1
|
Deng Z, Fei X, Zhang S, Xu M. A time window for memory consolidation during NREM sleep revealed by cAMP oscillation. Neuron 2025:S0896-6273(25)00220-X. [PMID: 40233747 DOI: 10.1016/j.neuron.2025.03.020] [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: 08/22/2024] [Revised: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025]
Abstract
Memory formation requires specific neural activity in coordination with intracellular signaling mediated by second messengers such as cyclic adenosine monophosphate (cAMP). However, the real-time dynamics of cAMP remain largely unknown. Here, using a genetically encoded cAMP sensor with high temporal resolution, we found neural-activity-dependent rapid cAMP elevation during learning. Interestingly, in slow-wave sleep, during which memory consolidation occurs, the cAMP level in mice was anti-correlated with neural activity and exhibited norepinephrine β1 receptor-dependent infra-slow oscillations that were synchronized across the hippocampus and cortex. Furthermore, the hippocampal-cortical interactions increased during the narrow time-window of the peak cAMP level; suppressing hippocampal activity specifically during this window impaired spatial memory consolidation. Thus, hippocampal-dependent memory consolidation occurs within a specific time window of high cAMP activity during slow-wave sleep.
Collapse
Affiliation(s)
- Ziru Deng
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiang Fei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
2
|
Chong YS, Ang SR, Sajikumar S. Beyond boundaries: extended temporal flexibility in synaptic tagging and capture. Commun Biol 2025; 8:553. [PMID: 40181131 PMCID: PMC11968991 DOI: 10.1038/s42003-025-07998-w] [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: 11/06/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
Synaptic tagging and capture (STC) is a mechanism that enables the formation of associative synaptic plasticity by marking activated synapses with "tags" to capture plasticity-related products (PRPs) essential for plasticity stabilization. Experimental evidence using long-term potentiation (LTP), a widely studied cellular correlate of memory, shows that the duration of synaptic tags varies, lasting up to 90 minutes in ex vivo hippocampal slices but shorter in in vivo conditions, likely due to higher metabolic activity. In this study, we investigate the time window for tag-PRP interactions in STC using a strong-before-weak paradigm, where protein synthesis-dependent late-LTP precedes protein synthesis-independent early-LTP at various intervals. Surprisingly, successful STC is observed even with a 9-hour interval in the strong-before-weak paradigm, suggesting a broader temporal flexibility for tag-PRP interactions than previously understood. This unexpected finding offers alternative explanations for associative memory formation by cataloguing memory events, allowing weaker memories to be strengthened when preceded by stronger ones.
Collapse
Affiliation(s)
- Yee Song Chong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Sheila Ruixia Ang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Life Sciences Institute Neurobiology Programme, National University of Singapore, Singapore, Singapore.
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Gielow MR, Headley DB, Herzallah MM, Paré D. Common Neocortical and Hippocampal Correlates of Performance Errors in a Timing Task. J Neurosci 2025; 45:e2003232024. [PMID: 39794130 PMCID: PMC11823390 DOI: 10.1523/jneurosci.2003-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
We aimed to identify the neuronal correlates of performance errors in a difficult timing task. Male rats were trained to seek rewards and avoid shocks depending on the position of photic conditioned stimuli (CS-R and CS-S, respectively). Then, they were exposed to conflict trials where they had to time the interval between the CS-R and CS-S to obtain rewards while avoiding footshocks. There were pronounced individual differences in behavioral strategies on conflict trials. When presented with a CS-S, some rats quickly left the shock sector, forsaking the option of earning a reward, and rarely got shocked. Others earned rewards by delaying avoidance based on the interval between the CS-R and CS-S but were shocked more often. The probability rats would fail a given trial was not stable across trials as rats engaged in incorrect trial runs that were longer than expected by chance. Since this finding suggested that rats shift between two quasi-stable processing modes, we next examined the neuronal correlates of errors. Incorrect trials coincided with reduced firing rates in CA1 and sensory cortical neurons. Moreover, trial-to-trial variations in the firing rates of simultaneously recorded neurons were more strongly correlated on error than correct trials. Last, the power of low-frequency local field potential oscillations was higher during incorrect trials. The finding that the neuronal correlates of correct and error trials are similar in the hippocampus and neocortex lead us to hypothesize that they depend on changes in the activity of common afferents, such as neuromodulatory inputs.
Collapse
Affiliation(s)
- Matthew R Gielow
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
| | - Drew B Headley
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
| | - Mohammad M Herzallah
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
- Palestinian Neuroscience Initiative, Al-Quds University, Jerusalem, Palestine
| | - Denis Paré
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, New Jersey 07102
| |
Collapse
|
4
|
Zhang Y, Li T, Miao J, Zhang Z, Yang M, Wang Z, Yang B, Zhang J, Li H, Su Q, Guo J. Gamma-glutamyl transferase 5 overexpression in cerebrovascular endothelial cells improves brain pathology, cognition, and behavior in APP/PS1 mice. Neural Regen Res 2025; 20:533-547. [PMID: 38819065 PMCID: PMC11317949 DOI: 10.4103/nrr.nrr-d-23-01525] [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: 09/10/2023] [Revised: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00030/figure1/v/2024-05-28T214302Z/r/image-tiff In patients with Alzheimer's disease, gamma-glutamyl transferase 5 (GGT5) expression has been observed to be downregulated in cerebrovascular endothelial cells. However, the functional role of GGT5 in the development of Alzheimer's disease remains unclear. This study aimed to explore the effect of GGT5 on cognitive function and brain pathology in an APP/PS1 mouse model of Alzheimer's disease, as well as the underlying mechanism. We observed a significant reduction in GGT5 expression in two in vitro models of Alzheimer's disease (Aβ1-42-treated hCMEC/D3 and bEnd.3 cells), as well as in the APP/PS1 mouse model. Additionally, injection of APP/PS1 mice with an adeno-associated virus encoding GGT5 enhanced hippocampal synaptic plasticity and mitigated cognitive deficits. Interestingly, increasing GGT5 expression in cerebrovascular endothelial cells reduced levels of both soluble and insoluble amyloid-β in the brains of APP/PS1 mice. This effect may be attributable to inhibition of the expression of β-site APP cleaving enzyme 1, which is mediated by nuclear factor-kappa B. Our findings demonstrate that GGT5 expression in cerebrovascular endothelial cells is inversely associated with Alzheimer's disease pathogenesis, and that GGT5 upregulation mitigates cognitive deficits in APP/PS1 mice. These findings suggest that GGT5 expression in cerebrovascular endothelial cells is a potential therapeutic target and biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi Province, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhina Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mingxuan Yang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhuoran Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Yang
- Department of Hernia and Abdominal Wall Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jiawei Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Haiting Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi Province, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
5
|
Haugland KG, Jordbræk SV, Knutsen E, Kjelstrup KB, Brun VH. Growth Hormone Alters Remapping in the Hippocampal Area CA1 in a Novel Environment. eNeuro 2025; 12:ENEURO.0237-24.2024. [PMID: 39900507 PMCID: PMC11814925 DOI: 10.1523/eneuro.0237-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Growth hormone (GH) is a neuromodulator that binds to receptors in the hippocampus and alters synaptic plasticity. A decline in GH levels is associated with normal aging, stress, and disease, and the mechanisms proposed involve the hippocampal circuit plasticity. To see how GH affects the hippocampal neural code, we recorded single neurons in the CA1 region of male Long-Evans rats with locally altered GH levels. Rats received injections of adeno-associated viruses into the hippocampus to make the cells overexpress either GH or an antagonizing mutated GH (aGH). Place cells were recorded in both familiar and novel environments to allow the assessment of pattern separation in the neural representations termed remapping. All the animals showed intact and stable place fields in the familiar environment. In the novel environment, aGH transfection increased the average firing rate, peak rate, and information density of the CA1 place fields. The tendency of global remapping increased in the GH animals compared with the controls, and only place cells of control animals showed significant rate remapping. Our results suggest that GH increases hippocampal sensitivity to novel information. Our findings show that GH is a significant neuromodulator in the hippocampus affecting how place cells represent the environment. These results could help us to understand the mechanisms behind memory impairments in GH deficiency as well as in normal aging.
Collapse
Affiliation(s)
- Kamilla G Haugland
- Departments of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø 9019, Norway
| | | | - Erik Knutsen
- Medical Biology, UiT - The Arctic University of Norway, Tromsø 9019, Norway
| | - Kirsten B Kjelstrup
- Departments of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø 9019, Norway
- University Hospital of North Norway, Tromsø 9019, Norway
| | - Vegard H Brun
- Departments of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø 9019, Norway
- University Hospital of North Norway, Tromsø 9019, Norway
| |
Collapse
|
6
|
Tessereau C, Xuan F, Mellor JR, Dayan P, Dombeck D. Navigating uncertainty: reward location variability induces reorganization of hippocampal spatial representations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631465. [PMID: 39829917 PMCID: PMC11741294 DOI: 10.1101/2025.01.06.631465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Navigating uncertainty is crucial for survival, with the location and availability of reward varying in different and unsignalled ways. Hippocampal place cell populations over-represent salient locations in an animal's environment, including those associated with rewards; however, how the spatial uncertainties impact the cognitive map is unclear. We report a virtual spatial navigation task designed to test the impact of different levels and types of uncertainty about reward on place cell populations. When the reward location changed on a trial-by-trial basis, inducing expected uncertainty, a greater proportion of place cells followed along, and the reward and the track end became anchors of a warped spatial metric. When the reward location then unexpectedly moved, the fraction of reward place cells that followed was greater when starting from a state of expected, compared to low, uncertainty. Overall, we show that different forms of potentially interacting uncertainty generate remapping in parallel, task-relevant, reference frames.
Collapse
Affiliation(s)
| | - Feng Xuan
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Jack, R. Mellor
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Peter Dayan
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- University of Tübingen, Tübingen, Germany
| | - Daniel Dombeck
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
7
|
Hiraga T, Hata T, Soya S, Shimoda R, Takahashi K, Soya M, Inoue K, Johansen JP, Okamoto M, Soya H. Light-exercise-induced dopaminergic and noradrenergic stimulation in the dorsal hippocampus: Using a rat physiological exercise model. FASEB J 2024; 38:e70215. [PMID: 39668509 PMCID: PMC11638517 DOI: 10.1096/fj.202400418rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Exercise activates the dorsal hippocampus that triggers synaptic and cellar plasticity and ultimately promotes memory formation. For decades, these benefits have been explored using demanding and stress-response-inducing exercise at moderate-to-vigorous intensities. In contrast, our translational research with animals and humans has focused on light-intensity exercise (light exercise) below the lactate threshold (LT), which almost anyone can safely perform with minimal stress. We found that even light exercise can stimulate hippocampal activity and enhance memory performance. Although the circuit mechanism of this boost remains unclear, arousal promotion even with light exercise implies the involvement of the ascending monoaminergic system that is essential to modulate hippocampal activity and impact memory. To test this hypothesis, we employed our physiological exercise model based on the LT of rats and immunohistochemically assessed the neuronal activation of the dorsal hippocampal sub-regions and brainstem monoaminergic neurons. Also, we monitored the extracellular concentration of monoamines in the dorsal hippocampus using in vivo microdialysis. We found that even light exercise increased neuronal activity in the dorsal hippocampal sub-regions and elevated the extracellular concentrations of noradrenaline and dopamine. Furthermore, we found that tyrosine hydroxylase-positive neurons in the locus coeruleus (LC) and the ventral tegmental area (VTA) were activated even by light exercise and were both positively correlated with the dorsal hippocampal activation. In conclusion, our findings demonstrate that light exercise stimulates dorsal hippocampal neurons, which are associated with LC-noradrenergic and VTA-dopaminergic activation. This shed light on the circuit mechanisms responsible for hippocampal neural activation during exercise, consequently enhancing memory function.
