1
|
Pirri F, McCormick CM. Oxytocin receptors within the caudal lateral septum regulate social approach-avoidance, long-term social discrimination, and anxiety-like behaviors in adult male and female rats. Neuropharmacology 2025; 271:110409. [PMID: 40074168 DOI: 10.1016/j.neuropharm.2025.110409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
OTR signaling promotes social approach or facilitates social avoidance, depending on the brain region involved. The lateral septum plays a critical role in regulating social interactions and memory. We investigated the role of OTR signaling in the caudodorsal lateral septum (LSc.d) in modulating social approach-avoidance behavior, long-term social discrimination memory, and anxiety-like behaviors in adult rats. Local infusion of the selective OTR antagonist L-368,899 (1 μg/0.5 μl) into the LSc.d decreased social approach, increased social vigilance, and reduced long-term social discrimination memory in both sexes. Administration of the biased OTR/Gq agonist carbetocin (0.5 μg/0.5 μl) reduced social approach and long-term social discrimination memory in both sexes, and had anxiogenic effects (increased latency to consume palatable food in test arena) only in males. In contrast, the full OTR agonist TGOT (50 ng/0.5 μl) had no effect on social approach or long-term social discrimination memory, and decreased latency to consume palatable food (anxiolytic effect). The results indicate that the oxytocin system can both promote and inhibit social behaviors depending on the differential activation of G-protein subunits and β-arrestins, as well as the pivotal role of the LS in modulating social and anxiety-like behavior in rats.
Collapse
Affiliation(s)
- Fardad Pirri
- Biological Sciences Department, Brock University, Canada
| | - Cheryl M McCormick
- Biological Sciences Department, Brock University, Canada; Psychology Department, Brock University, Canada.
| |
Collapse
|
2
|
Li X, Wang X, Xue L, Luo L, Hu L, Jiang W. RAGE/AP-1/OTR signaling pathway in rat hippocampus DG involved in CUS induced depressive-like behaviors. Behav Brain Res 2025; 485:115540. [PMID: 40090553 DOI: 10.1016/j.bbr.2025.115540] [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: 11/04/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
There has been a growing body of evidence indicating that the oxytocin (OT) system plays a significant role in the neurophysiology of chronic stress-related mood disorders in recent years. However, the precise alterations for the OT system in response to chronic stress and the underlying mechanism remains unclear. The present study demonstrated that chronic unpredictable stress (CUS) resulted in a reduction in the expression of RAGE and OTR, as well as an inhibition of AP-1 phosphorylation. RAGE knockdown in hippocampus DG induced depressive-like behaviors, down-regulated the OTR protein and mRNA levels, and reduced the AP-1 phosphorylation. The administration of OT via the nasal route reversed the depressive-like behaviors induced by RAGE knockdown, increased the levels of BDNF expression and AP-1 phosphorylation. On the other hand, RAGE over-expression in the hippocampus DG resisted the effects of CUS on depression-like behaviors, AP-1 phosphorylation, and OTR expression. These finding suggested that RAGE signaling pathway is involved in CUS induced depressive-like behaviors at least partially by regulating OTR expression.
Collapse
Affiliation(s)
- Xuemei Li
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xin Wang
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Lifen Xue
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Lan Luo
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Lingxiao Hu
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Wengao Jiang
- Key Laboratory of Molecular and Biochemical Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Song SC, Froemke RC. Lateralized local circuit tuning in female mouse auditory cortex. Neurosci Res 2025:S0168-0102(25)00068-9. [PMID: 40189152 DOI: 10.1016/j.neures.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Most offspring are born helpless, requiring intense caregiving from parents especially during the first few days of neonatal life. For many species, infant cries are a primary signal used by parents to provide caregiving. Previously we and others documented how maternal left auditory cortex rapidly becomes sensitized to pup calls over hours of parental experience, enabled by oxytocin. The speed and robustness of this maternal plasticity suggests cortical pre-tuning or initial bias for pup call stimulus features. Here we examine the circuit basis of left-lateralized tuning to vocalization features with whole-cell recordings in brain slices. We found that layer 2/3 pyramidal cells of female left auditory cortex show selective suppression of inhibitory inputs with repeated stimulation at the fundamental pup call rate (inter-stimulus interval ∼150 msec) in pup-naïve females and expanded with maternal experience. However, optogenetic stimulation of cortical inhibitory cells showed that inputs from somatostatin-positive and oxytocin-receptor-expressing interneurons were less suppressed at these rates. This suggested that disynaptic inhibition rather than monosynaptic depression was a major mechanism underlying pre-tuning of cortical excitatory neurons, confirmed with simulations. Thus cortical interneuron specializations can augment neuroplasticity mechanisms to ensure fast appropriate caregiving in response to infant cries.
