02/20/2024] Open
Abstract
INTRODUCTION Fragile X messenger ribonucleoprotein (FMRP) is a protein involved in many neuronal processes in the nervous system including the modulation of synaptic transmission. The loss of FMRP produces the fragile X syndrome (FXS), a neurodevelopmental disorder affecting synaptic and neuronal function and producing cognitive impairments. However, the effects of FXS on short-term processing of synaptic inputs and neuronal outputs in the hippocampus have not yet been sufficiently clarified. Furthermore, it is not known whether dorsal and ventral hippocampi are affected similarly or not in FXS. METHOD We used an Fmr1 knockout (KO) rat model of FXS and recordings of evoked field potentials from the CA1 field of transverse slices from both the dorsal and the ventral hippocampi of adult rats. RESULTS Following application of a frequency stimulation protocol consisting of a ten-pulse train and recordings of fEPSP, we found that the dorsal but not ventral KO hippocampus shows altered short-term synaptic plasticity. Furthermore, applying the frequency stimulation protocol and recordings of population spikes, both segments of the KO hippocampus display altered short-term neuronal dynamics. CONCLUSIONS These data suggest that short-term processing of synaptic inputs is affected in the dorsal, not ventral, FXS hippocampus, while short-term processing of neuronal output is affected in both segments of the FXS hippocampus in a similar way. These FXS-associated changes may have significant impact on the functions of the dorsal and ventral hippocampi in individuals with FXS.
Collapse
Affiliation(s)
- Leonidas J Leontiadis
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - Panagiotis Felemegkas
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - George Trompoukis
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - Giota Tsotsokou
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - Athina Miliou
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - Evangelia Karagianni
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | - Pavlos Rigas
- Laboratory of Neurophysiology, Department of Medicine, University of Patras, Rion, Greece
| | | |
Collapse
|
37
|
Nguyen R, Sivakumaran S, Lambe EK, Kim JC. Ventral hippocampal cholecystokinin interneurons gate contextual reward memory. iScience 2024; 27:108824. [PMID: 38303709 PMCID: PMC10831933 DOI: 10.1016/j.isci.2024.108824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Associating contexts with rewards depends on hippocampal circuits, with local inhibitory interneurons positioned to play an important role in shaping activity. Here, we demonstrate that the encoding of context-reward memory requires a ventral hippocampus (vHPC) to nucleus accumbens (NAc) circuit that is gated by cholecystokinin (CCK) interneurons. In a sucrose conditioned place preference (CPP) task, optogenetically inhibiting vHPC-NAc terminals impaired the acquisition of place preference. Transsynaptic rabies tracing revealed vHPC-NAc neurons were monosynaptically innervated by CCK interneurons. Using intersectional genetic targeting of CCK interneurons, ex vivo optogenetic activation of CCK interneurons increased GABAergic transmission onto vHPC-NAc neurons, while in vivo optogenetic inhibition of CCK interneurons increased cFos in these projection neurons. Notably, CCK interneuron inhibition during sucrose CPP learning increased time spent in the sucrose-associated location, suggesting enhanced place-reward memory. Our findings reveal a previously unknown hippocampal microcircuit crucial for modulating the strength of contextual reward learning.
Collapse
Affiliation(s)
- Robin Nguyen
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of OBGYN, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Thirtamara Rajamani K, Barbier M, Lefevre A, Niblo K, Cordero N, Netser S, Grinevich V, Wagner S, Harony-Nicolas H. Oxytocin activity in the paraventricular and supramammillary nuclei of the hypothalamus is essential for social recognition memory in rats. Mol Psychiatry 2024; 29:412-424. [PMID: 38052983 PMCID: PMC11116117 DOI: 10.1038/s41380-023-02336-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023]
Abstract
Oxytocin plays an important role in modulating social recognition memory. However, the direct implication of oxytocin neurons of the paraventricular nucleus of the hypothalamus (PVH) and their downstream hypothalamic targets in regulating short- and long-term forms of social recognition memory has not been fully investigated. In this study, we employed a chemogenetic approach to target the activity of PVH oxytocin neurons in male rats and found that specific silencing of this neuronal population led to an impairment in short- and long-term social recognition memory. We combined viral-mediated fluorescent labeling of oxytocin neurons with immunohistochemical techniques and identified the supramammillary nucleus (SuM) of the hypothalamus as a target of PVH oxytocinergic axonal projections in rats. We used multiplex fluorescence in situ hybridization to label oxytocin receptors in the SuM and determined that they are predominantly expressed in glutamatergic neurons, including those that project to the CA2 region of the hippocampus. Finally, we used a highly selective oxytocin receptor antagonist in the SuM to examine the involvement of oxytocin signaling in modulating short- and long-term social recognition memory and found that it is necessary for the formation of both. This study discovered a previously undescribed role for the SuM in regulating social recognition memory via oxytocin signaling and reinforced the specific role of PVH oxytocin neurons in regulating this form of memory.