Collapse
Affiliation(s)
- Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Shingo Soya
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaJapan
- Department of Molecular Behavioral Physiology, Institute of MedicineUniversity of TsukubaTsukubaJapan
| | - Ryo Shimoda
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Kanako Takahashi
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Mariko Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Department of Anatomy and Neuroscience, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshiro Inoue
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Center for Education in Liberal Arts and SciencesHealth Sciences University of HokkaidoIshikariJapan
| | - Joshua P. Johansen
- Laboratory for Neural Circuitry of MemoryRIKEN Center for Brain ScienceSaitamaJapan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| |
Collapse
|
8
|
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
|
9
|
Graf J, Samiee A, Flossmann T, Holthoff K, Kirmse K. Chemogenetic silencing reveals presynaptic G i/o protein-mediated inhibition of developing hippocampal synchrony in vivo. iScience 2024; 27:110997. [PMID: 39429781 PMCID: PMC11489827 DOI: 10.1016/j.isci.2024.110997] [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: 03/28/2024] [Revised: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Recent advances in understanding how neuronal activity shapes developing brain circuits increasingly rely on Gi/o-dependent inhibitory chemogenetic tools (Gi-DREADDs). However, their mechanisms of action and efficacy in neurons with immature Gi/o signaling are elusive. Here, we express the Gi-DREADD hM4Di in glutamatergic telencephalic neurons and analyze its impact on CA1 pyramidal neurons in neonatal mice. Using acousto-optic two-photon Ca2+ imaging, we report that activation of hM4Di leads to a complete arrest of spontaneous synchrony in CA1 in vitro. We demonstrate that hM4Di does not cause somatic hyperpolarization or shunting but rather mediates presynaptic silencing of glutamatergic neurotransmission. In vivo, inhibition through hM4Di potently suppresses early sharp waves (eSPWs) and discontinuous oscillatory network activity in CA1 of head-fixed mice before eye opening. Our findings provide insights into the role of Gi/o signaling in synchronized activity in the neonatal hippocampus and bear relevance for applying chemogenetic silencing at early developmental stages.
Collapse
Affiliation(s)
- Jürgen Graf
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Arash Samiee
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Tom Flossmann
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Institute of Physiology I, Jena University Hospital, 07743 Jena, Germany
| | - Knut Holthoff
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Knut Kirmse
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| |
Collapse
|
10
|
Dundon NM, Stuber A, Bullock T, Garcia JO, Babenko V, Rizor E, Yang D, Giesbrecht B, Grafton ST. Cardiac-Sympathetic Contractility and Neural Alpha-Band Power: Cross-Modal Collaboration during Approach-Avoidance Conflict. J Neurosci 2024; 44:e2008232024. [PMID: 39214705 PMCID: PMC11466073 DOI: 10.1523/jneurosci.2008-23.2024] [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: 10/24/2023] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
As evidence mounts that the cardiac-sympathetic nervous system reacts to challenging cognitive settings, we ask if these responses are epiphenomenal companions or if there is evidence suggesting a more intertwined role of this system with cognitive function. Healthy male and female human participants performed an approach-avoidance paradigm, trading off monetary reward for painful electric shock, while we recorded simultaneous electroencephalographic and cardiac-sympathetic signals. Participants were reward sensitive but also experienced approach-avoidance "conflict" when the subjective appeal of the reward was near equivalent to the revulsion of the cost. Drift-diffusion model parameters suggested that participants managed conflict in part by integrating larger volumes of evidence into choices (wider decision boundaries). Late alpha-band (neural) dynamics were consistent with widening decision boundaries serving to combat reward sensitivity and spread attention more fairly to all dimensions of available information. Independently, wider boundaries were also associated with cardiac "contractility" (an index of sympathetically mediated positive inotropy). We also saw evidence of conflict-specific "collaboration" between the neural and cardiac-sympathetic signals. In states of high conflict, the alignment (i.e., product) of alpha dynamics and contractility were associated with a further widening of the boundary, independent of either signal's singular association. Cross-trial coherence analyses provided additional evidence that the autonomic systems controlling cardiac-sympathetics might influence the assessment of information streams during conflict by disrupting or overriding reward processing. We conclude that cardiac-sympathetic control might play a critical role, in collaboration with cognitive processes, during the approach-avoidance conflict in humans.
Collapse
Affiliation(s)
- Neil M Dundon
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University of Freiburg, Freiburg 79104, Germany
| | - Alexander Stuber
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
| | - Tom Bullock
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
| | - Javier O Garcia
- Humans in Complex Systems Division, US DEVCOM Army Research Laboratory, Aberdeen Proving Ground, Maryland 21005
| | - Viktoriya Babenko
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- BIOPAC Systems Inc., Goleta, California 93117
| | - Elizabeth Rizor
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Interdepartmental Graduate Program in Dynamical Neuroscience, University of California, Santa Barbara, California 93106
| | - Dengxian Yang
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Department of Computer Science, University of California, Santa Barbara, California 93106
| | - Barry Giesbrecht
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Interdepartmental Graduate Program in Dynamical Neuroscience, University of California, Santa Barbara, California 93106
| | - Scott T Grafton
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
| |
Collapse
|
11
|
Benatti BM, Adiletta A, Sgadò P, Malgaroli A, Ferro M, Lamanna J. Epigenetic Modifications and Neuroplasticity in the Pathogenesis of Depression: A Focus on Early Life Stress. Behav Sci (Basel) 2024; 14:882. [PMID: 39457754 PMCID: PMC11504006 DOI: 10.3390/bs14100882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Major depressive disorder (MDD) is a debilitating mental illness, and it is considered to be one of the leading causes of disability globally. The etiology of MDD is multifactorial, involving an interplay between biological, psychological, and social factors. Early life represents a critical period for development. Exposure to adverse childhood experiences is a major contributor to the global burden of disease and disability, doubling the risk of developing MDD later in life. Evidence suggests that stressful events experienced during that timeframe play a major role in the emergence of MDD, leading to epigenetic modifications, which might, in turn, influence brain structure, function, and behavior. Neuroplasticity seems to be a primary pathogenetic mechanism of MDD, and, similarly to epigenetic mechanisms, it is particularly sensitive to stress in the early postnatal period. In this review, we will collect and discuss recent studies supporting the role of epigenetics and neuroplasticity in the pathogenesis of MDD, with a focus on early life stress (ELS). We believe that understanding the epigenetic mechanisms by which ELS affects neuroplasticity offers potential pathways for identifying novel therapeutic targets for MDD, ultimately aiming to improve treatment outcomes for this debilitating disorder.
Collapse
Affiliation(s)
- Bianca Maria Benatti
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
| | - Alice Adiletta
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Paola Sgadò
- Center for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (A.A.); (P.S.)
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Faculty of Psychology, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| | - Mattia Ferro
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Department of Psychology, Sigmund Freud Private University, 20143 Milan, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, 20132 Milan, Italy; (B.M.B.); (M.F.)
- Clinical Center Tourette Syndrome, IRCCS Ospedale San Raffaele, 20127 Milan, Italy
| |
Collapse
|
12
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [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: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
13
|
Ball JD, Davies A, Gurung D, Mankoo A, Panerai R, Minhas JS, Robinson T, Beishon L. The effect of posture on the age dependence of neurovascular coupling. Physiol Rep 2024; 12:e70031. [PMID: 39218618 PMCID: PMC11366444 DOI: 10.14814/phy2.70031] [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: 01/19/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Previous studies report contradicting age-related neurovascular coupling (NVC). Few studies assess postural effects, but less investigate relationships between age and NVC within different postures. Therefore, this study investigated the effect of age on NVC in different postures with varying cognitive stimuli. Beat-to-beat blood pressure, heart rate and end-tidal carbon dioxide were assessed alongside middle and posterior cerebral artery velocities (MCAv and PCAv, respectively) using transcranial Doppler ultrasonography in 78 participants (31 young-, 23 middle- and 24 older-aged) with visuospatial (VST) and attention tasks (AT) in various postures at two timepoints (T2 and T3). Between-group significance testing utilized one-way analysis-of-variance (ANOVA) (Tukey post-hoc). Mixed three-way/one-way ANOVAs explored task, posture, and age interactions. Significant effects of posture on NVC were driven by a 3.8% increase from seated to supine. For AT, mean supine %MCAv increase was greatest in younger (5.44%) versus middle (0.12%) and older-age (0.09%) at T3 (p = 0.005). For VST, mean supine %PCAv increase was greatest at T2 and T3 in middle (10.99%/10.12%) and older-age (17.36%/17.26%) versus younger (9.44%/8.89%) (p = 0.004/p = 0.002). We identified significant age-related NVC effects with VST-induced hyperactivation. This may reflect age-related compensatory processes in supine. Further work is required, using complex stimuli while standing/walking, examining NVC, aging and falls.
Collapse
Affiliation(s)
- James D. Ball
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Aaron Davies
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Dewakar Gurung
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Alex Mankoo
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Ronney Panerai
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Jatinder S. Minhas
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Thompson Robinson
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
- NIHR Leicester Biomedical Research Centre, British Heart Foundation Cardiovascular Research CentreGlenfield HospitalLeicesterUK
| | - Lucy Beishon
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| |
Collapse
|
14
|
Abdulmalek S, Connole LM, O'Sullivan NC, Beyna M, Pangalos MN, von Schack D, Ring RH, Murphy KJ. Midkine is upregulated in the hippocampus following both spatial and olfactory reward association learning and enhances memory. J Neurochem 2024; 168:2832-2847. [PMID: 39361112 DOI: 10.1111/jnc.16151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 10/06/2024]
Abstract
Hippocampal neuronal plasticity is a fundamental process underpinning learning and memory formation and requiring elaborate molecular mechanisms that result in the dynamic remodelling of synaptic connectivity. The neurotrophic properties of midkine (Mdk) have been implicated in the development and repair of the nervous system, while Mdk knockout resulted in deficits in the formation of certain types of memory. The role of Mdk in the process of memory-associated neuronal plasticity, however, remains poorly understood. We investigated the learning-induced regulation of Mdk in spatial navigation and association learning using the water maze and the odour reward association learning paradigms, characterising a temporal profile of Mdk protein expression post-learning. Both learning events revealed similar patterns of upregulation of expression of the protein in the rat hippocampal dentate gyrus, which were rapid and transient. Moreover, administration of recombinant Mdk during the endogenous Mdk upregulation following learning enhanced memory in the water maze task revealing a pro-cognitive action of Mdk. We further show that, within the adult hippocampus, Mdk mRNA is predominantly expressed in granular and pyramidal neurons and that hippocampal neuronal Mdk expression is regulated by the canonical plasticity-associated neurotransmitter glutamate. Finally, we confirm that the positive action of Mdk on neurite outgrowth previously noted in cortical and cerebellar neurons extends to hippocampal neurons. Together, our findings suggest a role for Mdk in glutamate-mediated hippocampal neuronal plasticity important for long-term memory consolidation.