Collapse
Affiliation(s)
- Soomin C Song
- Ion Laboratory, New York University Langone Health, New York, NY, USA; Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Robert C Froemke
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
4
|
Bair-Marshall CJ, Cassel NL, Agha AA, Bkhiet M, Froemke RC. Neural circuit plasticity transforms infant neglect into maternal care. RESEARCH SQUARE 2025:rs.3.rs-5983736. [PMID: 40060051 PMCID: PMC11888542 DOI: 10.21203/rs.3.rs-5983736/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Infants in distress evoke strong emotional responses in adults, which help to elicit caretaking behaviors from parents to ensure infant needs are met1-3. However, neonatal care can also be challenging, and interactions with infants can lead to stress and negative affect even in potential caregivers4-7. Child neglect and maltreatment rates in human populations make it important to understand the neural mechanisms of regulating negative emotions and stress in the parental brain8-10. Here we show how rapid plasticity in female mouse central amygdala (CeA) transforms infant aversion into attentiveness after initial pup experience. Projections from CeA to locus coeruleus (CeA→LC) were strongly activated upon initial pup contact leading to pup aversion. CeA→LC pup responses were reduced with parental experience and down-regulating CeA→LC activity led to less aversion. Oxytocin signaling in central amygdala was required to switch pup aversion to attention, inducing rapid long-term depression of excitatory inputs onto projection neurons. CeA→LC projectors released the stress hormone corticotropin-releasing factor onto LC neurons, modulating phasic firing to regulate attention. This circuit organization enables local CeA computations of pup valence to be broadcast throughout the brain by the LC central arousal system, leading to appropriate pup-directed behaviors depending on adult state.
Collapse
Affiliation(s)
- Chloe J Bair-Marshall
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016 USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Naomi L Cassel
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016 USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
- Barnard College, New York, NY 10027, USA
| | - Ayat A Agha
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016 USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Malak Bkhiet
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016 USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
- Barnard College, New York, NY 10027, USA
| | - Robert C Froemke
- Neuroscience Institute, New York University School of Medicine, New York, NY 10016 USA
- Department of Otolaryngology, New York University School of Medicine, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10003, USA
| |
Collapse
|
5
|
Robson E, Donahue MM, Mably AJ, Demetrovich PG, Hewitt LT, Colgin LL. Social odors drive hippocampal CA2 place cell responses to social stimuli. Prog Neurobiol 2025; 245:102708. [PMID: 39743170 PMCID: PMC11827691 DOI: 10.1016/j.pneurobio.2024.102708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/06/2024] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
Hippocampal region CA2 is essential for social memory processing. Interaction with social stimuli induces changes in CA2 place cell firing during active exploration and sharp wave-ripples during rest following a social interaction. However, it is unknown whether these changes in firing patterns are caused by integration of multimodal social stimuli or by a specific sensory modality associated with a social interaction. Rodents rely heavily on chemosensory cues in the form of olfactory signals for social recognition processes. To determine the extent to which social olfactory signals contribute to CA2 place cell responses to social stimuli, we recorded CA2 place cells in rats freely exploring environments containing stimuli that included or lacked olfactory content. We found that CA2 place cell firing patterns significantly changed only when social odors were prominent. Also, place cells that increased their firing in the presence of social odors alone preferentially increased their firing during subsequent sharp wave-ripples. Our results suggest that social olfactory cues are essential for changing CA2 place cell firing patterns during and after social interactions. These results support prior work suggesting CA2 performs social functions and shed light on processes underlying CA2 responses to social stimuli.
Collapse
Affiliation(s)
- Emma Robson
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Margaret M Donahue
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Alexandra J Mably
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Peyton G Demetrovich
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Lauren T Hewitt
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States
| | - Laura Lee Colgin
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, United States; Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States.
