1
|
Pollali E, Draguhn A. Differential suppression of hippocampal network oscillations by neuropeptide Y. Neuropharmacology 2025; 266:110281. [PMID: 39725122 DOI: 10.1016/j.neuropharm.2024.110281] [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: 10/19/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Neuropeptide Y (NPY) is the most abundant neuropeptide in the brain. It exerts anxiolytic and anticonvulsive actions, reduces stress and suppresses fear memory. While its effects at the behavioral and cellular levels have been well studied, much less is known about the modulation of physiological activity patterns at the network level. We therefore studied the impact of NPY on two prominent, memory-related hippocampal activity patterns, gamma oscillations and sharp wave-ripple complexes in C57BL/6 male mice. Using established in vitro brain slice models for both patterns, we assessed the effects of NPY and receptor-specific agonists and antagonists on network activity in the CA3 and CA1 subnetworks. We report that NPY strongly suppresses sharp waves, and has significant, but much weaker effects on the power of carbachol-induced gamma oscillations. Both effects are primarily mediated via Y2 receptors. Additionally, NPY effects are much more prominent in the CA1 region compared to CA3. Our results show pattern- and region-specific effects of NPY on hippocampal networks, which suggest specific modulatory actions on hippocampus-dependent memory processes.
Collapse
Affiliation(s)
- Evangelia Pollali
- Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany.
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| |
Collapse
|
2
|
Zheng J, Sun Y, Wang F, Xie Z, Wang Q, Peng JY, Ni J. Dynamic Routing of Theta-Frequency Synchrony in the Amygdalo-Hippocampal-Entorhinal Circuit Coordinates Retrieval of Competing Memories. Neurosci Bull 2025:10.1007/s12264-025-01356-w. [PMID: 39891844 DOI: 10.1007/s12264-025-01356-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 02/03/2025] Open
Affiliation(s)
- Jiahua Zheng
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yiqi Sun
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Fuhai Wang
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhongyu Xie
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qianyun Wang
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jian-Ya Peng
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jianguang Ni
- Department of Neurosurgery, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Lipshutz A, Saltz V, Anderson KR, Manganaro A, Dumitriu D. A localized tracing technique to explore intra-amygdala functional and structural correlates of individual variability in behavioral response. Front Mol Neurosci 2025; 18:1347539. [PMID: 39916773 PMCID: PMC11794228 DOI: 10.3389/fnmol.2025.1347539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/08/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction The neurobiological basis for individual variability in behavioral responses to stimuli remains poorly understood. Probing the neural substrates that underlie individual variability in stress responses may open the door for preventive approaches that use biological markers to identify at-risk populations. New developments of viral neuronal tracing tools have led to a recent increase in studies on long range circuits and their functional role in stress responses and social behavior. While these studies are necessary to untangle largescale connectivity, most social behaviors are mediated and fine-tuned by local subregional circuitry. Methods In order to probe this local, interregional connectivity, we present a new combination of a neuronal tracing system with immediate early gene immunohistochemistry for examining structural and functional connectivity within the same animal. Specifically, we combine a retrograde transsynaptic rabies tracing system with cFos colocalization immediately after an acute stressor to elucidate local structural and stress-activated connectivity within the amygdala complex in female and male mice. Results and discussion We show how specific structural and functional connections can predict individual variability along a spectrum of social approach/avoidance following acute social defeat stress. We demonstrate how our robust method can be used to elucidate structural and functional differences in local connectivity that mediate individual variability in behavioral response.
Collapse
Affiliation(s)
- Allie Lipshutz
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University, New York, NY, United States
- Division for Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| | - Victoria Saltz
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University, New York, NY, United States
- Division for Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
- Zuckerman Institute, Columbia University, New York, NY, United States
| | - Kristin R. Anderson
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University, New York, NY, United States
- Division for Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| | - Alessia Manganaro
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University, New York, NY, United States
- Division for Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
| | - Dani Dumitriu
- Division of Developmental Neuroscience, Department of Psychiatry, Columbia University, New York, NY, United States
- Division for Child and Adolescent Health, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, United States
- Zuckerman Institute, Columbia University, New York, NY, United States
| |
Collapse
|
4
|
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
|
5
|
Trompoukis G, Miliou A, Papatheodoropoulos C. β-adrenergic receptor-induced E-S potentiation in the dorsal and ventral hippocampus. Front Synaptic Neurosci 2024; 16:1511485. [PMID: 39758826 PMCID: PMC11695307 DOI: 10.3389/fnsyn.2024.1511485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
β-adrenergic receptors (β-ARs) play a critical role in modulating learning, memory, emotionality, and long-term synaptic plasticity. Recent studies indicate that β-ARs are necessary for long-term potentiation (LTP) induction in the ventral hippocampus under moderate synaptic activation conditions that do not typically induce LTP. To explore potential dorsoventral differences in β-AR-mediated effects, we applied the β-AR agonist isoproterenol (10 μM, 30 min) to dorsal and ventral hippocampal slices, recording field excitatory postsynaptic potentials (fEPSPs) and population spikes (PSs) from the CA1 region. Isoproterenol induced robust, long-lasting PS increases, with effects three times greater in the dorsal compared to the ventral hippocampus. Isoproterenol did not significantly affect fEPSP in either segment of the hippocampus, leading to strong excitatory-to-spike (E-S) potentiation-twice as large as that in the ventral hippocampus. E-S potentiation was not associated with significant paired-pulse inhibition changes in either hippocampal segment. These differences do not appear to result from β1-AR expression levels, as they are comparable across dorsal and ventral hippocampal regions. Overall, the findings suggest that β-AR activation enhances the dorsal hippocampus's role during stress, facilitating heightened alertness, rapid spatial information processing, and effective navigation necessary for "fight-or-flight" responses.
Collapse
|
6
|
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] [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
|
7
|
Lingg RT, Johnson SB, Hinz DC, Skog TD, Lizarazu M, Romig-Martin SA, LaLumiere RT, Narayanan NS, Radley JJ. Prefrontal projections to the bed nuclei of the stria terminalis modulate the specificity of aversive memories. RESEARCH SQUARE 2024:rs.3.rs-4241372. [PMID: 39569181 PMCID: PMC11577250 DOI: 10.21203/rs.3.rs-4241372/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Generalizing aversive memories helps organisms avoid danger, whereas discriminating between dissimilar situations promotes opportunistic behaviors. We identified a novel pathway that controls the contextual specificity of memory consolidation of inhibitory avoidance learning. Optogenetic inhibition of the rostral medial prefrontal cortex (mPFC)-to-anteroventral bed nuclei of the stria terminalis (avBST) pathway after a single footshock exacerbated stress hormonal output, and 2 d later promoted generalization to a novel context. Rostral mPFC-avBST influences were directly mnemonic rather than associated with stress hormone increases, as adrenalectomy did not prevent such influences on generalization. We next observed that fear discrimination between novel and aversive contexts engaged activity along the rostral mPFC and avBST pathway. Finally, post-footshock optogenetic pathway excitation enhanced 2-d discrimination. These findings highlight a prefrontal pathway in which activity immediately after aversive experiences promotes mnemonic discrimination between threatening and non-threatening contexts and may be importance for understanding trauma generalization in psychiatric illnesses.
Collapse
Affiliation(s)
- Ryan T. Lingg
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Shane B. Johnson
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
| | - Dalton C. Hinz
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Timothy D. Skog
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
| | - Manuela Lizarazu
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Sara A. Romig-Martin
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
| | - Ryan T. LaLumiere
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Nandakumar S. Narayanan
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Jason J. Radley
- Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Neuroscience Program, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
8
|
Biane JS, Ladow MA, Fan A, Choi HS, Zhou LZ, Hassan S, Apodaca-Montano DL, Kwon AO, Bratsch-Prince JX, Kheirbek MA. Representations of stimulus meaning in the hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618280. [PMID: 39464010 PMCID: PMC11507678 DOI: 10.1101/2024.10.14.618280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The ability to discriminate and categorize the meaning of environmental stimuli and respond accordingly is essential for survival. The ventral hippocampus (vHPC) controls emotional and motivated behaviors in response to environmental cues and is hypothesized to do so in part by deciphering the positive or negative quality of these cues. Yet, what features of the environment are represented in the activity patterns of vCA1 neurons, and whether the positive or negative meaning of a stimulus is present at this stage, remains unclear. Here, using 2-photon calcium imaging across six different experimental paradigms, we consistently found that vCA1 ensembles encode the identity, sensory features, and intensity of learned and innately salient stimuli, but not their overall valence. These results offer a reappraisal of vCA1 function, wherein information corresponding to individual stimulus features and their behavioral saliency predominates, while valence-related information is attached elsewhere.
Collapse
Affiliation(s)
- Jeremy S. Biane
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Max A. Ladow
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Austin Fan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hye Sun Choi
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lexi Zichen Zhou
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Shazreh Hassan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L. Apodaca-Montano
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew O. Kwon
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Joshua X. Bratsch-Prince
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A. Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences and Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Meng F, Song J, Huang X, Zhang M, Sun X, Jing Q, Cao S, Xie Z, Liu Q, Zhang H, Li C. Inhibiting endoplasmic reticulum stress alleviates perioperative neurocognitive disorders by reducing neuroinflammation mediated by NLRP3 inflammasome activation. CNS Neurosci Ther 2024; 30:e70049. [PMID: 39432407 PMCID: PMC11493103 DOI: 10.1111/cns.70049] [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: 03/12/2024] [Revised: 08/12/2024] [Accepted: 09/04/2024] [Indexed: 10/23/2024] Open
Abstract
AIM The aim of this study is to explore the key mechanisms of perioperative neurocognitive dysfunction (PND) after anesthesia/surgery (A/S) by screening hub genes. METHODS Transcriptome sequencing was conducted on hippocampal samples obtained from 18-month-old C57BL/6 mice assigned to control (Ctrl) and A/S groups. The functionality of differentially expressed genes (DEGs) was investigated using Metascape. Hub genes associated with changes between the two groups were screened by combining weighted gene coexpression network analysis within CytoHubba. Reverse transcription PCR and western blotting were used to validate changes in mRNA and protein expression, respectively. NLRP3 inflammasome activation was detected by western blotting and ELISA. Tauroursodeoxycholic acid (TUDCA), an inhibitor of endoplasmic reticulum (ER) stress, was administrated preoperatively to explore its effects on the occurrence of PND. Immunofluorescence analysis was performed to evaluate the activation of astrocytes and microglia in the hippocampus, and hippocampus-dependent learning and memory were assessed using behavioral experiments. RESULTS In total, 521 DEGs were detected between the control and A/S groups. These DEGs were significantly enriched in biological processes related to metabolic processes and their regulation. Four hub genes (Hspa5, Igf1r, Sfpq, and Xbp1) were identified. Animal experiments have shown that mice in the A/S group exhibited cognitive impairments accompanied by increased Hspa5 and Xbp1 expression, ER stress, and activation of NLRP3 inflammasome. CONCLUSIONS Inhibiting ER stress alleviated cognitive impairment in A/S mice; particularly, ER stress induced by A/S results in NLRP3 inflammasome activation and neuroinflammation. Moreover, the preoperative administration of TUDCA inhibited ER stress, NLRP3 inflammasome activation, and neuroinflammation.