Collapse
Affiliation(s)
- Sarah Abdulmalek
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Laura M Connole
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Niamh C O'Sullivan
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Mercedes Beyna
- Inflammation Research Unit, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, USA
| | | | - David von Schack
- Inflammation Research Unit, Pfizer Worldwide Research & Development, Cambridge, Massachusetts, USA
| | | | - Keith J Murphy
- Neurotherapeutics Research Group, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Roth RH, Ding JB. Cortico-basal ganglia plasticity in motor learning. Neuron 2024; 112:2486-2502. [PMID: 39002543 PMCID: PMC11309896 DOI: 10.1016/j.neuron.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 07/15/2024]
Abstract
One key function of the brain is to control our body's movements, allowing us to interact with the world around us. Yet, many motor behaviors are not innate but require learning through repeated practice. Among the brain's motor regions, the cortico-basal ganglia circuit is particularly crucial for acquiring and executing motor skills, and neuronal activity in these regions is directly linked to movement parameters. Cell-type-specific adaptations of activity patterns and synaptic connectivity support the learning of new motor skills. Functionally, neuronal activity sequences become structured and associated with learned movements. On the synaptic level, specific connections become potentiated during learning through mechanisms such as long-term synaptic plasticity and dendritic spine dynamics, which are thought to mediate functional circuit plasticity. These synaptic and circuit adaptations within the cortico-basal ganglia circuitry are thus critical for motor skill acquisition, and disruptions in this plasticity can contribute to movement disorders.
Collapse
Affiliation(s)
- Richard H Roth
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Bin Ibrahim MZ, Wang Z, Sajikumar S. Synapses tagged, memories kept: synaptic tagging and capture hypothesis in brain health and disease. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230237. [PMID: 38853570 PMCID: PMC11343274 DOI: 10.1098/rstb.2023.0237] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 06/11/2024] Open
Abstract
The synaptic tagging and capture (STC) hypothesis lays the framework on the synapse-specific mechanism of protein synthesis-dependent long-term plasticity upon synaptic induction. Activated synapses will display a transient tag that will capture plasticity-related products (PRPs). These two events, tag setting and PRP synthesis, can be teased apart and have been studied extensively-from their electrophysiological and pharmacological properties to the molecular events involved. Consequently, the hypothesis also permits interactions of synaptic populations that encode different memories within the same neuronal population-hence, it gives rise to the associativity of plasticity. In this review, the recent advances and progress since the experimental debut of the STC hypothesis will be shared. This includes the role of neuromodulation in PRP synthesis and tag integrity, behavioural correlates of the hypothesis and modelling in silico. STC, as a more sensitive assay for synaptic health, can also assess neuronal aberrations. We will also expound how synaptic plasticity and associativity are altered in ageing-related decline and pathological conditions such as juvenile stress, cancer, sleep deprivation and Alzheimer's disease. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Mohammad Zaki Bin Ibrahim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
| | - Zijun Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore119077, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117597, Singapore
| |
Collapse
|
17
|
Copenhaver AE, LeGates TA. Sex-Specific Mechanisms Underlie Long-Term Potentiation at Hippocampus→Medium Spiny Neuron Synapses in the Medial Shell of the Nucleus Accumbens. J Neurosci 2024; 44:e0100242024. [PMID: 38806250 PMCID: PMC11223474 DOI: 10.1523/jneurosci.0100-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp→NAc synapses is rewarding, and mice can establish learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigated sex differences in the mechanisms underlying Hipp→NAc LTP using whole-cell electrophysiology and pharmacology. We observed similarities in basal synaptic strength between males and females and found that LTP occurs postsynaptically with similar magnitudes in both sexes. However, key sex differences emerged as LTP in males required NMDA receptors (NMDAR), whereas LTP in females utilized an NMDAR-independent mechanism involving L-type voltage-gated Ca2+ channels (VGCCs) and estrogen receptor α (ERα). We also uncovered sex-similar features as LTP in both sexes depended on CaMKII activity and occurred independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders.
Collapse
Affiliation(s)
- Ashley E Copenhaver
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
| | - Tara A LeGates
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
18
|
Tamboli S, Singh S, Topolnik D, El Amine Barkat M, Radhakrishnan R, Guet-McCreight A, Topolnik L. Mouse hippocampal CA1 VIP interneurons detect novelty in the environment and support recognition memory. Cell Rep 2024; 43:114115. [PMID: 38607918 DOI: 10.1016/j.celrep.2024.114115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/17/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
In the CA1 hippocampus, vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) play a prominent role in disinhibitory circuit motifs. However, the specific behavioral conditions that lead to circuit disinhibition remain uncertain. To investigate the behavioral relevance of VIP-IN activity, we employed wireless technologies allowing us to monitor and manipulate their function in freely behaving mice. Our findings reveal that, during spatial exploration in new environments, VIP-INs in the CA1 hippocampal region become highly active, facilitating the rapid encoding of novel spatial information. Remarkably, both VIP-INs and pyramidal neurons (PNs) exhibit increased activity when encountering novel changes in the environment, including context- and object-related alterations. Concurrently, somatostatin- and parvalbumin-expressing inhibitory populations show an inverse relationship with VIP-IN and PN activity, revealing circuit disinhibition that occurs on a timescale of seconds. Thus, VIP-IN-mediated disinhibition may constitute a crucial element in the rapid encoding of novelty and the acquisition of recognition memory.
Collapse
Affiliation(s)
- Suhel Tamboli
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Sanjay Singh
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Dimitry Topolnik
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Mohamed El Amine Barkat
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | - Risna Radhakrishnan
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada
| | | | - Lisa Topolnik
- Neuroscience Axis, CRCHUQ-CHUL, Quebec City, PQ, Canada; Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Quebec City, PQ, Canada.
| |
Collapse
|
19
|
Cabrera Y, Koymans KJ, Poe GR, Kessels HW, Van Someren EJW, Wassing R. Overnight neuronal plasticity and adaptation to emotional distress. Nat Rev Neurosci 2024; 25:253-271. [PMID: 38443627 DOI: 10.1038/s41583-024-00799-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
Expressions such as 'sleep on it' refer to the resolution of distressing experiences across a night of sound sleep. Sleep is an active state during which the brain reorganizes the synaptic connections that form memories. This Perspective proposes a model of how sleep modifies emotional memory traces. Sleep-dependent reorganization occurs through neurophysiological events in neurochemical contexts that determine the fates of synapses to grow, to survive or to be pruned. We discuss how low levels of acetylcholine during non-rapid eye movement sleep and low levels of noradrenaline during rapid eye movement sleep provide a unique window of opportunity for plasticity in neuronal representations of emotional memories that resolves the associated distress. We integrate sleep-facilitated adaptation over three levels: experience and behaviour, neuronal circuits, and synaptic events. The model generates testable hypotheses for how failed sleep-dependent adaptation to emotional distress is key to mental disorders, notably disorders of anxiety, depression and post-traumatic stress with the common aetiology of insomnia.
Collapse
Affiliation(s)
- Yesenia Cabrera
- Department of Integrative Biology and Physiology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Karin J Koymans
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Gina R Poe
- Department of Integrative Biology and Physiology, Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- Department of Synaptic Plasticity and Behaviour, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Society for Arts and Sciences, Amsterdam, Netherlands
| | - Eus J W Van Someren
- Department of Sleep and Cognition, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Society for Arts and Sciences, Amsterdam, Netherlands
- Department of Integrative Neurophysiology and Psychiatry, VU University, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, VU University, Amsterdam UMC, Amsterdam, Netherlands
| | - Rick Wassing
- Sleep and Circadian Research, Woolcock Institute of Medical Research, Macquarie University, Sydney, New South Wales, Australia.
- School of Psychological Sciences, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia.
- Sydney Local Health District, Sydney, New South Wales, Australia.
| |
Collapse
|
20
|
Tian Y, Murphy MJH, Steiner LA, Kalia SK, Hodaie M, Lozano AM, Hutchison WD, Popovic MR, Milosevic L, Lankarany M. Modeling Instantaneous Firing Rate of Deep Brain Stimulation Target Neuronal Ensembles in the Basal Ganglia and Thalamus. Neuromodulation 2024; 27:464-475. [PMID: 37140523 DOI: 10.1016/j.neurom.2023.03.012] [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: 11/13/2022] [Revised: 01/27/2023] [Accepted: 03/02/2023] [Indexed: 05/05/2023]
Abstract
OBJECTIVE Deep brain stimulation (DBS) is an effective treatment for movement disorders, including Parkinson disease and essential tremor. However, the underlying mechanisms of DBS remain elusive. Despite the capability of existing models in interpreting experimental data qualitatively, there are very few unified computational models that quantitatively capture the dynamics of the neuronal activity of varying stimulated nuclei-including subthalamic nucleus (STN), substantia nigra pars reticulata (SNr), and ventral intermediate nucleus (Vim)-across different DBS frequencies. MATERIALS AND METHODS Both synthetic and experimental data were used in the model fitting; the synthetic data were generated by an established spiking neuron model that was reported in our previous work, and the experimental data were provided using single-unit microelectrode recordings (MERs) during DBS (microelectrode stimulation). Based on these data, we developed a novel mathematical model to represent the firing rate of neurons receiving DBS, including neurons in STN, SNr, and Vim-across different DBS frequencies. In our model, the DBS pulses were filtered through a synapse model and a nonlinear transfer function to formulate the firing rate variability. For each DBS-targeted nucleus, we fitted a single set of optimal model parameters consistent across varying DBS frequencies. RESULTS Our model accurately reproduced the firing rates observed and calculated from both synthetic and experimental data. The optimal model parameters were consistent across different DBS frequencies. CONCLUSIONS The result of our model fitting was in agreement with experimental single-unit MER data during DBS. Reproducing neuronal firing rates of different nuclei of the basal ganglia and thalamus during DBS can be helpful to further understand the mechanisms of DBS and to potentially optimize stimulation parameters based on their actual effects on neuronal activity.