| |
Collapse
|
6
|
Belge JB, Geenen V, Salado AL, Kaschten B, Martin D, Scantamburlo G. Case report: Non-linear evolution of oxytocin informs YBOCS changes post-DBS of the bed nucleus of the stria terminalis for treatment resistant OCD. Front Psychiatry 2025; 15:1473797. [PMID: 39931193 PMCID: PMC11807955 DOI: 10.3389/fpsyt.2024.1473797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/09/2024] [Indexed: 02/13/2025] Open
Abstract
Introduction Obsessive-compulsive disorder (OCD) is a challenging neuropsychiatric condition with a subset of patients remaining refractory to conventional treatments. Deep brain stimulation (DBS) of the bed nucleus of the stria terminalis (BNST) has shown promise for severe, treatment-resistant OCD. This case report examines the relationship between plasma oxytocin levels and OCD symptom severity following BNST-DBS. Methods A 36-year-old patient with long-standing, treatment-resistant OCD underwent stereotactic implantation of DBS electrodes at the BNST. Postoperative assessments included OCD symptom severity using the Yale-Brown Obsessive Compulsive Scale (YBOCS) and plasma oxytocin levels, measured at 12 time points over three years. Longitudinal and correlational analyses were performed using linear and polynomial regression models. Results Non-linear trends in oxytocin levels were identified, with polynomial regression revealing a significant quadratic term, suggesting a parabolic trend. Strong positive correlations were found between changes in oxytocin levels and YBOCS total, obsession, and compulsion scores. Conclusion The findings suggest a significant non-linear evolution of oxytocin levels and a positive correlation with OCD symptom changes following BNST-DBS. Oxytocin levels could serve as a biomarker for DBS efficacy if this finding is replicated in larger studies.
Collapse
Affiliation(s)
- Jean-Baptiste Belge
- Department of Psychiatry, Liège University Hospital, University of Liège, Liège, Belgium
- Psycho-Neuro-Endocrinology Unit, University of Liège, Liège, Belgium
| | - Vincent Geenen
- GIGA Research Institute, GIGA-Immunity, Inflammation and Infection (GIGA-I3), University of Liège, Liège, Belgium
| | - Anne L. Salado
- Department of Neurosurgery, Liège University Hospital, University of Liège, Liège, Belgium
| | - Bruno Kaschten
- Department of Neurosurgery, Liège University Hospital, University of Liège, Liège, Belgium
| | - Didier Martin
- Department of Neurosurgery, Liège University Hospital, University of Liège, Liège, Belgium
| | - Gabrielle Scantamburlo
- Department of Psychiatry, Liège University Hospital, University of Liège, Liège, Belgium
- Psycho-Neuro-Endocrinology Unit, University of Liège, Liège, Belgium
| |
Collapse
|
7
|
Sarahian N, Khodagholi F, Valian N, Ahmadiani A. Interplay of MeCP2/REST/Synaptophysin-BDNF and intranasal oxytocin influence on Aβ-induced memory and cognitive impairments. Behav Brain Res 2025; 476:115235. [PMID: 39236931 DOI: 10.1016/j.bbr.2024.115235] [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: 05/07/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is linked to the accumulation of Aβ, increased tau hyperphosphorylation, persistent neuroinflammation, and a decline in neurotrophic factors, neurogenesis, and synaptic plasticity. Oxytocin (OT) has a significant impact on memory and learning. We examined the influence of intranasal (IN) OT on synaptic plasticity, neurogenesis, histone acetylation, and spatial and cognitive memories in rats. METHODS Aβ25-35 (5 µg/2.5 µl) was administered bilaterally in the CA1 of male Wistar rats for four consecutive days. After seven days of recovery, OT (2 µg/µl, 10 µl in each nostril) was administered IN for seven consecutive days. Working, spatial, and cognitive memories, and gene expression of neurogenesis- and synaptic plasticity-involved factors were measured in the hippocampus. Histone acetylation (H3K9 and H4K8) was also measured using western blotting. RESULTS IN administration of OT significantly improved working and spatial memory impairment induced by Aβ and increased the factors involved in synaptic plasticity (MeCP2, REST, synaptophysin, and BDNF) and neurogenesis (Ki67 and DCX). We also found an enhancement in the levels of H3K9ac and H4K8ac following OT administration. CONCLUSION These findings indicated that IN OT could improve hippocampus-related behaviors by increasing synaptic plasticity, stimulating neurogenesis, and chromatin plasticity.
Collapse
Affiliation(s)
- Nahid Sarahian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| |
Collapse
|
8
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
9
|
Inada K. Neurobiological mechanisms underlying oxytocin-mediated parental behavior in rodents. Neurosci Res 2024; 207:1-12. [PMID: 38642676 DOI: 10.1016/j.neures.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/22/2024]
Abstract
Parental behavior is essential for mammalian offspring to survive. Because of this significance, elucidating the neurobiological mechanisms that facilitate parental behavior has received strong interest. Decades of studies utilizing pharmacology and molecular biology have revealed that in addition to its facilitatory effects on parturition and lactation, oxytocin (OT) promotes the expression of parental behavior in rodents. Recent studies have also described the modulation of sensory processing by OT and the interaction of the OT system with other brain regions associated with parental behavior. However, the precise neurobiological mechanisms underlying the facilitation of caregiving behaviors by OT remain unclear. In this Review, I summarize the findings from rats and mice with a view toward integrating past and recent progress. I then review recent advances in the understanding of the molecular, cellular, and circuit mechanisms of OT-mediated parental behavior. Based on these observations, I propose a hypothetical model that would explain the mechanisms underlying OT-mediated parental behavior. Finally, I conclude by discussing some major remaining questions and propose potential future research directions.