Collapse
Affiliation(s)
- Fanbing Meng
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Jian Song
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Xinwei Huang
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Meixian Zhang
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Xiaoxiao Sun
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Qi Jing
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Silu Cao
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Zheng Xie
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Qiong Liu
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Hui Zhang
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Cheng Li
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, School of Medicine, Tongji UniversityShanghaiChina
| |
Collapse
|
10
|
Hunsberger HC, Lee S, Jin M, Lanio M, Whye A, Cha J, Scarlata M, Matthews LC, Jayaseelan K, Denny CA. Sex-Specific Effects of Anxiety on Cognition and Activity-Dependent Neural Networks: Insights From (Female) Mice and (Wo)men. Biol Psychiatry 2024:S0006-3223(24)01621-4. [PMID: 39349155 DOI: 10.1016/j.biopsych.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Neuropsychiatric symptoms, such as depression and anxiety, are observed in 90% of patients with Alzheimer's disease (AD), two-thirds of whom are women. Neuropsychiatric symptoms usually manifest long before AD onset creating a therapeutic opportunity. Here, we examined the impact of anxiety on AD progression and the underlying brainwide neuronal mechanisms. METHODS To gain mechanistic insight into how anxiety affects AD progression, we performed a cross-sectional analysis on mood, cognition, and neural activity using the ArcCreERT2 x eYFP (enhanced yellow fluorescent protein) x amyloid precursor protein/presenilin 1 (APP/PS1) (AD) mice. The Alzheimer's Disease Neuroimaging Initiative dataset was used to determine the impact of anxiety on AD progression in humans. RESULTS Female APP/PS1 mice exhibited anxiety-like behavior and cognitive decline at an earlier age than control mice and male mice. Brainwide analysis of c-Fos+ revealed changes in regional correlations and overall network connectivity in APP/PS1 mice. Sex-specific eYFP+/c-Fos+ changes were observed; female APP/PS1 mice exhibited less eYFP+/c-Fos+ cells in dorsal CA3, whereas male APP/PS1 mice exhibited less eYFP+/c-Fos+ cells in the dorsal dentate gyrus. In the Alzheimer's Disease Neuroimaging Initiative dataset, anxiety predicted transition to dementia. Female participants positive for anxiety and amyloid transitioned more quickly to dementia than male participants. CONCLUSIONS While future studies are needed to understand whether anxiety is a predictor, a neuropsychiatric biomarker, or a comorbid symptom that occurs during disease onset, these results suggest that there are sex differences in AD network dysfunction and that personalized medicine may benefit male and female patients with AD rather than a one-size-fits-all approach.
Collapse
Affiliation(s)
- Holly C Hunsberger
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, New York
| | - Seonjoo Lee
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, New York; Mental Health Data Science, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, New York
| | - Michelle Jin
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, New York
| | - Marcos Lanio
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, New York
| | - Alicia Whye
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, New York
| | - Jiook Cha
- Department of Biostatistics (in Psychiatry), Mailman School of Public Health, Columbia University, New York, New York; Division of Child and Adolescent Psychiatry, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, New York; Data Science Institute, Columbia University, New York, New York; Department of Psychology, Seoul National University, Seoul, South Korea
| | - Miranda Scarlata
- Department of Neuroscience, Vassar College, Poughkeepsie, New York
| | - Louise C Matthews
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, New York; Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, New York
| | | | - Christine A Denny
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, New York; Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, New York.
| |
Collapse
|
11
|
Li K, Koukoutselos K, Sakaguchi M, Ciocchi S. Distinct ventral hippocampal inhibitory microcircuits regulating anxiety and fear behaviors. Nat Commun 2024; 15:8228. [PMID: 39300067 DOI: 10.1038/s41467-024-52466-4] [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: 11/17/2023] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
In emotion research, anxiety and fear have always been interconnected, sharing overlapping brain structures and neural circuitry. Recent investigations, however, have unveiled parallel long-range projection pathways originating from the ventral hippocampus, shedding light on their distinct roles in anxiety and fear. Yet, the mechanisms governing the emergence of projection-specific activity patterns to mediate different negative emotions remain elusive. Here, we show a division of labor in local GABAergic inhibitory microcircuits of the ventral hippocampus, orchestrating the activity of subpopulations of pyramidal neurons to shape anxiety and fear behaviors in mice. These findings offer a comprehensive insight into how distinct inhibitory microcircuits are dynamically engaged to encode different emotional states.
Collapse
Affiliation(s)
- Kaizhen Li
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland.
| | | | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Stéphane Ciocchi
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland.
| |
Collapse
|
12
|
Oleksiak CR, Plas SL, Carriaga D, Vasudevan K, Maren S, Moscarello JM. Ventral hippocampus mediates inter-trial responding in signaled active avoidance. Behav Brain Res 2024; 470:115071. [PMID: 38806099 DOI: 10.1016/j.bbr.2024.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/07/2024] [Accepted: 05/24/2024] [Indexed: 05/30/2024]
Abstract
The hippocampus has a central role in regulating contextual processes in memory. We have shown that pharmacological inactivation of ventral hippocampus (VH) attenuates the context-dependence of signaled active avoidance (SAA) in rats. Here, we explore whether the VH mediates intertrial responses (ITRs), which are putative unreinforced avoidance responses that occur between trials. First, we examined whether VH inactivation would affect ITRs. Male rats underwent SAA training and subsequently received intra-VH infusions of saline or muscimol before retrieval tests in the training context. Rats that received muscimol performed significantly fewer ITRs, but equivalent avoidance responses, compared to controls. Next, we asked whether chemogenetic VH activation would increase ITR vigor. In male and female rats expressing excitatory (hM3Dq) DREADDs, systemic CNO administration produced a robust ITR increase that was not due to nonspecific locomotor effects. Then, we examined whether chemogenetic VH activation potentiated ITRs in an alternate (non-training) test context and found it did. Finally, to determine if context-US associations mediate ITRs, we exposed rats to the training context for three days after SAA training to extinguish the context. Rats submitted to context extinction did not show a reliable decrease in ITRs during a retrieval test, suggesting that context-US associations are not responsible for ITRs. Collectively, these results reveal an important role for the VH in context-dependent ITRs during SAA. Further work is required to explore the neural circuits and associative basis for these responses, which may be underlie pathological avoidance that occurs in humans after threat has passed.
Collapse
Affiliation(s)
- Cecily R Oleksiak
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845, USA
| | - Samantha L Plas
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845, USA
| | - Denise Carriaga
- Department of Psychological Science, University of Texas Rio Grande Valley, TX 78539
| | - Krithika Vasudevan
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845, USA
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845, USA
| | - Justin M Moscarello
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845, USA; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845, USA.
| |
Collapse
|
13
|
Zhang BB, Ling XY, Shen QY, Zhang YX, Li QX, Xie ST, Li HZ, Zhang QP, Yung WH, Wang JJ, Ke Y, Zhang XY, Zhu JN. Suppression of excitatory synaptic transmission in the centrolateral amygdala via presynaptic histamine H3 heteroreceptors. J Physiol 2024. [PMID: 38953534 DOI: 10.1113/jp286392] [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: 02/06/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
The central histaminergic system has a pivotal role in emotional regulation and psychiatric disorders, including anxiety, depression and schizophrenia. However, the effect of histamine on neuronal activity of the centrolateral amygdala (CeL), an essential node for fear and anxiety processing, remains unknown. Here, using immunostaining and whole-cell patch clamp recording combined with optogenetic manipulation of histaminergic terminals in CeL slices prepared from histidine decarboxylase (HDC)-Cre rats, we show that histamine selectively suppresses excitatory synaptic transmissions, including glutamatergic transmission from the basolateral amygdala, on both PKC-δ- and SOM-positive CeL neurons. The histamine-induced effect is mediated by H3 receptors expressed on VGLUT1-/VGLUT2-positive presynaptic terminals in CeL. Furthermore, optoactivation of histaminergic afferent terminals from the hypothalamic tuberomammillary nucleus (TMN) also significantly suppresses glutamatergic transmissions in CeL via H3 receptors. Histamine neither modulates inhibitory synaptic transmission by presynaptic H3 receptors nor directly excites CeL neurons by postsynaptic H1, H2 or H4 receptors. These results suggest that histaminergic afferent inputs and presynaptic H3 heteroreceptors may hold a critical position in balancing excitatory and inhibitory synaptic transmissions in CeL by selective modulation of glutamatergic drive, which may not only account for the pathophysiology of psychiatric disorders but also provide potential psychotherapeutic targets. KEY POINTS: Histamine selectively suppresses the excitatory, rather than inhibitory, synaptic transmissions on both PKC-δ- and SOM-positive neurons in the centrolateral amygdala (CeL). H3 receptors expressed on VGLUT1- or VGLUT2-positive afferent terminals mediate the suppression of histamine on glutamatergic synaptic transmission in CeL. Optogenetic activation of hypothalamic tuberomammillary nucleus (TMN)-CeL histaminergic projections inhibits glutamatergic transmission in CeL via H3 receptors.
Collapse
Affiliation(s)
- Bei-Bei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xin-Yu Ling
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qing-Yi Shen
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yang-Xun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qian-Xiao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi-Peng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Wing-Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, SAR, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, National Resource Center for Mutant Mice, and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Joo B, Xu S, Park H, Kim K, Rah JC, Koo JW. Parietal-Frontal Pathway Controls Relapse of Fear Memory in a Novel Context. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100315. [PMID: 38726036 PMCID: PMC11078648 DOI: 10.1016/j.bpsgos.2024.100315] [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: 06/28/2023] [Revised: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 05/12/2024] Open
Abstract
Background Fear responses significantly affect daily life and shape our approach to uncertainty. However, the potential resurgence of fear in unfamiliar situations poses a significant challenge to exposure-based therapies for maladaptive fear responses. Nonetheless, how novel contextual stimuli are associated with the relapse of extinguished fear remains unknown. Methods Using a context-dependent fear renewal model, the functional circuits and underlying mechanisms of the posterior parietal cortex (PPC) and anterior cingulate cortex (ACC) were investigated using optogenetic, histological, in vivo, and ex vivo electrophysiological and pharmacological techniques. Results We demonstrated that the PPC-to-ACC pathway governs fear relapse in a novel context. We observed enhanced populational calcium activity in the ACC neurons that received projections from the PPC and increased synaptic activity in the basolateral amygdala-projecting PPC-to-ACC neurons upon renewal in a novel context, where excitatory postsynaptic currents amplitudes increased but inhibitory postsynaptic current amplitudes decreased. In addition, we found that parvalbumin-expressing interneurons controlled novel context-dependent fear renewal, which was blocked by the chronic administration of fluoxetine. Conclusions Our findings highlight the PPC-to-ACC pathway in mediating the relapse of extinguished fear in novel contexts, thereby contributing significant insights into the intricate neural mechanisms that govern fear renewal.