Collapse
Affiliation(s)
- Yupeng Tian
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada
| | | | - Leon A Steiner
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; Berlin Institute of Health, Berlin, Germany; Department of Surgery, University of Toronto, Toronto, ON, Canada; Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Suneil K Kalia
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Mojgan Hodaie
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Andres M Lozano
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - William D Hutchison
- CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Milos R Popovic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Luka Milosevic
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Milad Lankarany
- Krembil Research Institute - University Health Network, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; KITE Research Institute, Toronto Rehabilitation Institute - University Health Network, Toronto, ON, Canada; CRANIA, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Yin X, Liu W, Feng H, Huang J, Wang Q, Zhang Q, He J, Wang R. Bifidobacterium animalis subsp. lactis A6 attenuates hippocampal damage and memory impairments in an ADHD rat model. Food Funct 2024; 15:2668-2678. [PMID: 38374797 DOI: 10.1039/d3fo04665f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Attention deficit hyperactivity disorder (ADHD) is commonly accompanied by learning and memory deficits. This study aimed to demonstrate the effects of probiotic Bifidobacterium animalis subsp. lactis A6 (BAA6) on behaviour and memory function in spontaneously hypertensive rats (SHRs). The results showed that BAA6 treatment ameliorated spatial working memory deficits and inhibited hippocampal neuron loss in SHRs. The levels of neurotransmitters such as acetylcholine, dopamine, and norepinephrine, and the brain derived neurotrophic factor increased and that of glutamate decreased in the brain tissue of SHRs after BAA6 administration. Moreover, BAA6 reduced the levels of pro-inflammatory cytokines TNF-α and IL-1β, and increased the levels of anti-inflammatory IL-10 and antioxidant glutathione in SHRs. 16S rRNA high-throughput sequencing showed that BAA6 treatment changed the gut microbiota composition. BAA6 promoted beneficial Lactobacillus, Romboutsia, Blautia, and Turicibacter, and decreased the enrichment of bacterial genera such as Dietzia, Sporosarcina, Brevibacterium, NK4A214_group, Atopostipes, and Facklamia negatively associated with neurotransmitter release and anti-inflammatory effects in SHRs. Together, these results suggested that BAA6 improved memory function by ameliorating hippocampal damage, abnormal neurotransmitter release and cerebral inflammation by reshaping the gut microbiota in SHRs. This study provides a scientific basis for the development and application of BAA6 as a promising dietary intervention to reduce the risk of ADHD.
Collapse
Affiliation(s)
- Xindi Yin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Weichen Liu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Haihong Feng
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
- Hebei Engineering Research Center of Animal Product, Langfang 065200, China
| | - Jiaqiang Huang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Qi Wang
- Xinjiang Golden Camel Investment Co., Ltd., Wulumuqi 830039, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| | - Jingjing He
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| |
Collapse
|
22
|
Malone TJ, Tien NW, Ma Y, Cui L, Lyu S, Wang G, Nguyen D, Zhang K, Myroshnychenko MV, Tyan J, Gordon JA, Kupferschmidt DA, Gu Y. A consistent map in the medial entorhinal cortex supports spatial memory. Nat Commun 2024; 15:1457. [PMID: 38368457 PMCID: PMC10874432 DOI: 10.1038/s41467-024-45853-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/05/2024] [Indexed: 02/19/2024] Open
Abstract
The medial entorhinal cortex (MEC) is hypothesized to function as a cognitive map for memory-guided navigation. How this map develops during learning and influences memory remains unclear. By imaging MEC calcium dynamics while mice successfully learned a novel virtual environment over ten days, we discovered that the dynamics gradually became more spatially consistent and then stabilized. Additionally, grid cells in the MEC not only exhibited improved spatial tuning consistency, but also maintained stable phase relationships, suggesting a network mechanism involving synaptic plasticity and rigid recurrent connectivity to shape grid cell activity during learning. Increased c-Fos expression in the MEC in novel environments further supports the induction of synaptic plasticity. Unsuccessful learning lacked these activity features, indicating that a consistent map is specific for effective spatial memory. Finally, optogenetically disrupting spatial consistency of the map impaired memory-guided navigation in a well-learned environment. Thus, we demonstrate that the establishment of a spatially consistent MEC map across learning both correlates with, and is necessary for, successful spatial memory.
Collapse
Affiliation(s)
- Taylor J Malone
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nai-Wen Tien
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Yan Ma
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lian Cui
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shangru Lyu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Center of Neural Science, New York University, New York, NY, USA
| | - Kai Zhang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Maxym V Myroshnychenko
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jean Tyan
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joshua A Gordon
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
- Office of the Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David A Kupferschmidt
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
23
|
Sosa M, Plitt MH, Giocomo LM. Hippocampal sequences span experience relative to rewards. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.27.573490. [PMID: 38234842 PMCID: PMC10793396 DOI: 10.1101/2023.12.27.573490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Hippocampal place cells fire in sequences that span spatial environments and non-spatial modalities, suggesting that hippocampal activity can anchor to the most behaviorally salient aspects of experience. As reward is a highly salient event, we hypothesized that sequences of hippocampal activity can anchor to rewards. To test this, we performed two-photon imaging of hippocampal CA1 neurons as mice navigated virtual environments with changing hidden reward locations. When the reward moved, the firing fields of a subpopulation of cells moved to the same relative position with respect to reward, constructing a sequence of reward-relative cells that spanned the entire task structure. The density of these reward-relative sequences increased with task experience as additional neurons were recruited to the reward-relative population. Conversely, a largely separate subpopulation maintained a spatially-based place code. These findings thus reveal separate hippocampal ensembles can flexibly encode multiple behaviorally salient reference frames, reflecting the structure of the experience.
Collapse
Affiliation(s)
- Marielena Sosa
- Department of Neurobiology, Stanford University School of Medicine; Stanford, CA, USA
| | - Mark H. Plitt
- Department of Neurobiology, Stanford University School of Medicine; Stanford, CA, USA
- Present address: Department of Molecular and Cell Biology, University of California Berkeley; Berkeley, CA, USA
| | - Lisa M. Giocomo
- Department of Neurobiology, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
24
|
Wang XY, He SS, Zhou MM, Li XR, Wang CC, Zhao YC, Xue CH, Che HX. EPA and DHA Alleviated Chronic Dextran Sulfate Sodium Exposure-Induced Depressive-like Behaviors in Mice and Potential Mechanisms Involved. Mar Drugs 2024; 22:76. [PMID: 38393047 PMCID: PMC10890276 DOI: 10.3390/md22020076] [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: 01/09/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Patients with ulcerative colitis (UC) have higher rates of depression. However, the mechanism of depression development remains unclear. The improvements of EPA and DHA on dextran sulfate sodium (DSS)-induced UC have been verified. Therefore, the present study mainly focused on the effects of EPA and DHA on UC-induced depression in C57BL/6 mice and the possible mechanisms involved. A forced swimming test and tail suspension experiment showed that EPA and DHA significantly improved DSS-induced depressive-like behavior. Further analysis demonstrated that EPA and DHA could significantly suppress the inflammation response of the gut and brain by regulating the NLRP3/ASC signal pathway. Moreover, intestine and brain barriers were maintained by enhancing ZO-1 and occludin expression. In addition, EPA and DHA also increased the serotonin (5-HT) concentration and synaptic proteins. Interestingly, EPA and DHA treatments increased the proportion of dominant bacteria, alpha diversity, and beta diversity. In conclusion, oral administration of EPA and DHA alleviated UC-induced depressive-like behavior in mice by modulating the inflammation, maintaining the mucosal and brain barriers, suppressing neuronal damage and reverting microbiota changes.
Collapse
Affiliation(s)
- Xi-Yu Wang
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (X.-Y.W.); (S.-S.H.); (M.-M.Z.); (X.-R.L.)
| | - Shu-Sen He
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (X.-Y.W.); (S.-S.H.); (M.-M.Z.); (X.-R.L.)
| | - Miao-Miao Zhou
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (X.-Y.W.); (S.-S.H.); (M.-M.Z.); (X.-R.L.)
| | - Xiao-Ran Li
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (X.-Y.W.); (S.-S.H.); (M.-M.Z.); (X.-R.L.)
| | - Cheng-Cheng Wang
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China; (C.-C.W.); (Y.-C.Z.)
| | - Ying-Cai Zhao
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China; (C.-C.W.); (Y.-C.Z.)
| | - Chang-Hu Xue
- SKL of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299 Sansha Road, Qingdao 266404, China; (C.-C.W.); (Y.-C.Z.)
| | - Hong-Xia Che
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China; (X.-Y.W.); (S.-S.H.); (M.-M.Z.); (X.-R.L.)
| |
Collapse
|
25
|
Copenhaver AE, LeGates TA. Sex-specific mechanisms underlie long-term potentiation at hippocampus-nucleus accumbens synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575709. [PMID: 38293132 PMCID: PMC10827060 DOI: 10.1101/2024.01.15.575709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary in order to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp-NAc synapses is rewarding, and that mice can make learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigate sex differences in the mechanisms underlying Hipp-NAc LTP using whole-cell electrophysiology and pharmacology. We found that males and females display similar magnitudes of Hipp-NAc LTP which occurs postsynaptically. However, LTP in females requires L-type voltage-gated Ca 2+ channels (VGCC) for postsynaptic Ca 2+ influx, while males rely on NMDA receptors (NMDAR). Additionally, females require estrogen receptor α (ERα) activity for LTP while males do not. These differential mechanisms converge as LTP in both sexes depends on CAMKII activity and occurs independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral excitatory pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders. SIGNIFICANCE STATEMENT Strengthening of Hipp-NAc synapses drives reward-related behaviors. Male and female mice have similar magnitudes of long-term potentiation (LTP) and both sexes have a predicted postsynaptic locus of plasticity. Despite these similarities, we illustrate here that sex-specific molecular mechanisms are used to elicit LTP. Given the bidirectional relationship between Hipp-NAc synaptic strength in mediating reward-related behaviors, the use of distinct molecular mechanisms may explain sex differences observed in stress susceptibility or response to rewarding stimuli. Discovery and characterization of convergent sex differences provides mechanistic insight into the sex-specific function of Hipp-NAc circuitry and has widespread implications for circuits mediating learning and reward-related behavior.