Collapse
Affiliation(s)
- Keerthi Thirtamara Rajamani
- Department of Psychiatry and Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Marie Barbier
- Department of Psychiatry and Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Cortical Systems and Behavior Laboratory, University of California San Diego, San Diego, CA, USA
| | - Kristi Niblo
- Department of Psychiatry and Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas Cordero
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY, USA
| | - Shai Netser
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Shlomo Wagner
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Hala Harony-Nicolas
- Department of Psychiatry and Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Seaver Autism Center for Research and Treatment at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
39
|
Cum M, Santiago Pérez JA, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. A systematic review and meta-analysis of how social memory is studied. Sci Rep 2024; 14:2221. [PMID: 38278973 PMCID: PMC10817899 DOI: 10.1038/s41598-024-52277-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli-a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
Affiliation(s)
- Meghan Cum
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | | - Erika Wangia
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Naeliz Lopez
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Elizabeth S Wright
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Ryo L Iwata
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Albert Li
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Amelia R Chambers
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | |
Collapse
|
40
|
Lv SS, Lv XJ, Cai YQ, Hou XY, Zhang ZZ, Wang GH, Chen LQ, Lv N, Zhang YQ. Corticotropin-releasing hormone neurons control trigeminal neuralgia-induced anxiodepression via a hippocampus-to-prefrontal circuit. SCIENCE ADVANCES 2024; 10:eadj4196. [PMID: 38241377 PMCID: PMC10798562 DOI: 10.1126/sciadv.adj4196] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/21/2023] [Indexed: 01/21/2024]
Abstract
Anxiety and depression are frequently observed in patients suffering from trigeminal neuralgia (TN), but neural circuits and mechanisms underlying this association are poorly understood. Here, we identified a dedicated neural circuit from the ventral hippocampus (vHPC) to the medial prefrontal cortex (mPFC) that mediates TN-related anxiodepression. We found that TN caused an increase in excitatory synaptic transmission from vHPCCaMK2A neurons to mPFC inhibitory neurons marked by the expression of corticotropin-releasing hormone (CRH). Activation of CRH+ neurons subsequently led to feed-forward inhibition of layer V pyramidal neurons in the mPFC via activation of the CRH receptor 1 (CRHR1). Inhibition of the vHPCCaMK2A-mPFCCRH circuit ameliorated TN-induced anxiodepression, whereas activating this pathway sufficiently produced anxiodepressive-like behaviors. Thus, our studies identified a neural pathway driving pain-related anxiodepression and a molecular target for treating pain-related psychiatric disorders.
Collapse
Affiliation(s)
- Su-Su Lv
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xue-Jing Lv
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ya-Qi Cai
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xin-Yu Hou
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhi-Zhe Zhang
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Guo-Hong Wang
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Li-Qiang Chen
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ning Lv
- Department of Translational Neuroscience, Jing’an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | | |
Collapse
|
41
|
Walker H, Frost NA. Distinct transcriptional programs define a heterogeneous neuronal ensemble for social interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573153. [PMID: 38187723 PMCID: PMC10769355 DOI: 10.1101/2023.12.22.573153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Reliable representations of information regarding complex behaviors including social interactions require the coordinated activity of heterogeneous cell types within distributed brain regions. Activity in the medial prefrontal cortex is critical in regulating social behavior, but our understanding of the specific cell types which comprise the social ensemble has been limited by available mouse lines and molecular tagging strategies which rely on the expression of a single marker gene. Here we sought to quantify the heterogeneous neuronal populations which are recruited during social interaction in parallel in a non-biased manner and determine how distinct cell types are differentially active during social interactions. We identify distinct populations of prefrontal neurons activated by social interaction by quantification of immediate early gene (IEG) expression in transcriptomically clustered neurons. This approach revealed variability in the recruitment of different excitatory and inhibitory populations within the medial prefrontal cortex. Furthermore, evaluation of the populations of IEGs recruited following social interaction revealed both cell-type and region-specific transcriptional programs, suggesting that reliance on a single molecular marker is insufficient to quantify activation across all cell types. Our findings provide a comprehensive description of cell-type specific transcriptional programs invoked by social interactions and reveal new insights into the heterogeneous neuronal populations which compose the social ensemble.