Collapse
Affiliation(s)
- Bitna Joo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Shijie Xu
- Medical Research Center, Affiliated Cancer Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Hyungju Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Kipom Kim
- Research Strategy Office, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jong-Cheol Rah
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
- Sensory & Motor Systems Neuroscience Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| |
Collapse
|
15
|
Greiner EM, Petrovich GD. Recruitment of hippocampal and thalamic pathways to the central amygdala in the control of feeding behavior under novelty. Brain Struct Funct 2024; 229:1179-1191. [PMID: 38625554 DOI: 10.1007/s00429-024-02791-7] [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: 09/05/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
It is adaptive to restrict eating under uncertainty, such as during habituation to novel foods and unfamiliar environments. However, sustained restrictive eating can become maladaptive. Currently, the neural substrates of restrictive eating are poorly understood. Using a model of feeding avoidance under novelty, our recent study identified forebrain activation patterns and found evidence that the central nucleus of the amygdala (CEA) is a core integrating node. The current study analyzed the activity of CEA inputs in male and female rats to determine if specific pathways are recruited during feeding under novelty. Recruitment of direct inputs from the paraventricular nucleus of the thalamus (PVT), the infralimbic cortex (ILA), the agranular insular cortex (AI), the hippocampal ventral field CA1, and the bed nucleus of the stria terminals (BST) was assessed with combined retrograde tract tracing and Fos induction analysis. The study found that during consumption of a novel food in a novel environment, larger number of neurons within the PVTp and the CA1 that send monosynaptic inputs to the CEA were recruited compared to controls that consumed familiar food in a familiar environment. The ILA, AI, and BST inputs to the CEA were similarly recruited across conditions. There were no sex differences in activation of any of the pathways analyzed. These results suggest that the PVTp-CEA and CA1-CEA pathways underlie feeding inhibition during novelty and could be potential sites of malfunction in excessive food avoidance.
Collapse
Affiliation(s)
- Eliza M Greiner
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, MA, 02467, USA
| | - Gorica D Petrovich
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
16
|
Liu Y, Ye S, Li XN, Li WG. Memory Trace for Fear Extinction: Fragile yet Reinforceable. Neurosci Bull 2024; 40:777-794. [PMID: 37812300 PMCID: PMC11178705 DOI: 10.1007/s12264-023-01129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/08/2023] [Indexed: 10/10/2023] Open
Abstract
Fear extinction is a biological process in which learned fear behavior diminishes without anticipated reinforcement, allowing the organism to re-adapt to ever-changing situations. Based on the behavioral hypothesis that extinction is new learning and forms an extinction memory, this new memory is more readily forgettable than the original fear memory. The brain's cellular and synaptic traces underpinning this inherently fragile yet reinforceable extinction memory remain unclear. Intriguing questions are about the whereabouts of the engram neurons that emerged during extinction learning and how they constitute a dynamically evolving functional construct that works in concert to store and express the extinction memory. In this review, we discuss recent advances in the engram circuits and their neural connectivity plasticity for fear extinction, aiming to establish a conceptual framework for understanding the dynamic competition between fear and extinction memories in adaptive control of conditioned fear responses.
Collapse
Affiliation(s)
- Ying Liu
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Shuai Ye
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Xin-Ni Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Huashan Hospital, Institute for Translational Brain Research, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Zhang Y, Xu C, Gu Y. Context Processing in Contextual and Cued Fear Extinction. Neurosci Bull 2024; 40:835-839. [PMID: 38619694 PMCID: PMC11178715 DOI: 10.1007/s12264-024-01205-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 04/16/2024] Open
Affiliation(s)
- Yimu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chun Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Chen PB, Chen R, LaPierre N, Chen Z, Mefford J, Marcus E, Heffel MG, Soto DC, Ernst J, Luo C, Flint J. Complementation testing identifies genes mediating effects at quantitative trait loci underlying fear-related behavior. CELL GENOMICS 2024; 4:100545. [PMID: 38697120 PMCID: PMC11099346 DOI: 10.1016/j.xgen.2024.100545] [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: 01/03/2024] [Revised: 02/23/2024] [Accepted: 04/04/2024] [Indexed: 05/04/2024]
Abstract
Knowing the genes involved in quantitative traits provides an entry point to understanding the biological bases of behavior, but there are very few examples where the pathway from genetic locus to behavioral change is known. To explore the role of specific genes in fear behavior, we mapped three fear-related traits, tested fourteen genes at six quantitative trait loci (QTLs) by quantitative complementation, and identified six genes. Four genes, Lamp, Ptprd, Nptx2, and Sh3gl, have known roles in synapse function; the fifth, Psip1, was not previously implicated in behavior; and the sixth is a long non-coding RNA, 4933413L06Rik, of unknown function. Variation in transcriptome and epigenetic modalities occurred preferentially in excitatory neurons, suggesting that genetic variation is more permissible in excitatory than inhibitory neuronal circuits. Our results relieve a bottleneck in using genetic mapping of QTLs to uncover biology underlying behavior and prompt a reconsideration of expected relationships between genetic and functional variation.
Collapse
Affiliation(s)
- Patrick B Chen
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachel Chen
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nathan LaPierre
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zeyuan Chen
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joel Mefford
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emilie Marcus
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew G Heffel
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniela C Soto
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jason Ernst
- Department of Computer Science, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA, USA; Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chongyuan Luo
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan Flint
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Premachandran H, Wilkin J, Arruda-Carvalho M. Minimizing Variability in Developmental Fear Studies in Mice: Toward Improved Replicability in the Field. Curr Protoc 2024; 4:e1040. [PMID: 38713136 DOI: 10.1002/cpz1.1040] [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] [Indexed: 05/08/2024]
Abstract
In rodents, the first weeks of postnatal life feature remarkable changes in fear memory acquisition, retention, extinction, and discrimination. Early development is also marked by profound changes in brain circuits underlying fear memory processing, with heightened sensitivity to environmental influences and stress, providing a powerful model to study the intersection between brain structure, function, and the impacts of stress. Nevertheless, difficulties related to breeding and housing young rodents, preweaning manipulations, and potential increased variability within that population pose considerable challenges to developmental fear research. Here we discuss several factors that may promote variability in studies examining fear conditioning in young rodents and provide recommendations to increase replicability. We focus primarily on experimental conditions, design, and analysis of rodent fear data, with an emphasis on mouse studies. The convergence of anatomical, synaptic, physiological, and behavioral changes during early life may increase variability, but careful practice and transparency in reporting may improve rigor and consensus in the field. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Hanista Premachandran
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- These authors contributed equally to this work
| | - Jennifer Wilkin
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- These authors contributed equally to this work
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Masella G, Silva F, Corti E, Azkona G, Madeira MF, Tomé ÂR, Ferreira SG, Cunha RA, Duarte CB, Santos M. The amygdala NT3-TrkC pathway underlies inter-individual differences in fear extinction and related synaptic plasticity. Mol Psychiatry 2024; 29:1322-1337. [PMID: 38233468 PMCID: PMC11189811 DOI: 10.1038/s41380-024-02412-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024]
Abstract
Fear-related pathologies are among the most prevalent psychiatric conditions, having inappropriate learned fear and resistance to extinction as cardinal features. Exposure therapy represents a promising therapeutic approach, the efficiency of which depends on inter-individual variation in fear extinction learning, which neurobiological basis is unknown. We characterized a model of extinction learning, whereby fear-conditioned mice were categorized as extinction (EXT)-success or EXT-failure, according to their inherent ability to extinguish fear. In the lateral amygdala, GluN2A-containing NMDAR are required for LTP and stabilization of fear memories, while GluN2B-containing NMDAR are required for LTD and fear extinction. EXT-success mice showed attenuated LTP, strong LTD and higher levels of synaptic GluN2B, while EXT-failure mice showed strong LTP, no LTD and higher levels of synaptic GluN2A. Neurotrophin 3 (NT3) infusion in the lateral amygdala was sufficient to rescue extinction deficits in EXT-failure mice. Mechanistically, activation of tropomyosin receptor kinase C (TrkC) with NT3 in EXT-failure slices attenuated lateral amygdala LTP, in a GluN2B-dependent manner. Conversely, blocking endogenous NT3-TrkC signaling with TrkC-Fc chimera in EXT-success slices strengthened lateral amygdala LTP. Our data support a key role for the NT3-TrkC system in inter-individual differences in fear extinction in rodents, through modulation of amygdalar NMDAR composition and synaptic plasticity.
Collapse
Affiliation(s)
- Gianluca Masella
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal
| | - Francisca Silva
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal
| | - Elisa Corti
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal
| | - Garikoitz Azkona
- Department of Basic Psychological Processes and Their Development, School of Psychology, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Maria Francisca Madeira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal
| | - Ângelo R Tomé
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Samira G Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carlos B Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Mónica Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra (iiiUC), Coimbra, Portugal.
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
21
|
Oleksiak CR, Plas SL, Carriaga D, Vasudevan K, Maren S, Moscarello JM. Ventral hippocampus mediates inter-trial responding in signaled active avoidance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585627. [PMID: 38562746 PMCID: PMC10983994 DOI: 10.1101/2024.03.18.585627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The hippocampus has a central role in regulating contextual processes in memory. We have shown that pharmacological inactivation of ventral hippocampus (VH) attenuates the context-dependence of signaled active avoidance (SAA) in rats. Here, we explore whether the VH mediates intertrial responses (ITRs), which are putative unreinforced avoidance responses that occur between trials. First, we examined whether VH inactivation would affect ITRs. Male rats underwent SAA training and subsequently received intra-VH infusions of saline or muscimol before retrieval tests in the training context. Rats that received muscimol performed significantly fewer ITRs, but equivalent avoidance responses, compared to controls. Next, we asked whether chemogenetic VH activation would increase ITR vigor. In male and female rats expressing excitatory (hM3Dq) DREADDs, systemic CNO administration produced a robust ITR increase that was not due to nonspecific locomotor effects. Then, we examined whether chemogenetic VH activation potentiated ITRs in an alternate (non-training) test context and found it did. Finally, to determine if context-US associations mediate ITRs, we exposed rats to the training context for three days after SAA training to extinguish the context. Rats submitted to context extinction did not show a reliable decrease in ITRs during a retrieval test, suggesting that context-US associations are not responsible for ITRs. Collectively, these results reveal an important role for the VH in context-dependent ITRs during SAA. Further work is required to explore the neural circuits and associative basis for these responses, which may be underlie pathological avoidance that occurs in humans after threat has passed.