Collapse
|
26
|
Luppi AI, Girn M, Rosas FE, Timmermann C, Roseman L, Erritzoe D, Nutt DJ, Stamatakis EA, Spreng RN, Xing L, Huttner WB, Carhart-Harris RL. A role for the serotonin 2A receptor in the expansion and functioning of human transmodal cortex. Brain 2024; 147:56-80. [PMID: 37703310 DOI: 10.1093/brain/awad311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Integrating independent but converging lines of research on brain function and neurodevelopment across scales, this article proposes that serotonin 2A receptor (5-HT2AR) signalling is an evolutionary and developmental driver and potent modulator of the macroscale functional organization of the human cerebral cortex. A wealth of evidence indicates that the anatomical and functional organization of the cortex follows a unimodal-to-transmodal gradient. Situated at the apex of this processing hierarchy-where it plays a central role in the integrative processes underpinning complex, human-defining cognition-the transmodal cortex has disproportionately expanded across human development and evolution. Notably, the adult human transmodal cortex is especially rich in 5-HT2AR expression and recent evidence suggests that, during early brain development, 5-HT2AR signalling on neural progenitor cells stimulates their proliferation-a critical process for evolutionarily-relevant cortical expansion. Drawing on multimodal neuroimaging and cross-species investigations, we argue that, by contributing to the expansion of the human cortex and being prevalent at the apex of its hierarchy in the adult brain, 5-HT2AR signalling plays a major role in both human cortical expansion and functioning. Owing to its unique excitatory and downstream cellular effects, neuronal 5-HT2AR agonism promotes neuroplasticity, learning and cognitive and psychological flexibility in a context-(hyper)sensitive manner with therapeutic potential. Overall, we delineate a dual role of 5-HT2ARs in enabling both the expansion and modulation of the human transmodal cortex.
Collapse
Affiliation(s)
- Andrea I Luppi
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
- Leverhulme Centre for the Future of Intelligence, University of Cambridge, Cambridge, CB2 1SB, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | - Manesh Girn
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
| | - Fernando E Rosas
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
- Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Timmermann
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Leor Roseman
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David J Nutt
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - R Nathan Spreng
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Robin L Carhart-Harris
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
27
|
Su J, Huang F, Tian Y, Tian R, Qianqian G, Bello ST, Zeng D, Jendrichovsky P, Lau CG, Xiong W, Yu D, Tortorella M, Chen X, He J. Entorhinohippocampal cholecystokinin modulates spatial learning by facilitating neuroplasticity of hippocampal CA3-CA1 synapses. Cell Rep 2023; 42:113467. [PMID: 37979171 DOI: 10.1016/j.celrep.2023.113467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
The hippocampus is broadly impacted by neuromodulations. However, how neuropeptides shape the function of the hippocampus and the related spatial learning and memory remains unclear. Here, we discover the crucial role of cholecystokinin (CCK) in heterosynaptic neuromodulation from the medial entorhinal cortex (MEC) to the hippocampus. Systematic knockout of the CCK gene impairs CA3-CA1 LTP and space-related performance. The MEC provides most of the CCK-positive neurons projecting to the hippocampal region, which potentiates CA3-CA1 long-term plasticity heterosynaptically in a frequency- and NMDA receptor (NMDAR)-dependent manner. Selective inhibition of MEC CCKergic neurons or downregulation of their CCK mRNA levels also impairs CA3-CA1 LTP formation and animals' performance in the water maze. This excitatory extrahippocampal projection releases CCK upon high-frequency excitation and is active during animal exploration. Our results reveal the critical role of entorhinal CCKergic projections in bridging intra- and extrahippocampal circuitry at electrophysiological and behavioral levels.
Collapse
Affiliation(s)
- Junfeng Su
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Fengwen Huang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China.
| | - Yu Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Ran Tian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Gao Qianqian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Stephen Temitayo Bello
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China
| | - Dingxaun Zeng
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Peter Jendrichovsky
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - C Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China
| | - Wenjun Xiong
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China
| | - Daiguan Yu
- Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Micky Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R. China; Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China.
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, P.R. China.
| |
Collapse
|
28
|
Krishnan S, Sheffield ME. Reward Expectation Reduces Representational Drift in the Hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572809. [PMID: 38187677 PMCID: PMC10769341 DOI: 10.1101/2023.12.21.572809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Spatial memory in the hippocampus involves dynamic neural patterns that change over days, termed representational drift. While drift may aid memory updating, excessive drift could impede retrieval. Memory retrieval is influenced by reward expectation during encoding, so we hypothesized that diminished reward expectation would exacerbate representational drift. We found that high reward expectation limited drift, with CA1 representations on one day gradually re-emerging over successive trials the following day. Conversely, the absence of reward expectation resulted in increased drift, as the gradual re-emergence of the previous day's representation did not occur. At the single cell level, lowering reward expectation caused an immediate increase in the proportion of place-fields with low trial-to-trial reliability. These place fields were less likely to be reinstated the following day, underlying increased drift in this condition. In conclusion, heightened reward expectation improves memory encoding and retrieval by maintaining reliable place fields that are gradually reinstated across days, thereby minimizing representational drift.
Collapse
|
29
|
Congiu M, Mondoloni S, Zouridis IS, Schmors L, Lecca S, Lalive AL, Ginggen K, Deng F, Berens P, Paolicelli RC, Li Y, Burgalossi A, Mameli M. Plasticity of neuronal dynamics in the lateral habenula for cue-punishment associative learning. Mol Psychiatry 2023; 28:5118-5127. [PMID: 37414924 PMCID: PMC11041652 DOI: 10.1038/s41380-023-02155-3] [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: 01/12/2023] [Revised: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
The brain's ability to associate threats with external stimuli is vital to execute essential behaviours including avoidance. Disruption of this process contributes instead to the emergence of pathological traits which are common in addiction and depression. However, the mechanisms and neural dynamics at the single-cell resolution underlying the encoding of associative learning remain elusive. Here, employing a Pavlovian discrimination task in mice we investigate how neuronal populations in the lateral habenula (LHb), a subcortical nucleus whose excitation underlies negative affect, encode the association between conditioned stimuli and a punishment (unconditioned stimulus). Large population single-unit recordings in the LHb reveal both excitatory and inhibitory responses to aversive stimuli. Additionally, local optical inhibition prevents the formation of cue discrimination during associative learning, demonstrating a critical role of LHb activity in this process. Accordingly, longitudinal in vivo two-photon imaging tracking LHb calcium neuronal dynamics during conditioning reveals an upward or downward shift of individual neurons' CS-evoked responses. While recordings in acute slices indicate strengthening of synaptic excitation after conditioning, support vector machine algorithms suggest that postsynaptic dynamics to punishment-predictive cues represent behavioral cue discrimination. To examine the presynaptic signaling in LHb participating in learning we monitored neurotransmitter dynamics with genetically-encoded indicators in behaving mice. While glutamate, GABA, and serotonin release in LHb remain stable across associative learning, we observe enhanced acetylcholine signaling developing throughout conditioning. In summary, converging presynaptic and postsynaptic mechanisms in the LHb underlie the transformation of neutral cues in valued signals supporting cue discrimination during learning.
Collapse
Affiliation(s)
- Mauro Congiu
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Sarah Mondoloni
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Ioannis S Zouridis
- Institute of Neurobiology and Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076, Tübingen, Germany
- Graduate Training Centre of Neuroscience, International Max Planck Research School (IMPRS), University of Tübingen, Tübingen, Germany
| | - Lisa Schmors
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Tübingen, Germany
| | - Salvatore Lecca
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Arnaud L Lalive
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Kyllian Ginggen
- The Department of Biomedical Sciences, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Fei Deng
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Philipp Berens
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Tübingen AI Center, University of Tübingen, Tübingen, Germany
| | - Rosa Chiara Paolicelli
- The Department of Biomedical Sciences, The University of Lausanne, 1005, Lausanne, Switzerland
| | - Yulong Li
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Andrea Burgalossi
- Institute of Neurobiology and Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076, Tübingen, Germany
| | - Manuel Mameli
- The Department of Fundamental Neuroscience, The University of Lausanne, 1005, Lausanne, Switzerland.
- Inserm, UMR-S 839, 75005, Paris, France.
| |
Collapse
|
30
|
Yang P, Chen H, Wang T, Su H, Li J, He Y, Su S. Electroacupuncture promotes synaptic plasticity in rats with chronic inflammatory pain-related depression by upregulating BDNF/TrkB/CREB signaling pathway. Brain Behav 2023; 13:e3310. [PMID: 37948105 PMCID: PMC10726860 DOI: 10.1002/brb3.3310] [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: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Chronic inflammatory pain (CIP) frequently coincides with depression among patients. The onset and development of pain and depression are associated with altered neural synaptic plasticity. Electroacupuncture (EA) can effectively relieve CIP and depression. However, the underlying mechanisms have not been fully illustrated. OBJECTIVE To explore whether EA can relieve CIP and depression by regulating hippocampal synaptic plasticity, and the present study offers foundational evidence for the efficacy of EA in treating CIP-related depression (CIPD). METHODS Rats were divided into four groups: 0.9% normal saline group, complete Freund's adjuvant (CFA) group, CFA + duloxetine group, and CFA + EA group. Pain hypersensitivity was detected by mechanical withdrawal threshold and thermal paw withdrawal latency, and the depression level was gauged using the open field test, the sucrose preference test, and the forced swimming test. The morphology of the hippocampal neurons was observed using Nissl staining. The protein expression levels of synuclein (Syn), postsynaptic density protein-95 (PSD-95), brain-derived neurotrophic factors (BDNFs), tyrosine-protein kinase B (TrKB), p-TrkB, cAMP response element binding protein (CREB), and p-CREB were measured by western blotting and immunofluorescence staining. BDNF and TrkB mRNA expression were detected using quantitative real-time polymerase chain reaction (PCR) (qRT-PCR). The content of 5-hydroxytryptamine (5-HT) and γ-aminobutyric acid (GABA) was detected using enzyme-linked immunosorbent assay, and the glutamic acid (Glu) content was determined using the ultraviolet colorimetry method. The hippocampal neuron ultrastructure was observed using transmission electron microscopy. RESULTS EA could alleviate CIP and related depressive behaviors as well as protect the hippocampal neuronal structure from damage and regulate 5-HT/GABA/Glu levels in the hippocampus. Additionally, EA could significantly increase the expression of synapse-associated proteins such as PSD-95 and Syn by activating the BDNF/TrKB/CREB signaling pathway. CONCLUSION EA improves pain and depressive behaviors in CIPD rats, and the mechanism may be related to synaptic plasticity mediated by the BDNF/TrKB/CREB signaling pathway.