Collapse
|
42
|
Cum M, Pérez JS, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. Mind the gap: A systematic review and meta-analysis of how social memory is studied. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572606. [PMID: 38187659 PMCID: PMC10769336 DOI: 10.1101/2023.12.20.572606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli - a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
|
43
|
Orciani C, Do Carmo S, Foret MK, Hall H, Bonomo Q, Lavagna A, Huang C, Cuello AC. Early treatment with an M1 and sigma-1 receptor agonist prevents cognitive decline in a transgenic rat model displaying Alzheimer-like amyloid pathology. Neurobiol Aging 2023; 132:220-232. [PMID: 37864952 DOI: 10.1016/j.neurobiolaging.2023.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/24/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
The application of the selective allosteric M1 muscarinic and sigma-1 receptor agonist, AF710B (aka ANAVEX3-71), has shown to attenuate Alzheimer's disease-like hallmarks in McGill-R-Thy1-APP transgenic rats when administered at advanced pathological stages. It remains unknown whether preventive treatment strategies applying this compound may be equally effective. We tested whether daily oral administration of AF710B (10 µg/kg) in 7-month-old, preplaque, McGill-R-Thy1-APP rats for 7 months, followed by a 4-week washout period, could prevent Alzheimer's disease-like pathological hallmarks. Long-term AF710B treatment prevented the cognitive impairment of McGill-R-Thy1-APP rats. The effect was accompanied by a reduction in the number of amyloid plaques in the hippocampus and the levels of Aβ42 and Aβ40 peptides in the cerebral cortex. AF710B treatment also reduced microglia and astrocyte recruitment toward CA1 hippocampal Aβ-burdened neurons compared to vehicle-treated McGill-R-Thy1-APP rats, also altering the inflammatory cytokines profile. Lastly, AF710B treatment rescued the conversion of brain-derived neurotrophic factor precursor to its mature and biologically active form. Overall, these results suggest preventive and disease-modifying properties of the compound.
Collapse
Affiliation(s)
- Chiara Orciani
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 2B4, Canada
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Morgan K Foret
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Helene Hall
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Quentin Bonomo
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 2B4, Canada
| | - Agustina Lavagna
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Chunwei Huang
- Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - A Claudio Cuello
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 2B4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal H3G 1Y6, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Canada,; Department of Pharmacology, Oxford University, Oxford, UK.
| |
Collapse
|
44
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau CA, Robinson HA, Phillips ML, Yang T, Longo F, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566435. [PMID: 37986936 PMCID: PMC10659425 DOI: 10.1101/2023.11.09.566435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in methyl-CpG-binding protein-2 (MECP2), encoding a transcriptional regulator of many genes, including brain-derived neurotrophic factor (Bdnf). BDNF mRNA and protein levels are lower in RTT autopsy brains and in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of a brain penetrant, small molecule ligand of its TrkB receptors. Applied in vitro, LM22A-4 increased dendritic spine density in pyramidal neurons in cultured hippocampal slices from postnatal day (P) 7 male Mecp2 knockout (KO) mice as much as recombinant BDNF, and both effects were prevented by the TrkB receptor inhibitors K-252a and ANA-12. Consistent with its partial agonist activity, LM22A-4 did not affect spine density in CA1 pyramidal neurons in slice cultures from male wildtype (WT) mice, where typical BDNF levels outcompete its binding to TrkB. To identify neurons of known genotypes in the "mosaic" brain of female Mecp2 heterozygous (HET) mice, we treated 4-6-month-old female MeCP2-GFP WT and HET mice with peripheral injections of LM22A-4 for 4 weeks. Surprisingly, mutant neurons lacking MeCP2-GFP showed dendritic spine volumes comparable to that in WT controls, while MeCP2-GFP-expressing neurons showed larger spines, similar to the phenotype we described in symptomatic male Mecp2 KO mice where all neurons lack MeCP2. Consistent with this non-cell-autonomous mechanism, a 4-week systemic treatment with LM22A-4 had an effect only in MeCP2-GFP-expressing neurons in female Mecp2 HET mice, bringing dendritic spine volumes down to WT control levels, and without affecting spines of MeCP2-GFP-lacking neurons. At the behavioral level, we found that female Mecp2 HET mice engaged in aggressive behaviors significantly more than WT controls, which were reduced to WT levels by a 4-week systemic treatment with LM22A-4. Altogether, these data revealed differences in dendritic spine size and altered behaviors in Mecp2 HET mice, while providing support to the potential usefulness of BDNF-related therapeutic approaches such as the partial TrkB agonist LM22A-4.