Collapse
Affiliation(s)
- Cecily R. Oleksiak
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845
| | - Samantha L. Plas
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845
| | - Denise Carriaga
- Department of Psychological Science, University of Texas Rio Grande Valley, TX 78539
| | - Krithika Vasudevan
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845
| | - Stephen Maren
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845
| | - Justin M. Moscarello
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77845
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77845
| |
Collapse
|
22
|
Dirven BCJ, van Melis L, Daneva T, Dillen L, Homberg JR, Kozicz T, Henckens MJAG. Hippocampal Trauma Memory Processing Conveying Susceptibility to Traumatic Stress. Neuroscience 2024; 540:87-102. [PMID: 38220126 DOI: 10.1016/j.neuroscience.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/16/2024]
Abstract
While the majority of the population is ever exposed to a traumatic event during their lifetime, only a fraction develops posttraumatic stress disorder (PTSD). Disrupted trauma memory processing has been proposed as a core factor underlying PTSD symptomatology. We used transgenic Targeted-Recombination-in-Active-Populations (TRAP) mice to investigate potential alterations in trauma-related hippocampal memory engrams associated with the development of PTSD-like symptomatology. Mice were exposed to a stress-enhanced fear learning paradigm, in which prior exposure to a stressor affects the learning of a subsequent fearful event (contextual fear conditioning using foot shocks), during which neuronal activity was labeled. One week later, mice were behaviorally phenotyped to identify mice resilient and susceptible to developing PTSD-like symptomatology. Three weeks post-learning, mice were re-exposed to the conditioning context to induce remote fear memory recall, and associated hippocampal neuronal activity was assessed. While no differences in the size of the hippocampal neuronal ensemble activated during fear learning were observed between groups, susceptible mice displayed a smaller ensemble activated upon remote fear memory recall in the ventral CA1, higher regional hippocampal parvalbuminneuronal density and a relatively lower activity of parvalbumininterneurons upon recall. Investigation of potential epigenetic regulators of the engram revealed rather generic (rather than engram-specific) differences between groups, with susceptible mice displaying lower hippocampal histone deacetylase 2 expression, and higher methylation and hydroxymethylation levels. These finding implicate variation in epigenetic regulation within the hippocampus, as well as reduced regional hippocampal activity during remote fear memory recall in interindividual differences in susceptibility to traumatic stress.
Collapse
Affiliation(s)
- Bart C J Dirven
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Lennart van Melis
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Teya Daneva
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Lieke Dillen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Tamas Kozicz
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands; Center for Individualized Medicine, Department of Clinical Genomics, and Biochemical Genetics Laboratory, Mayo Clinic, Rochester, MN 55905, USA; University of Pecs Medical School, Department of Anatomy, Pecs, Hungary
| | - Marloes J A G Henckens
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
23
|
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
|
24
|
Qiu S, Hu Y, Huang Y, Gao T, Wang X, Wang D, Ren B, Shi X, Chen Y, Wang X, Wang D, Han L, Liang Y, Liu D, Liu Q, Deng L, Chen Z, Zhan L, Chen T, Huang Y, Wu Q, Xie T, Qian L, Jin C, Huang J, Deng W, Jiang T, Li X, Jia X, Yuan J, Li A, Yan J, Xu N, Xu L, Luo Q, Poo MM, Sun Y, Li CT, Yao H, Gong H, Sun YG, Xu C. Whole-brain spatial organization of hippocampal single-neuron projectomes. Science 2024; 383:eadj9198. [PMID: 38300992 DOI: 10.1126/science.adj9198] [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: 07/27/2023] [Accepted: 12/19/2023] [Indexed: 02/03/2024]
Abstract
Mapping single-neuron projections is essential for understanding brain-wide connectivity and diverse functions of the hippocampus (HIP). Here, we reconstructed 10,100 single-neuron projectomes of mouse HIP and classified 43 projectome subtypes with distinct projection patterns. The number of projection targets and axon-tip distribution depended on the soma location along HIP longitudinal and transverse axes. Many projectome subtypes were enriched in specific HIP subdomains defined by spatial transcriptomic profiles. Furthermore, we delineated comprehensive wiring diagrams for HIP neurons projecting exclusively within the HIP formation (HPF) and for those projecting to both intra- and extra-HPF targets. Bihemispheric projecting neurons generally projected to one pair of homologous targets with ipsilateral preference. These organization principles of single-neuron projectomes provide a structural basis for understanding the function of HIP neurons.
Collapse
Affiliation(s)
- Shou Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yachuang Hu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yiming Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Lingang Laboratory, Shanghai 200031, China
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Taosha Gao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaofei Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Danying Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Biyu Ren
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoxue Shi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinran Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luyao Han
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yikai Liang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dechen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qingxu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Deng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhaoqin Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lijie Zhan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianzhi Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuzhe Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Lingang Laboratory, Shanghai 200031, China
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qingge Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Taorong Xie
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liuqin Qian
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenxi Jin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiawen Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Deng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Xiangning Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Xueyan Jia
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Jing Yuan
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Anan Li
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Ninglong Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Qingming Luo
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Mu-Ming Poo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201210, China
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chengyu T Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Lingang Laboratory, Shanghai 200031, China
| | - Haishan Yao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Gong
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chun Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of the Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
25
|
Xia F, Fascianelli V, Vishwakarma N, Ghinger FG, Fusi S, Kheirbek MA. Identifying and modulating neural signatures of stress susceptibility and resilience enables control of anhedonia. RESEARCH SQUARE 2024:rs.3.rs-3581329. [PMID: 38343839 PMCID: PMC10854313 DOI: 10.21203/rs.3.rs-3581329/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Anhedonia is a core aspect of major depressive disorder. Traditionally viewed as a blunted emotional state in which individuals are unable to experience joy, anhedonia also diminishes the drive to seek rewards and the ability to value and learn about them 1-4.The neural underpinnings of anhedonia and how this emotional state drives related behavioral changes remain unclear. Here, we investigated these questions by taking advantage of the fact that when mice are exposed to traumatic social stress, susceptible animals become socially withdrawn and anhedonic, where they cease to seek high-value rewards, while others remain resilient. By performing high density electrophysiological recordings and comparing neural activity patterns of these groups in the basolateral amygdala (BLA) and ventral CA1 (vCA1) of awake behaving animals, we identified neural signatures of susceptibility and resilience to anhedonia. When animals actively sought rewards, BLA activity in resilient mice showed stronger discrimination between upcoming reward choices. In contrast, susceptible mice displayed a rumination-like signature, where BLA neurons encoded the intention to switch or stay on a previously chosen reward. When animals were at rest, the spontaneous BLA activity of susceptible mice was higher dimensional than in controls, reflecting a greater number of distinct neural population states. Notably, this spontaneous activity allowed us to decode group identity and to infer if a mouse had a history of stress better than behavioral outcomes alone. Finally, targeted manipulation of vCA1 inputs to the BLA in susceptible mice rescued dysfunctional neural dynamics, amplified dynamics associated with resilience, and reversed their anhedonic behavior. This work reveals population-level neural signatures that explain individual differences in responses to traumatic stress, and suggests that modulating vCA1-BLA inputs can enhance resilience by regulating these dynamics.
Collapse
Affiliation(s)
- Frances Xia
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Valeria Fascianelli
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
| | - Nina Vishwakarma
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
| | - Frances Grace Ghinger
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
| | - Stefano Fusi
- Center for Theoretical Neuroscience, Columbia University, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, NY, USA
- Department of Neuroscience, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, NY, USA
- Kavli Institute for Brain Science, Columbia University Irving Medical Center, NY, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, USA
| |
Collapse
|
26
|
Chen PB, Chen R, LaPierre N, Chen Z, Mefford J, Marcus E, Heffel MG, Soto DC, Ernst J, Luo C, Flint J. Complementation testing identifies causal genes at quantitative trait loci underlying fear related behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574060. [PMID: 38260483 PMCID: PMC10802323 DOI: 10.1101/2024.01.03.574060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Knowing the genes involved in quantitative traits provides a critical entry point to understanding the biological bases of behavior, but there are very few examples where the pathway from genetic locus to behavioral change is known. Here we address a key step towards that goal by deploying a test that directly queries whether a gene mediates the effect of a quantitative trait locus (QTL). To explore the role of specific genes in fear behavior, we mapped three fear-related traits, tested fourteen genes at six QTLs, and identified six genes. Four genes, Lsamp, Ptprd, Nptx2 and Sh3gl, have known roles in synapse function; the fifth gene, Psip1, is a transcriptional co-activator not previously implicated in behavior; the sixth is a long non-coding RNA 4933413L06Rik with no known function. Single nucleus transcriptomic and epigenetic analyses implicated excitatory neurons as likely mediating the genetic effects. Surprisingly, variation in transcriptome and epigenetic modalities between inbred strains occurred preferentially in excitatory neurons, suggesting that genetic variation is more permissible in excitatory than inhibitory neuronal circuits. Our results open a bottleneck in using genetic mapping of QTLs to find novel biology underlying behavior and prompt a reconsideration of expected relationships between genetic and functional variation.
Collapse
|
27
|
Vasudevan K, Hassell JE, Maren S. Hippocampal Engrams and Contextual Memory. ADVANCES IN NEUROBIOLOGY 2024; 38:45-66. [PMID: 39008010 DOI: 10.1007/978-3-031-62983-9_4] [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] [Indexed: 07/16/2024]
Abstract
Memories are not formed in a vacuum and often include rich details about the time and place in which events occur. Contextual stimuli promote the retrieval of events that have previously occurred in the encoding context and limit the retrieval of context-inappropriate information. Contexts that are associated with traumatic or harmful events both directly elicit fear and serve as reminders of aversive events associated with trauma. It has long been appreciated that the hippocampus is involved in contextual learning and memory and is central to contextual fear conditioning. However, little is known about the underlying neuronal mechanisms underlying the encoding and retrieval of contextual fear memories. Recent advancements in neuronal labeling methods, including activity-dependent tagging of cellular ensembles encoding memory ("engrams"), provide unique insight into the neural substrates of memory in the hippocampus. Moreover, these methods allow for the selective manipulation of memory ensembles. Attenuating or erasing fear memories may have considerable therapeutic value for patients with post-traumatic stress disorder or other trauma- or stressor-related conditions. In this chapter, we review the role of the hippocampus in contextual fear conditioning in rodents and explore recent work implicating hippocampal ensembles in the encoding and retrieval of aversive memories.