Collapse
Affiliation(s)
- Pu Yang
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Haiyan Chen
- Department of NursingThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningGuangxiChina
| | - Tian Wang
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Hong Su
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Jing Li
- The First School of Clinical MedicineGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Yujun He
- Faculty of Acupuncture, Moxibustion and TuinaGuangxi University of Chinese MedicineNanningGuangxiChina
| | - Shengyong Su
- Department of Acupuncture and MoxibustionThe First Affiliated Hospital of Guangxi University of Chinese MedicineNanningGuangxiChina
- Guangxi Key Laboratory of Molecular Biology of Preventive Medicine of Traditional Chinese MedicineNanningGuangxiChina
| |
Collapse
|
31
|
KLOCKE B, MOORE C, OTT H, PITYCHOUTIS PM. Chronic pharmacological activation of SERCA with CDN1163 affects spatial cognitive flexibility but not attention and impulsivity in mice. Behav Pharmacol 2023; 34:477-487. [PMID: 37917567 PMCID: PMC10624114 DOI: 10.1097/fbp.0000000000000756] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Intracellular calcium (Ca2+) homeostasis is critical for many neural processes, including learning, memory and synaptic plasticity. The sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) is among the key regulators that preserve Ca2+ homeostasis in neurons. SERCAs comprise a set of ubiquitously expressed Ca2+ pumps that primarily function to sequester cytosolic Ca2+ into endoplasmic reticular stores. As SERCA has been implicated in the neurobiology of several neuropsychiatric and neurodegenerative diseases, pharmacological harnessing of its function is critical in understanding SERCA's role in brain physiology and pathophysiology. In the current study, we employed the Morris water maze and 5-choice serial reaction time task (5-CSRTT) to investigate the effects of chronic pharmacological activation of SERCA, using the small allosteric SERCA activator CDN1163, on spatial learning and memory, and executive functioning in naive C57BL/6J mice. Our data show that chronic pharmacological SERCA activation with CDN1163 (20 mg/kg) selectively impairs spatial cognitive flexibility and reversal learning in the Morris water maze while leaving executive functions such as attention and impulsivity intact. Present findings contribute to the growing field of the role of SERCA function in the brain and behavior and expand current knowledge on the use of the small allosteric activator CDN1163 as an investigational tool to study the role of SERCA in regulating neurobehavioral processes and as a potential therapeutic candidate for debilitating brain disorders.
Collapse
Affiliation(s)
- Benjamin KLOCKE
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | - Carter MOORE
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | - Hayden OTT
- Department of Biology, University of Dayton, Dayton, Ohio 45469, USA
| | | |
Collapse
|
32
|
Iannucci J, O’Neill K, Wang X, Mukherjee S, Wang J, Shapiro LA. Sex-Specific and Traumatic Brain Injury Effects on Dopamine Receptor Expression in the Hippocampus. Int J Mol Sci 2023; 24:16084. [PMID: 38003274 PMCID: PMC10671736 DOI: 10.3390/ijms242216084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health concern. Each year, over 50 million individuals worldwide suffer from TBI, and this leads to a number of acute and chronic health issues. These include affective and cognitive impairment, as well as an increased risk of alcohol and drug use. The dopaminergic system, a key component of reward circuitry, has been linked to alcohol and other substance use disorders, and previous research indicates that TBI can induce plasticity within this system. Understanding how TBI modifies the dopaminergic system may offer insights into the heightened substance use and reward-seeking behavior following TBI. The hippocampus, a critical component of the reward circuit, is responsible for encoding and integrating the spatial and salient aspects of rewarding stimuli. This study explored TBI-related changes in neuronal D2 receptor expression within the hippocampus, examining the hypothesis that sex differences exist in both baseline hippocampal D2 receptor expression and its response to TBI. Utilizing D2-expressing tdTomato transgenic male and female mice, we implemented either a sham injury or the lateral fluid percussion injury (FPI) model of TBI and subsequently performed a region-specific quantification of D2 expression in the hippocampus. The results show that male mice exhibit higher baseline hippocampal D2 expression compared to female mice. Additionally, there was a significant interaction effect between sex and injury on the expression of D2 in the hippocampus, particularly in regions of the dentate gyrus. Furthermore, TBI led to significant reductions in hippocampal D2 expression in male mice, while female mice remained mostly unaffected. These results suggest that hippocampal D2 expression varies between male and female mice, with the female dopaminergic system demonstrating less susceptibility to TBI-induced plasticity.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Katherine O’Neill
- Department of Biological Science, Texas A&M University, College Station, TX 77843, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Sanjib Mukherjee
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Lee A. Shapiro
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| |
Collapse
|
33
|
Wu D, Sun Q, Wei W, Bai Y, Zhai L, Jia L. Nrf2-mediated protective effect of alpha-lipoic acid on synaptic oxidative damage and inhibition of PKC/ERK/CREB pathway in bisphenol A-exposed HT-22 cells. Food Chem Toxicol 2023; 181:114112. [PMID: 37858839 DOI: 10.1016/j.fct.2023.114112] [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: 05/10/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
The harmful effects of bisphenol A (BPA) on learning and memory may involve hippocampal oxidative damage; however, the underlying mechanism remains unclear. Antioxidants that antagonize BPA-induced neuronal oxidative damage lack research. This study aimed to develop an in vitro model using the HT-22 mouse hippocampal neuronal cell line to investigate the neurotoxic mechanism of BPA and the protective effect of alpha-lipoic acid (ALA) on nuclear factor erythroid 2-related factor 2 (Nrf2) inhibition. The results showed that ALA reduced BPA-induced reactive oxygen species and neuronal nitric oxide synthase (nNOS) levels; however, inhibiting Nrf2 weakened the protective effects of ALA. BPA reduced mitochondrial complex I/III activity and ATP levels, but ALA ameliorated this damage. ALA improved the BPA-induced downregulation of the kelch-like ECH-associated protein 1 (keap1)/Nrf2 system, synaptic-related proteins, and the protein kinase C (PKC)/extracellular signal-regulated kinase (ERK)/cAMP response element binding protein (CREB) pathway; however, the protective effects of ALA were weakened when Nrf2 was inhibited. Our results suggest that BPA causes oxidative damage to HT-22 cells by damaging mitochondrial function, nNOS, and the keap1/Nrf2 system, thereby impairing synaptic-related proteins and the PKC/ERK/CREB pathway. ALA counters BPA-induced damage via Nrf2, which may be a significant target for the protective action of ALA.
Collapse
Affiliation(s)
- Dan Wu
- Department of Child and Adolescent Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Qi Sun
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Wei Wei
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Yinglong Bai
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Lingling Zhai
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Lihong Jia
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang, Liaoning, 110122, China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
34
|
Malone TJ, Tien NW, Ma Y, Cui L, Lyu S, Wang G, Nguyen D, Zhang K, Myroshnychenko MV, Tyan J, Gordon JA, Kupferschmidt DA, Gu Y. A consistent map in the medial entorhinal cortex supports spatial memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560254. [PMID: 37986767 PMCID: PMC10659391 DOI: 10.1101/2023.09.30.560254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
The medial entorhinal cortex (MEC) is hypothesized to function as a cognitive map for memory-guided navigation. How this map develops during learning and influences memory remains unclear. By imaging MEC calcium dynamics while mice successfully learned a novel virtual environment over ten days, we discovered that the dynamics gradually became more spatially consistent and then stabilized. Additionally, grid cells in the MEC not only exhibited improved spatial tuning consistency, but also maintained stable phase relationships, suggesting a network mechanism involving synaptic plasticity and rigid recurrent connectivity to shape grid cell activity during learning. Increased c-Fos expression in the MEC in novel environments further supports the induction of synaptic plasticity. Unsuccessful learning lacked these activity features, indicating that a consistent map is specific for effective spatial memory. Finally, optogenetically disrupting spatial consistency of the map impaired memory-guided navigation in a well-learned environment. Thus, we demonstrate that the establishment of a spatially consistent MEC map across learning both correlates with, and is necessary for, successful spatial memory.
Collapse
Affiliation(s)
- Taylor J. Malone
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Nai-Wen Tien
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Current address: Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- These authors contributed equally to this work
| | - Yan Ma
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- These authors contributed equally to this work
| | - Lian Cui
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shangru Lyu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Current address: Center of Neural Science, New York University, New York, NY, USA
| | - Kai Zhang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Maxym V. Myroshnychenko
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jean Tyan
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua A. Gordon
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
- Office of the Director, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A. Kupferschmidt
- Integrative Neuroscience Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
35
|
Dahl MJ, Kulesza A, Werkle-Bergner M, Mather M. Declining locus coeruleus-dopaminergic and noradrenergic modulation of long-term memory in aging and Alzheimer's disease. Neurosci Biobehav Rev 2023; 153:105358. [PMID: 37597700 PMCID: PMC10591841 DOI: 10.1016/j.neubiorev.2023.105358] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023]
Abstract
Memory is essential in defining our identity by guiding behavior based on past experiences. However, aging leads to declining memory, disrupting older adult's lives. Memories are encoded through experience-dependent modifications of synaptic strength, which are regulated by the catecholamines dopamine and noradrenaline. While cognitive aging research demonstrates how dopaminergic neuromodulation from the substantia nigra-ventral tegmental area regulates hippocampal synaptic plasticity and memory, recent findings indicate that the noradrenergic locus coeruleus sends denser inputs to the hippocampus. The locus coeruleus produces dopamine as biosynthetic precursor of noradrenaline, and releases both to modulate hippocampal plasticity and memory. Crucially, the locus coeruleus is also the first site to accumulate Alzheimer's-related abnormal tau and severely degenerates with disease development. New in-vivo assessments of locus coeruleus integrity reveal associations with Alzheimer's markers and late-life memory impairments, which likely stem from impaired dopaminergic and noradrenergic neurotransmission. Bridging research across species, the reviewed findings suggest that degeneration of the locus coeruleus results in deficient dopaminergic and noradrenergic modulation of hippocampal plasticity and thus memory decline.
Collapse
Affiliation(s)
- Martin J Dahl
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany; Leonard Davis School of Gerontology, University of Southern California, 90089 Los Angeles, CA, USA.
| | - Agnieszka Kulesza
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany
| | - Markus Werkle-Bergner
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195 Berlin, Germany
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, 90089 Los Angeles, CA, USA; Department of Psychology, University of Southern California, Los Angeles, California, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
36
|
Nelson ED, Maynard KR, Nicholas KR, Tran MN, Divecha HR, Collado-Torres L, Hicks SC, Martinowich K. Activity-regulated gene expression across cell types of the mouse hippocampus. Hippocampus 2023; 33:1009-1027. [PMID: 37226416 PMCID: PMC11129873 DOI: 10.1002/hipo.23548] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/19/2023] [Accepted: 05/06/2023] [Indexed: 05/26/2023]
Abstract
Activity-regulated gene (ARG) expression patterns in the hippocampus (HPC) regulate synaptic plasticity, learning, and memory, and are linked to both risk and treatment responses for many neuropsychiatric disorders. The HPC contains discrete classes of neurons with specialized functions, but cell type-specific activity-regulated transcriptional programs are not well characterized. Here, we used single-nucleus RNA-sequencing (snRNA-seq) in a mouse model of acute electroconvulsive seizures (ECS) to identify cell type-specific molecular signatures associated with induced activity in HPC neurons. We used unsupervised clustering and a priori marker genes to computationally annotate 15,990 high-quality HPC neuronal nuclei from N = 4 mice across all major HPC subregions and neuron types. Activity-induced transcriptomic responses were divergent across neuron populations, with dentate granule cells being particularly responsive to activity. Differential expression analysis identified both upregulated and downregulated cell type-specific gene sets in neurons following ECS. Within these gene sets, we identified enrichment of pathways associated with varying biological processes such as synapse organization, cellular signaling, and transcriptional regulation. Finally, we used matrix factorization to reveal continuous gene expression patterns differentially associated with cell type, ECS, and biological processes. This work provides a rich resource for interrogating activity-regulated transcriptional responses in HPC neurons at single-nuclei resolution in the context of ECS, which can provide biological insight into the roles of defined neuronal subtypes in HPC function.