Collapse
|
45
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
46
|
Kim J, Jung MW, Lee D. Reward learning improves social signal processing in autism model mice. Cell Rep 2023; 42:113228. [PMID: 37815916 DOI: 10.1016/j.celrep.2023.113228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/03/2023] [Accepted: 09/21/2023] [Indexed: 10/12/2023] Open
Abstract
Social and reward signal processing and their association are critical elements of social motivation. Despite the use of reward learning to improve the social interactions of patients with autism spectrum disorder (ASD), the underlying neural mechanisms are unknown. Here, we found different yet conjunct neuronal representations of social and reward signals in the mouse medial prefrontal cortex (mPFC). We also found that social signal processing is selectively disrupted, whereas reward signal processing is intact in the mPFC of Shank2-knockout mice, a mouse model of ASD. Furthermore, reward learning not only allows Shank2-knockout mice to associate social stimuli with reward availability, but it also rescues the impaired social signal processing. These findings provide insights into the neural basis for the therapeutic use of reward learning in ASD.
Collapse
Affiliation(s)
- Joowon Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Min Whan Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea; Center for Synaptic Brain Dysfunction, Institute for Basic Science, Daejeon 34141, Korea.
| | - Doyun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea.
| |
Collapse
|
47
|
Bakoyiannis I, Ducourneau EG, Parkes SL, Ferreira G. Pathway specific interventions reveal the multiple roles of ventral hippocampus projections in cognitive functions. Rev Neurosci 2023; 34:825-838. [PMID: 37192533 DOI: 10.1515/revneuro-2023-0009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/28/2023] [Indexed: 05/18/2023]
Abstract
Since the 1950s study of Scoville and Milner on the case H.M., the hippocampus has attracted neuroscientists' attention. The hippocampus has been traditionally divided into dorsal and ventral parts, each of which projects to different brain structures and mediates various functions. Despite a predominant interest in its dorsal part in animal models, especially regarding episodic-like and spatial cognition, recent data highlight the role of the ventral hippocampus (vHPC), as the main hippocampal output, in cognitive processes. Here, we review recent studies conducted in rodents that have used advanced in vivo functional techniques to specifically monitor and manipulate vHPC efferent pathways and delineate the roles of these specific projections in learning and memory processes. Results highlight that vHPC projections to basal amygdala are implicated in emotional memory, to nucleus accumbens in social memory and instrumental actions and to prefrontal cortex in all the above as well as in object-based memory. Some of these hippocampal projections also modulate feeding and anxiety-like behaviours providing further evidence that the "one pathway-one function" view is outdated and future directions are proposed to better understand the role of hippocampal pathways and shed further light on its connectivity and function.