Collapse
Affiliation(s)
- Krithika Vasudevan
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - James E Hassell
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
28
|
Rahimi-Danesh M, Samizadeh MA, Sajadi AE, Rezvankhah T, Vaseghi S. Sex difference affects fear extinction but not lithium efficacy in rats following fear-conditioning with respect to the hippocampal level of BDNF. Pharmacol Biochem Behav 2024; 234:173675. [PMID: 37972713 DOI: 10.1016/j.pbb.2023.173675] [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: 08/29/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
In rodents, exposure to electrical shock and creating a strong fear memory using fear-conditioning model can induce PTSD-like behavior. In this study, we induced a fear-conditioning model in rats and investigated freezing (PTSD-like) behavior, 21 days after three shocks exposure (0.6 mA, 3 s, 30 seconds interval) in both male and female rats. Lithium was injected intraperitoneally (100 mg/kg) in three protocols: (1) 1 h after fear-conditioning (2) 1 h, 24 h, and 48 h after fear-conditioning (3), 1 h, 24 h, 48 h, 72 h, and 96 h after fear-conditioning. Extinction training (20 sounds without shocks, 75 dB, 3 s, 30 seconds interval) was performed in three protocols: (1) 1 h after fear-conditioning (one session), (2) 1 h, 24 h, and 48 h after fear-conditioning (three sessions), (3), 1 h, 24 h, 48 h, 72 h, and 96 h after fear-conditioning (five sessions). Forced swim test (FST) and hot plate were used to assess behavior. Results showed that lithium in all protocols had no effect on freezing behavior, FST, and pain subthreshold in all rats. Extinction training decreased freezing behavior, with more efficacy in females. In males, only 5-session training was effective, while in females all protocols were effective. Extinction training also altered pain perception and the results of FST, depending on the sessions and was different in males and females. Brain-derived neurotrophic factor (BDNF) mRNA level was increased in females following 3 and 5 sessions, and in males following 5 sessions extinction training. In conclusion, we suggested that there is a sex difference for the effect of extinction training on freezing behavior and BDNF mRNA level in a rat model of fear-conditioning.
Collapse
Affiliation(s)
- Mehrsa Rahimi-Danesh
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Mohammad-Ali Samizadeh
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Amir-Ehsan Sajadi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Tara Rezvankhah
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
29
|
Casagrande MA, Porto RR, Haubrich J, Kautzmann A, de Oliveira Álvares L. Emotional Value of Fear Memory and the Role of the Ventral Hippocampus in Systems Consolidation. Neuroscience 2023; 535:184-193. [PMID: 37944583 DOI: 10.1016/j.neuroscience.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/31/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
Recent studies have explored the circuitry involving the ventral hippocampus (vHPC), the amygdala, and the prefrontal cortex, a pathway mainly activated to store contextual information efficiently. Lesions in the vHPC impair remote memory, but not in the short term. However, how the vHPC is affected by distinct memory strength or its role in systems consolidation has not yet been elucidated. Here, we investigated how distinct training intensities, with strong or weak contextual fear conditioning, affect activation of the dorsal hippocampus (dHPC) and the vHPC. We found that the time course of memory consolidation differs in fear memories of different training intensities in both the dHPC and vHPC. Our results also indicate that memory generalization happens alongside greater activation of the vHPC, and these processes occur faster with stronger fear memories. The vHPC is required for the expression of remote fear memory and may control contextual fear generalization, a view corroborated by the fact that inactivation of the vHPC suppresses generalized fear expression, making memory more precise again. Systems consolidation occurs concomitantly with greater activation of the vHPC, which is accelerated in stronger fear memories. These findings lead us to propose that greater activation of the vHPC could be used as a marker for memory generalization.
Collapse
Affiliation(s)
- M A Casagrande
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves 9500, Prédio 43422, Sala 216, CEP 91.501-970 Porto Alegre, Rio Grande do Sul, Brasil
| | - R R Porto
- Behavioural Neuroscience Laboratory, Western Sydney University, School of Medicine, Cnr David Pilgrim Ave & Goldsmith Ave, Building 30, Campbelltown, NSW 2560, Australia
| | - J Haubrich
- Dept. of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Universitätsstraße, 150 MA 4/150, 44780 Bochum, Germany
| | - A Kautzmann
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves 9500, Prédio 43422, Sala 216, CEP 91.501-970 Porto Alegre, Rio Grande do Sul, Brasil
| | - L de Oliveira Álvares
- Laboratório de Neurobiologia da Memória, Departamento de Biofísica, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), Av. Bento Gonçalves 9500, Prédio 43422, Sala 216, CEP 91.501-970 Porto Alegre, Rio Grande do Sul, Brasil.
| |
Collapse
|
30
|
Santos TB, Kramer-Soares JC, Coelho CAO, Oliveira MGM. Temporal association activates projections from the perirhinal cortex and ventral CA1 to the prelimbic cortex and from the prelimbic cortex to the basolateral amygdala. Cereb Cortex 2023; 33:11456-11470. [PMID: 37823340 DOI: 10.1093/cercor/bhad375] [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: 08/04/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
In trace fear conditioning, the prelimbic cortex exhibits persistent activity during the interval between the conditioned and unconditioned stimuli, which maintains a conditioned stimulus representation. Regions cooperating for this function or encoding the conditioned stimulus before the interval could send inputs to the prelimbic cortex, supporting learning. The basolateral amygdala has conditioned stimulus- and unconditioned stimulus-responsive neurons, convergently activated. The prelimbic cortex could directly project to the basolateral amygdala to associate the transient memory of the conditioned stimulus with the unconditioned stimulus. We investigated the neuronal circuit supporting temporal associations using contextual fear conditioning with a 5-s interval, in which 5 s separates the contextual conditioned stimulus from the unconditioned stimulus. Injecting retrobeads, we quantified c-Fos in prelimbic cortex- or basolateral amygdala-projecting neurons from 9 regions after contextual fear conditioning with a 5-s interval or contextual fear conditioning, in which the conditioned and unconditioned stimuli overlap. The contextual fear conditioning with a 5-s interval activated ventral CA1 and perirhinal cortex neurons projecting to the prelimbic cortex and prelimbic cortex neurons projecting to basolateral amygdala. Both fear conditioning activated ventral CA1 and lateral entorhinal cortex neurons projecting to basolateral amygdala and basolateral amygdala neurons projecting to prelimbic cortex. The perirhinal cortex → prelimbic cortex and ventral CA1 → prelimbic cortex connections are the first identified prelimbic cortex afferent projections participating in temporal associations. These results help to understand time-linked memories, a process required in episodic and working memories.
Collapse
Affiliation(s)
- Thays B Santos
- Departamento de Psicobiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| | - Juliana C Kramer-Soares
- Departamento de Psicobiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
- Universidade Cruzeiro do Sul-UNICSUL, São Paulo 08060-070, Brazil
| | - Cesar A O Coelho
- Neuroscience and Mental Health, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Maria G M Oliveira
- Departamento de Psicobiologia, Universidade Federal de São Paulo-UNIFESP, São Paulo 04023-062, Brazil
| |
Collapse
|
31
|
Hochgerner H, Singh S, Tibi M, Lin Z, Skarbianskis N, Admati I, Ophir O, Reinhardt N, Netser S, Wagner S, Zeisel A. Neuronal types in the mouse amygdala and their transcriptional response to fear conditioning. Nat Neurosci 2023; 26:2237-2249. [PMID: 37884748 PMCID: PMC10689239 DOI: 10.1038/s41593-023-01469-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
The amygdala is a brain region primarily associated with emotional response. The use of genetic markers and single-cell transcriptomics can provide insights into behavior-associated cell state changes. Here we present a detailed cell-type taxonomy of the adult mouse amygdala during fear learning and memory consolidation. We perform single-cell RNA sequencing on naïve and fear-conditioned mice, identify 130 neuronal cell types and validate their spatial distributions. A subset of all neuronal types is transcriptionally responsive to fear learning and memory retrieval. The activated engram cells upregulate activity-response genes and coordinate the expression of genes associated with neurite outgrowth, synaptic signaling, plasticity and development. We identify known and previously undescribed candidate genes responsive to fear learning. Our molecular atlas may be used to generate hypotheses to unveil the neuron types and neural circuits regulating the emotional component of learning and memory.
Collapse
Affiliation(s)
- Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shelly Singh
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Muhammad Tibi
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zhige Lin
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nuphar Reinhardt
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shai Netser
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
32
|
Abouelnaga KH, Huff AE, O'Neill OS, Messer WS, Winters BD. Activating M1 muscarinic cholinergic receptors induces destabilization of resistant contextual fear memories in rats. Neurobiol Learn Mem 2023; 205:107821. [PMID: 37666411 DOI: 10.1016/j.nlm.2023.107821] [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: 05/18/2023] [Revised: 07/27/2023] [Accepted: 08/27/2023] [Indexed: 09/06/2023]
Abstract
Destabilization of previously consolidated memories places them in a labile state in which they are open to modification. However, strongly encoded fear memories tend to be destabilization-resistant and the conditions required to destabilize such memories remain poorly understood. Our lab has previously shown that exposure to salient novel contextual cues during memory reactivation can destabilize strongly encoded object location memories and that activity at muscarinic cholinergic receptors is critical for this effect. In the current study, we similarly targeted destabilization-resistant fear memories, hypothesizing that exposure to salient novelty at the time of reactivation would induce destabilization of strongly encoded fear memories in a muscarinic receptor-dependent manner. First, we show that contextual fear memories induced by 3 context-shock pairings readily destabilize upon memory reactivation, and that this destabilization is blocked by systemic (ip) administration of the muscarinic receptor antagonist scopolamine (0.3 mg/kg) in male rats. Following that, we confirm that this effect is dorsal hippocampus (dHPC)-dependent by targeting M1 receptors in the CA1 region with pirenzepine. Next, we show that more strongly encoded fear memories (induced with 5 context-shock pairings) resist destabilization. Consistent with our previous work, however, we report that salient novelty (a change in floor texture) presented during the reactivation session promotes destabilization of resistant contextual fear memories in a muscarinic receptor-dependent manner. Finally, the effect of salient novelty on memory destabilization was mimicked by stimulating muscarinic receptors with the selective M1 agonist CDD-0102A (ip, 0.3 mg/kg). These findings reveal further generalizability of our previous results implicating novel cues and M1 muscarinic signaling in promoting destabilization of resistant memories and suggest possible therapeutic options for disorders characterized by persistent, maladaptive fear memories such as PTSD and phobias.