Collapse
Affiliation(s)
- Erik D. Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kyndall R. Nicholas
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew N Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205
| |
Collapse
|
37
|
Jain A, Nakahata Y, Watabe T, Rusina P, South K, Adachi K, Yan L, Simorowski N, Furukawa H, Yasuda R. Dendritic, delayed, and stochastic CaMKII activation underlies behavioral time scale plasticity in CA1 synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.549180. [PMID: 37577549 PMCID: PMC10418109 DOI: 10.1101/2023.08.01.549180] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Behavioral time scale plasticity (BTSP), is a form of non-Hebbian plasticity induced by integrating pre- and postsynaptic components separated by behavioral time scale (seconds). BTSP in the hippocampal CA1 neurons underlies place cell formation. However, the molecular mechanisms underlying this behavioral time scale (eligibility trace) and synapse specificity are unknown. CaMKII can be activated in a synapse-specific manner and remain active for a few seconds, making it a compelling candidate for the eligibility trace during BTSP. Here, we show that BTSP can be induced in a single dendritic spine using 2-photon glutamate uncaging paired with postsynaptic current injection temporally separated by behavioral time scale. Using an improved CaMKII sensor, we saw no detectable CaMKII activation during this BTSP induction. Instead, we observed a dendritic, delayed, and stochastic CaMKII activation (DDSC) associated with Ca 2+ influx and plateau 20-40 s after BTSP induction. DDSC requires both pre-and postsynaptic activity, suggesting that CaMKII can integrate these two signals. Also, optogenetically blocking CaMKII 30 s after the BTSP protocol inhibited synaptic potentiation, indicating that DDSC is an essential mechanism of BTSP. IP3-dependent intracellular Ca 2+ release facilitates both DDSC and BTSP. Thus, our study suggests that the non-synapse specific CaMKII activation provides an instructive signal with an extensive time window over tens of seconds during BTSP.
Collapse
|
38
|
Guskjolen A, Cembrowski MS. Engram neurons: Encoding, consolidation, retrieval, and forgetting of memory. Mol Psychiatry 2023; 28:3207-3219. [PMID: 37369721 PMCID: PMC10618102 DOI: 10.1038/s41380-023-02137-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Tremendous strides have been made in our understanding of the neurobiological substrates of memory - the so-called memory "engram". Here, we integrate recent progress in the engram field to illustrate how engram neurons transform across the "lifespan" of a memory - from initial memory encoding, to consolidation and retrieval, and ultimately to forgetting. To do so, we first describe how cell-intrinsic properties shape the initial emergence of the engram at memory encoding. Second, we highlight how these encoding neurons preferentially participate in synaptic- and systems-level consolidation of memory. Third, we describe how these changes during encoding and consolidation guide neural reactivation during retrieval, and facilitate memory recall. Fourth, we describe neurobiological mechanisms of forgetting, and how these mechanisms can counteract engram properties established during memory encoding, consolidation, and retrieval. Motivated by recent experimental results across these four sections, we conclude by proposing some conceptual extensions to the traditional view of the engram, including broadening the view of cell-type participation within engrams and across memory stages. In collection, our review synthesizes general principles of the engram across memory stages, and describes future avenues to further understand the dynamic engram.
Collapse
Affiliation(s)
- Axel Guskjolen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Mark S Cembrowski
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
39
|
Sun N, Cui WQ, Min XM, Zhang GM, Liu JZ, Wu HY. A new perspective on hippocampal synaptic plasticity and post-stroke depression. Eur J Neurosci 2023; 58:2961-2984. [PMID: 37518943 DOI: 10.1111/ejn.16093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Post-stroke depression, a common complication after stroke, severely affects the recovery and quality of life of patients with stroke. Owing to its complex mechanisms, post-stroke depression treatment remains highly challenging. Hippocampal synaptic plasticity is one of the key factors leading to post-stroke depression; however, the precise molecular mechanisms remain unclear. Numerous studies have found that neurotrophic factors, protein kinases and neurotransmitters influence depressive behaviour by modulating hippocampal synaptic plasticity. This review further elaborates on the role of hippocampal synaptic plasticity in post-stroke depression by summarizing recent research and analysing possible molecular mechanisms. Evidence for the correlation between hippocampal mechanisms and post-stroke depression helps to better understand the pathological process of post-stroke depression and improve its treatment.
Collapse
Affiliation(s)
- Ning Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Man Min
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia-Zheng Liu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
40
|
Abstract
Flexible behavior requires the creation, updating, and expression of memories to depend on context. While the neural underpinnings of each of these processes have been intensively studied, recent advances in computational modeling revealed a key challenge in context-dependent learning that had been largely ignored previously: Under naturalistic conditions, context is typically uncertain, necessitating contextual inference. We review a theoretical approach to formalizing context-dependent learning in the face of contextual uncertainty and the core computations it requires. We show how this approach begins to organize a large body of disparate experimental observations, from multiple levels of brain organization (including circuits, systems, and behavior) and multiple brain regions (most prominently the prefrontal cortex, the hippocampus, and motor cortices), into a coherent framework. We argue that contextual inference may also be key to understanding continual learning in the brain. This theory-driven perspective places contextual inference as a core component of learning.
Collapse
Affiliation(s)
- James B Heald
- Department of Neuroscience and Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; ,
| | - Daniel M Wolpert
- Department of Neuroscience and Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; ,
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, Cambridge, United Kingdom;
| | - Máté Lengyel
- Computational and Biological Learning Lab, Department of Engineering, University of Cambridge, Cambridge, United Kingdom;
- Center for Cognitive Computation, Department of Cognitive Science, Central European University, Budapest, Hungary
| |
Collapse
|
41
|
Kim S, Seong KM, Lee SH. Acetylcholine titre regulation by non-neuronal acetylcholinesterase 1 and its putative roles in honey bee physiology. INSECT MOLECULAR BIOLOGY 2023. [PMID: 37130064 DOI: 10.1111/imb.12845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 04/11/2023] [Indexed: 05/03/2023]
Abstract
Similar to other insects, honey bees have two acetylcholinesterases (AChEs), AmAChE1 and AmAChE2. The primary catalytic enzyme for acetylcholine (ACh) hydrolysis in synapses is AmAChE2, which is predominantly expressed in neuronal tissues, whereas AmAChE1 is expressed in both neuronal and non-neuronal tissues, with limited catalytic activity. Unlike constitutively expressed AmAChE2, AmAChE1 expression is induced under stressful conditions such as heat shock and brood rearing suppression, but its role in regulating ACh titre remains unclear. In this paper, to elucidate the role of AmAChE1, the expression of AmAChE1 was suppressed via RNA interference (RNAi) in AmAChE1-induced worker bees. The ACh titre measurement following RNAi revealed that the expression of AmAChE1 downregulated the overall ACh titre in all tissues examined without altering AmAChE2 expression. Transcriptome analysis showed that AmAChE1 knockdown upregulated protein biosynthesis, cell respiration, and thermogenesis in the head. These findings suggest that AmAChE1 is involved in decreasing neuronal activity, enhancing energy conservation, and potentially extending longevity under stressful conditions via ACh titre regulation.
Collapse
Affiliation(s)
- Sanghyeon Kim
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Keon Mook Seong
- Department of Applied Biology, Chungnam National University, Daejeon, South Korea
| | - Si Hyeock Lee
- Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
- Department of Agricultural Biotechnology, College of Agriculture and Life Science, Seoul National University, Seoul, South Korea
| |
Collapse
|
42
|
Ramirez-Mejia G, Gil-Lievana E, Urrego-Morales O, Galvez-Marquez D, Hernández-Ortiz E, Carrillo-Lorenzo JA, Bermúdez-Rattoni F. Salience to remember: VTA-IC dopaminergic pathway activity is necessary for object recognition memory formation. Neuropharmacology 2023; 228:109464. [PMID: 36804534 DOI: 10.1016/j.neuropharm.2023.109464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
Previous studies have shown that dopaminergic activity modulates the salience of novel stimuli enabling the formation of recognition memories. In this work, we hypothesize that dopamine released into the insular cortex (IC) from the ventral tegmental area (VTA) inputs enables the acquisition to consolidate object recognition memory. It has been reported that short training produces weak recognition memories; on the contrary, longer training produces lasting and robust recognition memories. Using a Cre-recombinase under the tyrosine hydroxylase (TH+) promoter mouse model, we photostimulated the VTA-IC dopaminergic pathway during short training or photoinhibited the same pathway during long training while mice explored objects. Our results showed that the photostimulation of the VTA-IC pathway during a short training enables the acquisition of recognition memory. Conversely, photoinhibition of the same pathway during a long training prevents the acquisition of recognition memory. Interestingly, the exploration time of the objects under photoinhibition or photostimulation of the dopaminergic VTA-IC pathway was not altered. Significantly, this enhancement of acquisition of the object recognition memory through the photostimulation of the VTA dopaminergic neurons could be impaired by the blockage of the D1-like receptors into the IC, either before or after the photostimulation. Altogether, our results suggest that dopamine released by the VTA is required during the acquisition to consolidate the object recognition memory through D1-like receptors into the IC without affecting the activity or the motivation to explore objects.
Collapse
Affiliation(s)
- Gerardo Ramirez-Mejia
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Elvi Gil-Lievana
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Oscar Urrego-Morales
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Donovan Galvez-Marquez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Eduardo Hernández-Ortiz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - José Alberto Carrillo-Lorenzo
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
43
|
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: 32] [Impact Index Per Article: 16.0] [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
|
44
|
Sartori BM, Moreira Júnior RE, Paiva IM, Moraes IB, Murgas LDS, Brunialti-Godard AL. Acute ethanol exposure leads to long-term effects on memory, behavior, and transcriptional regulation in the zebrafish brain. Behav Brain Res 2023; 444:114352. [PMID: 36842314 DOI: 10.1016/j.bbr.2023.114352] [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: 10/28/2022] [Revised: 02/02/2023] [Accepted: 02/15/2023] [Indexed: 02/27/2023]
Abstract
Alcohol consumption is associated with alterations in memory and learning processes in humans and animals. In this context, research models such as the zebrafish (Danio rerio) arise as key organisms in behavioral and molecular studies that attempt to clarify alterations in the Central Nervous System (CNS), like those related to alcohol use. Accordingly, we used the zebrafish as a model to evaluate the effects of ethanol on the learning and memory process, as well as its relationship with behavior and transcriptional regulation of lrfn2, lrrk2, grin1a, and bdnf genes in the brain. To this end, for the memory and learning evaluation, we conducted the Novel Object Recognition test (NOR); for behavior, the Novel Tank test; and for gene transcription, qPCR, after 2 h, 24 h, and 8 days of ethanol exposure. As a result, we noticed in the NOR that after 8 days of ethanol exposure, the control group spent more time exploring the novel object than when compared to 2 h post-exposure, indicating that naturally zebrafish remember familiar objects. In animals in the Treatment group, however, no object recognition behavior was observed, suggesting that alcohol affected the learning and memory processes of the animals and stimulated an anxiolytic effect in them. Regarding transcriptional regulation, 24 h after alcohol exposure, we found hyper-regulation of bdnf and, after 8 days, a hypo-regulation of lrfn2 and lrrk2. To conclude, we demonstrated that ethanol exposure may have influenced learning ability and memory formation in zebrafish, as well as behavior and regulation of gene transcription. These data are relevant for further understanding the application of zebrafish in research associated with ethanol consumption and behavior.