Collapse
Affiliation(s)
- Ioannis Bakoyiannis
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Eva-Gunnel Ducourneau
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| | - Shauna L Parkes
- University of Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Guillaume Ferreira
- University of Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33077 Bordeaux, France
| |
Collapse
|
48
|
Hasegawa Y, Kim J, Ursini G, Jouroukhin Y, Zhu X, Miyahara Y, Xiong F, Madireddy S, Obayashi M, Lutz B, Sawa A, Brown SP, Pletnikov MV, Kamiya A. Microglial cannabinoid receptor type 1 mediates social memory deficits in mice produced by adolescent THC exposure and 16p11.2 duplication. Nat Commun 2023; 14:6559. [PMID: 37880248 PMCID: PMC10600150 DOI: 10.1038/s41467-023-42276-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Adolescent cannabis use increases the risk for cognitive impairments and psychiatric disorders. Cannabinoid receptor type 1 (Cnr1) is expressed not only in neurons and astrocytes, but also in microglia, which shape synaptic connections during adolescence. However, the role of microglia in mediating the adverse cognitive effects of delta-9-tetrahydrocannabinol (THC), the principal psychoactive constituent of cannabis, is not fully understood. Here, we report that in mice, adolescent THC exposure produces microglial apoptosis in the medial prefrontal cortex (mPFC), which was exacerbated in a model of 16p11.2 duplication, a representative copy number variation (CNV) risk factor for psychiatric disorders. These effects are mediated by microglial Cnr1, leading to reduction in the excitability of mPFC pyramidal-tract neurons and deficits in social memory in adulthood. Our findings suggest the microglial Cnr1 may contribute to adverse effect of cannabis exposure in genetically vulnerable individuals.
Collapse
Affiliation(s)
- Yuto Hasegawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juhyun Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Gianluca Ursini
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Yan Jouroukhin
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences SUNY, University at Buffalo, Buffalo, NY, USA
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Miyahara
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Feiyi Xiong
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samskruthi Madireddy
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mizuho Obayashi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Leibniz Institute for Resilience Research (LIR) gGmbH, Mainz, Germany
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences SUNY, University at Buffalo, Buffalo, NY, USA.
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Frost NA, Donohue KC, Sohal V. Context-invariant socioemotional encoding by prefrontal ensembles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563015. [PMID: 37961143 PMCID: PMC10634670 DOI: 10.1101/2023.10.19.563015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The prefrontal cortex plays a key role in social interactions, anxiety-related avoidance, and flexible context- dependent behaviors, raising the question: how do prefrontal neurons represent socioemotional information across different environments? Are contextual and socioemotional representations segregated or intermixed, and does this cause socioemotional encoding to remap or generalize across environments? To address this, we imaged neuronal activity in the medial prefrontal cortex of mice engaged in social interactions or anxiety-related avoidance within different environments. Neuronal ensembles representing context and social interaction overlapped more than expected while remaining orthogonal. Anxiety-related representations similarly generalized across environments while remaining orthogonal to contextual information. This shows how prefrontal cortex multiplexes parallel information streams using the same neurons, rather than distinct subcircuits, achieving context-invariant encoding despite context-specific reorganization of population-level activity.
Collapse
|
50
|
Hung YC, Wu YJ, Chien ME, Lin YT, Tsai CF, Hsu KS. Loss of oxytocin receptors in hilar mossy cells impairs social discrimination. Neurobiol Dis 2023; 187:106311. [PMID: 37769745 DOI: 10.1016/j.nbd.2023.106311] [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: 12/28/2022] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023] Open
Abstract
Hippocampal oxytocin receptor (OXTR) signaling is crucial for discrimination of social stimuli to guide social recognition, but circuit mechanisms and cell types involved remain incompletely understood. Here, we report a role for OXTR-expressing hilar mossy cells (MCs) of the dentate gyrus in social stimulus discrimination by regulating granule cell (GC) activity. Using a Cre-loxP recombination approach, we found that ablation of Oxtr from MCs impairs discrimination of social, but not object, stimuli in adult male mice. Ablation of MC Oxtr increases spontaneous firing rate of GCs, synaptic excitation to inhibition ratio of MC-to-GC circuit, and GC firing when temporally associated with the lateral perforant path inputs. Using mouse hippocampal slices, we found that bath application of OXTR agonist [Thr4,Gly7]-oxytocin causes membrane depolarization and increases MC firing activity. Optogenetic activation of MC-to-GC circuit ameliorates social discrimination deficit in MC OXTR deficient mice. Together, our results uncover a previously unknown role of MC OXTR signaling for discrimination of social stimuli and delineate a MC-to-GC circuit responsible for social information processing.
Collapse
Affiliation(s)
- Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Jen Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan; Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Miao-Er Chien
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan
| | - Yu-Ting Lin
- Institute of Systems Neuroscience, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Fang Tsai
- Department of Physical Medicine and Rehabilitation, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|