Collapse
Affiliation(s)
- Karim H Abouelnaga
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road E, N1G 2W1 Guelph, ON, Canada.
| | - Andrew E Huff
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road E, N1G 2W1 Guelph, ON, Canada.
| | - Olivia S O'Neill
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road E, N1G 2W1 Guelph, ON, Canada.
| | - William S Messer
- Departments of Pharmacology and Experimental Therapeutics, University of Toledo, 2801 West Bancroft St, Toledo, OH 43606, USA.
| | - Boyer D Winters
- Department of Psychology and Collaborative Neuroscience Program, University of Guelph, 50 Stone Road E, N1G 2W1 Guelph, ON, Canada.
| |
Collapse
|
33
|
Pronier É, Morici JF, Girardeau G. The role of the hippocampus in the consolidation of emotional memories during sleep. Trends Neurosci 2023; 46:912-925. [PMID: 37714808 DOI: 10.1016/j.tins.2023.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 09/17/2023]
Abstract
Episodic memory relies on the hippocampus, a heterogeneous brain region with distinct functions. Spatial representations in the dorsal hippocampus (dHPC) are crucial for contextual memory, while the ventral hippocampus (vHPC) is more involved in emotional processing. Here, we review the literature in rodents highlighting the anatomical and functional properties of the hippocampus along its dorsoventral axis that underlie its role in contextual and emotional memory encoding, consolidation, and retrieval. We propose that the coordination between the dorsal and vHPC through theta oscillations during rapid eye movement (REM) sleep, and through sharp-wave ripples during non-REM (NREM) sleep, might facilitate the transfer of contextual information for integration with valence-related processing in other structures of the network. Further investigation into the physiology of the vHPC and its connections with other brain areas is needed to deepen the current understanding of emotional memory consolidation during sleep.
Collapse
Affiliation(s)
- Éléonore Pronier
- Institut du Fer à Moulin, Inserm U1270, Sorbonne Université, Paris, France
| | | | | |
Collapse
|
34
|
Brockway ET, Simon S, Drew MR. Ventral hippocampal projections to infralimbic cortex and basolateral amygdala are differentially activated by contextual fear and extinction recall. Neurobiol Learn Mem 2023; 205:107832. [PMID: 37757953 PMCID: PMC10919432 DOI: 10.1016/j.nlm.2023.107832] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/23/2023] [Accepted: 09/24/2023] [Indexed: 09/29/2023]
Abstract
Fear and extinction learning are thought to generate distinct and competing memory representations in the hippocampus. How these memory representations modulate the expression of appropriate behavioral responses remains unclear. To investigate this question, we used cholera toxin B subunit to retrolabel ventral hippocampal (vHPC) neurons projecting to the infralimbic cortex (IL) and basolateral amygdala (BLA) and then quantified c-Fos immediate early gene activity within these populations following expression of either contextual fear recall or contextual fear extinction recall. Fear recall was associated with increased c-Fos expression in vHPC projections to the BLA, whereas extinction recall was associated with increased activity in vHPC projections to IL. A control experiment was performed to confirm that the apparent shift in projection neuron activity was associated with extinction learning rather than mere context exposure. Overall, results indicate that hippocampal contextual fear and extinction memory representations differentially activate vHPC projections to IL and BLA. These findings suggest that hippocampal memory representations orchestrate appropriate behavioral responses through selective activation of projection pathways.
Collapse
Affiliation(s)
- Emma T Brockway
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Sarah Simon
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
35
|
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: 3] [Impact Index Per Article: 1.5] [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
|
36
|
Lim SC, Fusi S, Hen R. Ventral CA1 Population Codes for Anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.559358. [PMID: 37808689 PMCID: PMC10557595 DOI: 10.1101/2023.09.25.559358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The ventral hippocampus is a critical node in the distributed brain network that controls anxiety. Using miniature microscopy and calcium imaging, we recorded ventral CA1 (vCA1) neurons in freely moving mice as they explored variants of classic behavioral assays for anxiety. Unsupervised behavioral segmentation revealed clusters of behavioral motifs that corresponded to exploratory and vigilance-like states. We discovered multiple vCA1 population codes that represented the anxiogenic features of the environment, such as bright light and openness, as well as the moment-to-moment anxiety state of the animals. These population codes possessed distinct generalization properties: neural representations of anxiogenic features were different for open field and elevated plus/zero maze tasks, while neural representations of moment-to-moment anxiety state were similar across both experimental contexts. Our results suggest that anxiety is not tied to the aversive compartments of these mazes but is rather defined by a behavioral state and its corresponding population code that generalizes across environments.
Collapse
|
37
|
Greiner EM, Petrovich G. Recruitment of Hippocampal and Thalamic Pathways to the Central Amygdala in the Control of Feeding Behavior Under Novelty. RESEARCH SQUARE 2023:rs.3.rs-3328572. [PMID: 37790294 PMCID: PMC10543251 DOI: 10.21203/rs.3.rs-3328572/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
It is adaptive to restrict eating under uncertainty, such as during habituation to novel foods and unfamiliar environments. However, sustained restrictive eating is a core symptom of eating disorders and has serious long-term health consequences. Current therapeutic efforts are limited, because the neural substrates of restrictive eating are poorly understood. Using a model of feeding avoidance under novelty, our recent study identified forebrain activation patterns and found evidence that the central nucleus of the amygdala (CEA) is a core integrating node. The current study analyzed the activity of CEA inputs in male and female rats to determine if specific pathways are recruited during feeding under novelty. Recruitment of direct inputs from the paraventricular nucleus of the thalamus (PVT), the infralimbic cortex (ILA), the agranular insular cortex (AI), the hippocampal ventral field CA1, and the bed nucleus of the stria terminals (BST) was assessed with combined retrograde tract tracing and Fos induction analysis. The study found that during consumption of a novel food in a novel environment, larger number of neurons within the PVTp and the CA1 that send monosynaptic inputs to the CEA were recruited compared to controls that consumed familiar food in a familiar environment. The ILA, AI, and BST inputs to the CEA were similarly recruited across conditions. There were no sex differences in activation of any of the pathways analyzed. These results suggest that the PVTp-CEA and CA1-CEA pathways underlie feeding inhibition during novelty and could be potential sites of malfunction in excessive food avoidance.
Collapse
|
38
|
Zhu Y, Xie SZ, Peng AB, Yu XD, Li CY, Fu JY, Shen CJ, Cao SX, Zhang Y, Chen J, Li XM. Distinct Circuits From the Central Lateral Amygdala to the Ventral Part of the Bed Nucleus of Stria Terminalis Regulate Different Fear Memory. Biol Psychiatry 2023:S0006-3223(23)01553-6. [PMID: 37678543 DOI: 10.1016/j.biopsych.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The ability to differentiate stimuli that predict fear is critical for survival; however, the underlying molecular and circuit mechanisms remain poorly understood. METHODS We combined transgenic mice, in vivo transsynaptic circuit-dissecting anatomical approaches, optogenetics, pharmacological methods, and electrophysiological recording to investigate the involvement of specific extended amygdala circuits in different fear memory. RESULTS We identified the projections from central lateral amygdala (CeL) protein kinase C δ (PKCδ)-positive neurons and somatostatin (SST)-positive neurons to GABAergic (gamma-aminobutyric acidergic) and glutamatergic neurons in the ventral part of the bed nucleus of stria terminalis (vBNST). Prolonged optogenetic activation or inhibition of the PKCδCeL-vBNST pathway specifically reduced context fear memory, whereas the SSTCeL-vBNST pathway mainly reduced tone fear memory. Intriguingly, optogenetic manipulation of vBNST neurons that received the projection from PKCδCeL neurons exerted bidirectional regulation of context fear, whereas manipulation of vBNST neurons that received the projection from SSTCeL neurons could bidirectionally regulate both context and tone fear memory. We subsequently demonstrated the presence of δ and κ opioid receptor protein expression within the CeL-vBNST circuits, potentially accounting for the discrepancy between prolonged activation of GABAergic circuits and inhibition of downstream vBNST neurons. Finally, administration of an opioid receptor antagonist cocktail on the PKCδCeL-vBNST or SSTCeL-vBNST pathway successfully restored context or tone fear memory reduction induced by prolonged activation of the circuits. CONCLUSIONS Together, these findings establish a functional role for distinct CeL-vBNST circuits in the differential regulation and appropriate maintenance of fear.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Shi-Ze Xie
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Ai-Bing Peng
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-Dan Yu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Chun-Yue Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jia-Yu Fu
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Chen-Jie Shen
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Shu-Xia Cao
- Department of Neurology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Zhang
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Jiadong Chen
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-Ming Li
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
39
|
Yang S, Zhu G. Phytotherapy of abnormality of fear memory: A narrative review of mechanisms. Fitoterapia 2023; 169:105618. [PMID: 37482307 DOI: 10.1016/j.fitote.2023.105618] [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: 05/23/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
It is generally believed that in post-traumatic stress disorder (PTSD), the high expression of fear memory is mainly determined by amygdala hyperactivity and hippocampus hypoactivity. In this review, we firstly updated the mechanisms of fear memory, and then searched the experimental evidence of phytotherapy for fear memory in the past five years. Based on the summary of those experimental studies, we further discussed the future research strategies of plant medicines, including the study of the mechanism of specific brain regions, the optimal time for the prevention and treatment of fear memory-related diseases such as PTSD, and the development of new drugs with active components of plant medicines. Accordingly, plant medicines play a clear role in improving fear memory abnormalities and have the drug development potential in the treatment of fear-related disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei, Anhui 230061, China; Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
40
|
Bai T, Zhan L, Zhang N, Lin F, Saur D, Xu C. Learning-prolonged maintenance of stimulus information in CA1 and subiculum during trace fear conditioning. Cell Rep 2023; 42:112853. [PMID: 37481720 DOI: 10.1016/j.celrep.2023.112853] [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: 09/01/2022] [Revised: 04/12/2023] [Accepted: 07/08/2023] [Indexed: 07/25/2023] Open
Abstract
Temporal associative learning binds discontiguous conditional stimuli (CSs) and unconditional stimuli (USs), possibly by maintaining CS information in the hippocampus after its offset. Yet, how learning regulates such maintenance of CS information in hippocampal circuits remains largely unclear. Using the auditory trace fear conditioning (TFC) paradigm, we identify a projection from the CA1 to the subiculum critical for TFC. Deep-brain calcium imaging shows that the peak of trace activity in the CA1 and subiculum is extended toward the US and that the CS representation during the trace period is enhanced during learning. Interestingly, such plasticity is consolidated only in the CA1, not the subiculum, after training. Moreover, CA1 neurons, but not subiculum neurons, increasingly become active during CS-and-trace and shock periods, respectively, and correlate with CS-evoked fear retrieval afterward. These results indicate that learning dynamically enhances stimulus information maintenance in the CA1-subiculum circuit during learning while storing CS and US memories primarily in the CA1 area.