Collapse
Affiliation(s)
- Barbara Miranda Sartori
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Renato Elias Moreira Júnior
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Isadora Marques Paiva
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; Centro de Pesquisas em Doenças Inflamatórias (CRID), Faculdade de Medicina de Ribeirão Preto, Departamento de Farmacologia, Universidade de São Paulo (FMRP), Ribeirão Preto, Brazil
| | - Izabela Barbosa Moraes
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; Centro das Ciências Biológicas e da Saúde, Universidade Federal do Oeste da Bahia (UFOB), Barreiras, Brazil
| | - Luis David Solis Murgas
- Biotério Central, Departamento de Medicina Veterinária, Universidade Federal de Lavras (UFLA), Lavras, Brazil
| | - Ana Lúcia Brunialti-Godard
- Laboratório de Genética Animal e Humana, Departamento de Genética, Ecologia e Evolução, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| |
Collapse
|
45
|
Zhuo B, Deng S, Li B, Zhu W, Zhang M, Qin C, Meng Z. Possible Effects of Acupuncture in Poststroke Aphasia. Behav Neurol 2023; 2023:9445381. [PMID: 37091130 PMCID: PMC10115536 DOI: 10.1155/2023/9445381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 12/30/2022] [Accepted: 04/02/2023] [Indexed: 04/25/2023] Open
Abstract
Neural plasticity promotes the reorganization of language networks and is an essential recovery mechanism for poststroke aphasia (PSA). Neuroplasticity may be a pivotal bridge to elucidate the potential recovery mechanisms of acupuncture for aphasia. Therefore, understanding the neuroplasticity mechanism of acupuncture in PSA is crucial. However, the underlying therapeutic mechanism of neuroplasticity in PSA after acupuncture needs to be explored. Excitotoxicity after brain injury affects the activity of neurotransmitters and disrupts the transmission of normal neuron information. Thus, a helpful strategy of acupuncture might be to improve PSA by affecting the availability of these neurotransmitters and glutamate receptors at synapses. In addition, the regulation of neuroplasticity by acupuncture may also be related to the regulation of astrocytes. Considering the guiding significance of acupuncture for clinical treatment, it is necessary to carry out further study about the influence of acupuncture on the recovery of aphasia after stroke. This study summarizes the current research on the neural mechanism of acupuncture in treating PSA. It seeks to elucidate the potential effect of acupuncture on the recovery of PSA from the perspective of synaptic plasticity and integrity of gray and white matter.
Collapse
Affiliation(s)
- Bifang Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shizhe Deng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Boxuan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Weiming Zhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Menglong Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chenyang Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhihong Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
46
|
Kim HH, Lee SH, Ho WK, Eom K. Dopamine Receptor Supports the Potentiation of Intrinsic Excitability and Synaptic LTD in Temporoammonic-CA1 Synapse. Exp Neurobiol 2022; 31:361-375. [PMID: 36631845 PMCID: PMC9841748 DOI: 10.5607/en22028] [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: 08/11/2022] [Revised: 11/15/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Dopaminergic projection to the hippocampus from the ventral tegmental area or locus ceruleus has been considered to play an essential role in the acquisition of novel information. Hence, the dopaminergic modulation of synaptic plasticity in the hippocampus has been widely studied. We examined how the D1 and D2 receptors influenced the mGluR5-mediated synaptic plasticity of the temporoammonic-CA1 synapses and showed that the dopaminergic modulation of the temporoammonic-CA1 synapses was expressed in various ways. Our findings suggest that the dopaminergic system in the hippocampal CA1 region regulates the long-term synaptic plasticity and processing of the novel information.
Collapse
Affiliation(s)
- Hye-Hyun Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea,Neuroscience Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Suk-Ho Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea,Neuroscience Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won-Kyung Ho
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea,Neuroscience Research Center, Seoul National University College of Medicine, Seoul 03080, Korea,Won-Kyung Ho, TEL: 82-2-740-8226, FAX: 82-2-763-9667, e-mail:
| | - Kisang Eom
- Department of Physiology, School of Medicine, Keimyung University, Daegu 42601, Korea,To whom correspondence should be addressed. Kisang Eom, TEL: 82-53-258-7416, FAX: 82-53-258-7412, e-mail:
| |
Collapse
|
47
|
Hirai H, Sakaba T, Hashimotodani Y. Subcortical glutamatergic inputs exhibit a Hebbian form of long-term potentiation in the dentate gyrus. Cell Rep 2022; 41:111871. [PMID: 36577371 DOI: 10.1016/j.celrep.2022.111871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/19/2022] [Accepted: 12/01/2022] [Indexed: 12/28/2022] Open
Abstract
The hippocampus receives glutamatergic and GABAergic inputs from subcortical regions. Despite the important roles of these subcortical inputs in the regulation of hippocampal circuit, it has not been explored whether associative activation of the subcorticohippocampal pathway induces Hebbian plasticity of subcortical inputs. Here, we demonstrate that the hypothalamic supramammillary nucleus (SuM) to the dentate granule cell (GC) synapses, which co-release glutamate and GABA, undergo associative long-term potentiation (LTP) of glutamatergic, but not GABAergic, co-transmission. This LTP is induced by pairing of SuM inputs with GC spikes. We found that this Hebbian LTP is input-specific, requires NMDA receptors and CaMKII activation, and is expressed postsynaptically. By the net increase in excitatory drive of SuM inputs following LTP induction, associative inputs of SuM and the perforant path effectively discharge GCs. Our results highlight the important role of associative plasticity at SuM-GC synapses in the regulation of dentate gyrus activity and for the encoding of SuM-related information.
Collapse
Affiliation(s)
- Himawari Hirai
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Takeshi Sakaba
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan
| | - Yuki Hashimotodani
- Graduate School of Brain Science, Doshisha University, Kyoto 610-0394, Japan.
| |
Collapse
|
48
|
Hegedüs P, Sviatkó K, Király B, Martínez-Bellver S, Hangya B. Cholinergic activity reflects reward expectations and predicts behavioral responses. iScience 2022; 26:105814. [PMID: 36636356 PMCID: PMC9830220 DOI: 10.1016/j.isci.2022.105814] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/22/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Basal forebrain cholinergic neurons (BFCNs) play an important role in associative learning, suggesting that BFCNs may participate in processing stimuli that predict future outcomes. However, the impact of outcome probabilities on BFCN activity remained elusive. Therefore, we performed bulk calcium imaging and recorded spiking of identified cholinergic neurons from the basal forebrain of mice performing a probabilistic Pavlovian cued outcome task. BFCNs responded more to sensory cues that were often paired with reward. Reward delivery also activated BFCNs, with surprising rewards eliciting a stronger response, whereas punishments evoked uniform positive-going responses. We propose that BFCNs differentially weigh predictions of positive and negative reinforcement, reflecting divergent relative salience of forecasting appetitive and aversive outcomes, partially explained by a simple reinforcement learning model of a valence-weighed unsigned prediction error. Finally, the extent of cue-driven cholinergic activation predicted subsequent decision speed, suggesting that the expectation-gated cholinergic firing is instructive to reward-seeking behaviors.
Collapse
Affiliation(s)
- Panna Hegedüs
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, H-1083 Budapest, Hungary,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, H-1085 Budapest, Hungary
| | - Katalin Sviatkó
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, H-1083 Budapest, Hungary,János Szentágothai Doctoral School of Neurosciences, Semmelweis University, H-1085 Budapest, Hungary
| | - Bálint Király
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, H-1083 Budapest, Hungary,Department of Biological Physics, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Sergio Martínez-Bellver
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, H-1083 Budapest, Hungary,Department of Anatomy and Human Embryology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, Institute of Experimental Medicine, H-1083 Budapest, Hungary,Corresponding author
| |
Collapse
|
49
|
Reich N, Hölscher C. Beyond Appetite: Acylated Ghrelin As A Learning, Memory and Fear Behavior-modulating Hormone. Neurosci Biobehav Rev 2022; 143:104952. [DOI: 10.1016/j.neubiorev.2022.104952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/27/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
|
50
|
Gieske A, Sommer T. Independent effects of emotional arousal and reward anticipation on episodic memory formation. Cereb Cortex 2022; 33:4527-4541. [PMID: 36205480 DOI: 10.1093/cercor/bhac359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/26/2022] [Accepted: 08/18/2022] [Indexed: 11/12/2022] Open
Abstract
Events that elicit emotional arousal or are associated with reward are more likely remembered. Emotional arousal activates the amygdala and the central noradrenergic system, whereas reward anticipation results in an activity in the mesocorticolimbic dopaminergic system. The activation of both pathways enhances memory formation in the hippocampus where their effects are based on similar neural substrates, e.g. tagging of active hippocampal synapses. Moreover, emotional arousal and reward anticipation both enhance attention, which can also affect memory formation. In addition, both neuromodulators interact on the cellular level. Therefore, we tested in the current functional magnetic resonance imaging study whether simultaneously occurring emotional arousal and reward anticipation might have interacting effects on memory formation. We did not find evidence for such an interaction, neither on the behavioral nor on the neural level. Our results further suggest that reward anticipation enhances memory formation rather by an increase in anticipation-related arousal-reflected in activity in the dorsal anterior cingulate cortex-and not dopaminergic midbrain activity. Accompanying behavioral experiments indicated that the effect of reward anticipation on memory is (i) caused at least to some extent by anticipating the speeded response to obtain the reward and not by the valance of the outcome and (ii) can be observed already immediately after encoding, i.e. before consolidation.
Collapse
Affiliation(s)
- Astrid Gieske
- Medical Center Hamburg-Eppendorf, Institute for Systems Neuroscience, Hamburg, Germany
| | - Tobias Sommer
- Medical Center Hamburg-Eppendorf, Institute for Systems Neuroscience, Hamburg, Germany
| |
Collapse
|