Collapse
Affiliation(s)
- Tao Bai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Lijie Zhan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Na Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Feikai Lin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dieter Saur
- Department of Internal Medicine 2, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - Chun Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201210, China.
| |
Collapse
|
41
|
Kalinovskii AP, Pushkarev AP, Mikhailenko AD, Kudryavtsev DS, Belozerova OA, Shmygarev VI, Yatskin ON, Korolkova YV, Kozlov SA, Osmakov DI, Popov A, Andreev YA. Dual Modulator of ASIC Channels and GABA A Receptors from Thyme Alters Fear-Related Hippocampal Activity. Int J Mol Sci 2023; 24:13148. [PMID: 37685955 PMCID: PMC10487430 DOI: 10.3390/ijms241713148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated ion channels that mediate nociception in the peripheral nervous system and contribute to fear and learning in the central nervous system. Sevanol was reported previously as a naturally-occurring ASIC inhibitor from thyme with favorable analgesic and anti-inflammatory activity. Using electrophysiological methods, we found that in the high micromolar range, the compound effectively inhibited homomeric ASIC1a and, in sub- and low-micromolar ranges, positively modulated the currents of α1β2γ2 GABAA receptors. Next, we tested the compound in anxiety-related behavior models using a targeted delivery into the hippocampus with parallel electroencephalographic measurements. In the open field, 6 µM sevanol reduced both locomotor and θ-rhythmic activity similar to GABA, suggesting a primary action on the GABAergic system. At 300 μM, sevanol markedly suppressed passive avoidance behavior, implying alterations in conditioned fear memory. The observed effects could be linked to distinct mechanisms involving GABAAR and ASIC1a. These results elaborate the preclinical profile of sevanol as a candidate for drug development and support the role of ASIC channels in fear-related functions of the hippocampus.
Collapse
Affiliation(s)
- Aleksandr P. Kalinovskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Anton P. Pushkarev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Anastasia D. Mikhailenko
- Moscow State Academy of Veterinary Medicine and Biotechnology—MVA named after K.I. Skryabin, ul. Akademika Skryabina, 23, 109472 Moscow, Russia
| | - Denis S. Kudryavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Olga A. Belozerova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Vladimir I. Shmygarev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Oleg N. Yatskin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Yuliya V. Korolkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Sergey A. Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Dmitry I. Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, bld. 2, 119991 Moscow, Russia
| | - Alexander Popov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
| | - Yaroslav A. Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia (D.S.K.); (O.A.B.); (S.A.K.); (A.P.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, bld. 2, 119991 Moscow, Russia
| |
Collapse
|
42
|
Senba E, Kami K. Exercise therapy for chronic pain: How does exercise change the limbic brain function? NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100143. [PMID: 38099274 PMCID: PMC10719519 DOI: 10.1016/j.ynpai.2023.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 12/17/2023]
Abstract
We are exposed to various external and internal threats which might hurt us. The role of taking flexible and appropriate actions against threats is played by "the limbic system" and at the heart of it there is the ventral tegmental area and nucleus accumbens (brain reward system). Pain-related fear causes excessive excitation of amygdala, which in turn causes the suppression of medial prefrontal cortex, leading to chronification of pain. Since the limbic system of chronic pain patients is functionally impaired, they are maladaptive to their situations, unable to take goal-directed behavior and are easily caught by fear-avoidance thinking. We describe the neural mechanisms how exercise activates the brain reward system and enables chronic pain patients to take goal-directed behavior and overcome fear-avoidance thinking. A key to getting out from chronic pain state is to take advantage of the behavioral switching function of the basal nucleus of amygdala. We show that exercise activates positive neurons in this nucleus which project to the nucleus accumbens and promote reward behavior. We also describe fear conditioning and extinction are affected by exercise. In chronic pain patients, the fear response to pain is enhanced and the extinction of fear memories is impaired, so it is difficult to get out of "fear-avoidance thinking". Prolonged avoidance of movement and physical inactivity exacerbate pain and have detrimental effects on the musculoskeletal and cardiovascular systems. Based on the recent findings on multiple bran networks, we propose a well-balanced exercise prescription considering the adherence and pacing of exercise practice. We conclude that therapies targeting the mesocortico-limbic system, such as exercise therapy and cognitive behavioral therapy, may become promising tools in the fight against chronic pain.
Collapse
Affiliation(s)
- Emiko Senba
- Department of Physical Therapy, Osaka Yukioka College of Health Science, 1-1-41 Sojiji, Ibaraki-City, Osaka 567-0801, Japan
- Department of Rehabilitation Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 641-8509, Japan
| | - Katsuya Kami
- Department of Rehabilitation, Wakayama Faculty of Health Care Sciences, Takarazuka University of Medical and Health Care, 2252 Nakanoshima, Wakayama City, Wakayama 640-8392, Japan
- Department of Rehabilitation Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama 641-8509, Japan
| |
Collapse
|
43
|
Hunsberger HC, Lee S, Jin M, Lanio M, Whye A, Cha J, Scarlata M, Jayaseelan K, Denny CA. Sex-Specific Effects of Anxiety on Cognition and Activity-Dependent Neural Networks: Insights from (Female) Mice and (Wo)Men. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548180. [PMID: 37503264 PMCID: PMC10369916 DOI: 10.1101/2023.07.07.548180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Neuropsychiatric symptoms (NPS), such as depression and anxiety, are observed in 90% of Alzheimer's disease (AD) patients, two-thirds of whom are women. NPS usually manifest long before AD onset creating a therapeutic opportunity. Here, we examined the impact of anxiety on AD progression and the underlying brain-wide neuronal mechanisms. METHODS To gain mechanistic insight into how anxiety impacts AD progression, we performed a cross-sectional analysis on mood, cognition, and neural activity utilizing the ArcCreERT2 x enhanced yellow fluorescent protein (eYFP) x APP/PS1 (AD) mice. The ADNI dataset was used to determine the impact of anxiety on AD progression in human subjects. RESULTS Female AD mice exhibited anxiety-like behavior and cognitive decline at an earlier age than control (Ctrl) mice and male mice. Brain-wide analysis of c-Fos+ revealed changes in regional correlations and overall network connectivity in AD mice. Sex-specific memory trace changes were observed; female AD mice exhibited impaired memory traces in dorsal CA3 (dCA3), while male AD mice exhibited impaired memory traces in the dorsal dentate gyrus (dDG). In the ADNI dataset, anxiety predicted transition to dementia. Female subjects positive for anxiety and amyloid transitioned more quickly to dementia than male subjects. CONCLUSIONS While future studies are needed to understand whether anxiety is a predictor, a neuropsychiatric biomarker, or a comorbid symptom that occurs during disease onset, these results suggest that AD network dysfunction is sexually dimorphic, and that personalized medicine may benefit male and female AD patients rather than a one size fits all approach.
Collapse
Affiliation(s)
- Holly C. Hunsberger
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, USA
- Center for Neurodegenerative Diseases and Therapeutics, Rosalind Franklin University of Medicine and Science/The Chicago Medical School; North Chicago, IL, USA
| | - Seonjoo Lee
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC); New York, NY, USA
- Mental Health Data Science, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, USA
| | - Michelle Jin
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY, USA
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, USA
| | - Marcos Lanio
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY, USA
- Medical Scientist Training Program (MSTP), Columbia University Irving Medical Center (CUIMC), New York, NY, USA
| | - Alicia Whye
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC); New York, NY, USA
| | - Jiook Cha
- Department of Biostatistics (in Psychiatry), Mailman School of Public Health, Columbia University; New York, NY, USA
- Division of Child and Adolescent Psychiatry, NYSPI/RFMH; New York, NY, USA
- Data Science Institute, Columbia University; New York, NY, USA
- Department of Psychology, Seoul National University; Seoul, South Korea
| | - Miranda Scarlata
- Department of Neuroscience, Vassar College; Poughkeepsie, NY USA
- Department of Social Policy and Intervention, University of Oxford; Oxford, England
| | - Keerthana Jayaseelan
- Barnard College, Columbia University; New York, NY, USA
- Department of Medicine, New York Medical College/Westchester Medical Center; Valhalla, NY, USA
| | - Christine. A. Denny
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH) / New York State Psychiatric Institute (NYSPI), New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center (CUIMC); New York, NY, USA
| |
Collapse
|
44
|
Du W, Li E, Guo J, Arano R, Kim Y, Chen YT, Thompson A, Oh SJ, Samuel A, Li Y, Oyibo HK, Xu W. Directed stepwise tracing of polysynaptic neuronal circuits with replication-deficient pseudorabies virus. CELL REPORTS METHODS 2023; 3:100506. [PMID: 37426757 PMCID: PMC10326449 DOI: 10.1016/j.crmeth.2023.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 03/17/2023] [Accepted: 05/24/2023] [Indexed: 07/11/2023]
Abstract
Brain functions are accomplished by polysynaptic circuits formed by neurons wired together through multiple orders of synaptic connections. Polysynaptic connectivity has been difficult to examine due to a lack of methods of continuously tracing the pathways in a controlled manner. Here, we demonstrate directed, stepwise retrograde polysynaptic tracing by inducible reconstitution of replication-deficient trans-neuronal pseudorabies virus (PRVΔIE) in the brain. Furthermore, PRVΔIE replication can be temporally restricted to minimize its neurotoxicity. With this tool, we delineate a wiring diagram between the hippocampus and striatum-two major brain systems for learning, memory, and navigation-that consists of projections from specific hippocampal domains to specific striatal areas via distinct intermediate brain regions. Therefore, this inducible PRVΔIE system provides a tool for dissecting polysynaptic circuits underlying complex brain functions.
Collapse
Affiliation(s)
- Wenqin Du
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabeth Li
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Guo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rachel Arano
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yerim Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuh-Tarng Chen
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alyssa Thompson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - So Jung Oh
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Aspen Samuel
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Li
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hassana K. Oyibo
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Wei Xu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
45
|
Nguyen R, Koukoutselos K, Forro T, Ciocchi S. Fear extinction relies on ventral hippocampal safety codes shaped by the amygdala. SCIENCE ADVANCES 2023; 9:eadg4881. [PMID: 37256958 PMCID: PMC10413664 DOI: 10.1126/sciadv.adg4881] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/25/2023] [Indexed: 06/02/2023]
Abstract
Extinction memory retrieval is influenced by spatial contextual information that determines responding to conditioned stimuli (CS). However, it is poorly understood whether contextual representations are imbued with emotional values to support memory selection. Here, we performed activity-dependent engram tagging and in vivo single-unit electrophysiological recordings from the ventral hippocampus (vH) while optogenetically manipulating basolateral amygdala (BLA) inputs during the formation of cued fear extinction memory. During fear extinction when CS acquire safety properties, we found that CS-related activity in the vH reactivated during sleep consolidation and was strengthened upon memory retrieval. Moreover, fear extinction memory was facilitated when the extinction context exhibited precise coding of its affective zones. Last, these activity patterns along with the retrieval of the fear extinction memory were dependent on glutamatergic transmission from the BLA during extinction learning. Thus, fear extinction memory relies on the formation of contextual and stimulus safety representations in the vH instructed by the BLA.
Collapse
Affiliation(s)
| | | | - Thomas Forro
- Laboratory of Systems Neuroscience, Department of Physiology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
46
|
Biane JS, Ladow MA, Stefanini F, Boddu SP, Fan A, Hassan S, Dundar N, Apodaca-Montano DL, Zhou LZ, Fayner V, Woods NI, Kheirbek MA. Neural dynamics underlying associative learning in the dorsal and ventral hippocampus. Nat Neurosci 2023; 26:798-809. [PMID: 37012382 PMCID: PMC10448873 DOI: 10.1038/s41593-023-01296-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/07/2023] [Indexed: 04/05/2023]
Abstract
Animals associate cues with outcomes and update these associations as new information is presented. This requires the hippocampus, yet how hippocampal neurons track changes in cue-outcome associations remains unclear. Using two-photon calcium imaging, we tracked the same dCA1 and vCA1 neurons across days to determine how responses evolve across phases of odor-outcome learning. Initially, odors elicited robust responses in dCA1, whereas, in vCA1, odor responses primarily emerged after learning and embedded information about the paired outcome. Population activity in both regions rapidly reorganized with learning and then stabilized, storing learned odor representations for days, even after extinction or pairing with a different outcome. Additionally, we found stable, robust signals across CA1 when mice anticipated outcomes under behavioral control but not when mice anticipated an inescapable aversive outcome. These results show how the hippocampus encodes, stores and updates learned associations and illuminates the unique contributions of dorsal and ventral hippocampus.
Collapse
Affiliation(s)
- Jeremy S Biane
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Max A Ladow
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Fabio Stefanini
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY, USA
| | - Sayi P Boddu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Austin Fan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Shazreh Hassan
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Naz Dundar
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel L Apodaca-Montano
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lexi Zichen Zhou
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Varya Fayner
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nicholas I Woods
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
47
|
Dos Santos MB, de Oliveira Guarnieri L, Lunardi P, Schenatto Pereira G. On the effect of social cue valence in contextual memory persistence. Behav Brain Res 2023; 447:114398. [PMID: 36966939 DOI: 10.1016/j.bbr.2023.114398] [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: 01/23/2023] [Revised: 02/13/2023] [Accepted: 03/10/2023] [Indexed: 04/15/2023]
Abstract
Social cues are valuable sensorial stimuli to the acquisition and retrieval of contextual memories. Here, we asked whether the valence of social cues would impact the formation of contextual memories. Adult male C57/BL6 mice were exposed to either conditioned place preference (CPP) or avoidance (CPA). As positive stimuli we used social interaction with a female (IF), while interaction with a male CD1 mice (IM) was used as negative stimulus. Contextual memory was tested 24 h and 7 days after conditioning. Aggressive behavior of CD1, as well as interaction with the female were quantified along the conditioning sessions. IM, but not IF, was salient enough to induce contextual memory estimated by the difference between the time in the conditioned context during test and habituation. Next, we chose two odors with innate behavioral responses and opposite valence to narrow down the sociability to one of its sensorial sources of information - the olfaction. We used urine from females in proestrus (U) and 2,4,5-trimethyl thiazoline (TMT), a predator odor. TMT decreased and U increased the time in the conditioned context during the test performed 24 h and 7 days after conditioning. Taken together, our results suggest that contextual memories conditioned to social encounters are difficult to stablish in mice, specially the one with positive valence. On the other hand, using odors with ecological relevance is a promising strategy to study long-term contextual memories with opposite valences. Ultimately, the behavioral protocol proposed here offers the advantage of studying contextual memories with opposite valences using unconditioned stimulus from the same sensorial category such as olfaction.
Collapse
Affiliation(s)
- Matheus Barbosa Dos Santos
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leonardo de Oliveira Guarnieri
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Paula Lunardi
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Grace Schenatto Pereira
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
48
|
Ito W, Palmer AJ, Morozov A. Social Synchronization of Conditioned Fear in Mice Requires Ventral Hippocampus Input to the Amygdala. Biol Psychiatry 2023; 93:322-330. [PMID: 36244803 PMCID: PMC10069289 DOI: 10.1016/j.biopsych.2022.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/17/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Social organisms synchronize behaviors as an evolutionary-conserved means of thriving. Synchronization under threat, in particular, benefits survival and occurs across species, including humans, but the underlying mechanisms remain unknown because of the scarcity of relevant animal models. Here, we developed a rodent paradigm in which mice synchronized a classically conditioned fear response and identified an underlying neuronal circuit. METHODS Male and female mice were trained individually using auditory fear conditioning and then tested 24 hours later as dyads while allowing unrestricted social interaction during exposure to the conditioned stimulus under visible or infrared illumination to eliminate visual cues. The synchronization of the immobility or freezing bouts was quantified by calculating the effect size Cohen's d for the difference between the actual freezing time overlap and the overlap by chance. The inactivation of the dorsomedial prefrontal cortex, dorsal hippocampus, or ventral hippocampus was achieved by local infusions of muscimol. The chemogenetic disconnection of the hippocampus-amygdala pathway was performed by expressing hM4D(Gi) in the ventral hippocampal neurons and infusing clozapine N-oxide in the amygdala. RESULTS Mice synchronized cued but not contextual fear. It was higher in males than in females and attenuated in the absence of visible light. Inactivation of the ventral but not dorsal hippocampus or dorsomedial prefrontal cortex abolished fear synchronization. Finally, the disconnection of the hippocampus-amygdala pathway diminished fear synchronization. CONCLUSIONS Mice synchronize expression of conditioned fear relying on the ventral hippocampus-amygdala pathway, suggesting that the hippocampus transmits social information to the amygdala to synchronize threat response.
Collapse
Affiliation(s)
- Wataru Ito
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Neurobiology Research, Roanoke, Virginia.
| | - Alexander J Palmer
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Neurobiology Research, Roanoke, Virginia
| | - Alexei Morozov
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Neurobiology Research, Roanoke, Virginia; Carilion Clinic Department of Psychiatry and Behavioral Medicine, Roanoke, Virginia.
| |
Collapse
|
49
|
Xu F, Cong P, Zhang B, Dong H, Zuo W, Wu T, Tian L, Xiong L. A decrease in NR2B expression mediated by DNA hypermethylation induces perioperative neurocognitive disorder in aged mice. CNS Neurosci Ther 2023; 29:1229-1242. [PMID: 36694341 PMCID: PMC10068472 DOI: 10.1111/cns.14097] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
AIMS This study was designed to investigate the role of NR2B and the contribution of DNA methylation to NR2B expression in the pathogenesis of PND. METHODS Eighteen-month-old C57BL/6J mice were subjected to experimental laparotomy under 1.4% isoflurane anesthesia. Hippocampus-dependent learning and memory were evaluated by using the Barnes maze and contextual fear conditioning tests. The protein and mRNA expression levels of NR2B were evaluated by western blotting and qRT-PCR respectively, and the methylation of the NR2B gene was examined by using targeted bisulfite sequencing. Long-term synaptic plasticity (LTP) was measured by electrophysiology. RESULTS Mice that underwent laparotomy exhibited hippocampus-dependent cognitive deficits accompanied by decreased NR2B expressions and LTP deficiency. The overexpression of NR2B in the dorsal hippocampus could improve learning and memory in mice subjected to laparotomy. In particular, the decreased NR2B expressions induced by laparotomy was attributed to the NR2B gene hypermethylation. Preoperative administration of S-adenosylmethionine (SAM) could hypomethylate the NR2B gene, upregulate NR2B expression and improve LTP, exerting a dose-dependent therapeutic effect against PND. Moreover, inhibiting NR2B abrogated the benefits of SAM pretreatment. CONCLUSIONS Laparotomy cause hippocampus-dependent cognitive decline by hypermethylating the NR2B gene, allowing us to understand the pathogenesis of PND in an epigenetic landscape.
Collapse
Affiliation(s)
- Feifei Xu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peilin Cong
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence Affiliated to Tongji University School of Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Bingqian Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tingmei Wu
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence Affiliated to Tongji University School of Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Li Tian
- Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence Affiliated to Tongji University School of Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| | - Lize Xiong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Translational Research Institute of Brain and Brain-Like Intelligence Affiliated to Tongji University School of Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Shanghai, China
| |
Collapse
|
50
|
Massi L, Hagihara KM, Courtin J, Hinz J, Müller C, Fustiñana MS, Xu C, Karalis N, Lüthi A. Disynaptic specificity of serial information flow for conditioned fear. SCIENCE ADVANCES 2023; 9:eabq1637. [PMID: 36652513 PMCID: PMC10957099 DOI: 10.1126/sciadv.abq1637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Memory encoding and retrieval rely on specific interactions across multiple brain areas. Although connections between individual brain areas have been extensively studied, the anatomical and functional specificity of neuronal circuit organization underlying information transfer across multiple brain areas remains unclear. Here, we combine transsynaptic viral tracing, optogenetic manipulations, and calcium dynamics recordings to dissect the multisynaptic functional connectivity of the amygdala. We identify a distinct basolateral amygdala (BLA) subpopulation that connects disynaptically to the periaqueductal gray (PAG) via the central amygdala (CeA). This disynaptic pathway serves as a core circuit element necessary for the learning and expression of conditioned fear and exhibits learning-related plasticity. Together, our findings demonstrate the utility of multisynaptic approaches for functional circuit analysis and indicate that disynaptic specificity may be a general feature of neuronal circuit organization.
Collapse
Affiliation(s)
- Léma Massi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Kenta M. Hagihara
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| | - Julien Courtin
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Julian Hinz
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| | - Christian Müller
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Maria Sol Fustiñana
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Chun Xu
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nikolaos Karalis
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|