Collapse
Affiliation(s)
- Kengo Inada
- RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
10
|
Robson E, Donahue MM, Mably AJ, Demetrovich PG, Hewitt LT, Colgin LL. Social odors drive hippocampal CA2 place cell responses to social stimuli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603738. [PMID: 39071428 PMCID: PMC11275720 DOI: 10.1101/2024.07.16.603738] [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: 07/30/2024]
Abstract
Hippocampal region CA2 is essential for social memory processing. Interaction with social stimuli induces changes in CA2 place cell firing during active exploration and sharp wave-ripples during rest following a social interaction. However, it is unknown whether these changes in firing patterns are caused by integration of multimodal social stimuli or by a specific sensory modality associated with a social interaction. Rodents rely heavily on chemosensory cues in the form of olfactory signals for social recognition processes. To determine the extent to which social olfactory signals contribute to CA2 place cell responses to social stimuli, we recorded CA2 place cells in rats freely exploring environments containing stimuli that included or lacked olfactory content. We found that CA2 place cell firing patterns significantly changed only when social odors were prominent. Also, place cells that increased their firing in the presence of social odors alone preferentially increased their firing during subsequent sharp wave-ripples. Our results suggest that social olfactory cues are essential for changing CA2 place cell firing patterns during and after social interactions. These results support prior work suggesting CA2 performs social functions and shed light on processes underlying CA2 responses to social stimuli.
Collapse
Affiliation(s)
- Emma Robson
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Margaret M. Donahue
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Alexandra J. Mably
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Peyton G. Demetrovich
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Lauren T. Hewitt
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712
| | - Laura Lee Colgin
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712
| |
Collapse
|
11
|
Rolls ET, Treves A. A theory of hippocampal function: New developments. Prog Neurobiol 2024; 238:102636. [PMID: 38834132 DOI: 10.1016/j.pneurobio.2024.102636] [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: 01/27/2024] [Revised: 04/15/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
We develop further here the only quantitative theory of the storage of information in the hippocampal episodic memory system and its recall back to the neocortex. The theory is upgraded to account for a revolution in understanding of spatial representations in the primate, including human, hippocampus, that go beyond the place where the individual is located, to the location being viewed in a scene. This is fundamental to much primate episodic memory and navigation: functions supported in humans by pathways that build 'where' spatial view representations by feature combinations in a ventromedial visual cortical stream, separate from those for 'what' object and face information to the inferior temporal visual cortex, and for reward information from the orbitofrontal cortex. Key new computational developments include the capacity of the CA3 attractor network for storing whole charts of space; how the correlations inherent in self-organizing continuous spatial representations impact the storage capacity; how the CA3 network can combine continuous spatial and discrete object and reward representations; the roles of the rewards that reach the hippocampus in the later consolidation into long-term memory in part via cholinergic pathways from the orbitofrontal cortex; and new ways of analysing neocortical information storage using Potts networks.
Collapse
Affiliation(s)
- Edmund T Rolls
- Oxford Centre for Computational Neuroscience, Oxford, UK; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK.
| | | |
Collapse
|
12
|
Walia V, Wal P, Mishra S, Agrawal A, Kosey S, Dilipkumar Patil A. Potential role of oxytocin in the regulation of memories and treatment of memory disorders. Peptides 2024; 177:171222. [PMID: 38649032 DOI: 10.1016/j.peptides.2024.171222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 04/25/2024]
Abstract
Oxytocin (OXT) is an "affiliative" hormone or neurohormone or neuropeptide consists of nine amino acids, synthesized in magnocellular neurons of paraventricular (PVN) and supraoptic nuclei (SON) of hypothalamus. OXT receptors are widely distributed in various region of brain and OXT has been shown to regulate various social and nonsocial behavior. Hippocampus is the main region which regulates the learning and memory. Hippocampus particularly regulates the acquisition of new memories and retention of acquired memories. OXT has been shown to regulate the synaptic plasticity, neurogenesis, and consolidation of memories. Further, findings from both preclinical and clinical studies have suggested that the OXT treatment improves performance in memory related task. Various trials have suggested the positive impact of intranasal OXT in the dementia patients. However, these studies are limited in number. In the present study authors have highlighted the role of OXT in the formation and retrieval of memories. Further, the study demonstrated the outcome of OXT treatment in various memory and related disorders.
Collapse
Affiliation(s)
- Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India.
| | - Pranay Wal
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kanpur, UP 209305, India
| | - Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India
| | - Ankur Agrawal
- Jai Institute of Pharmaceutical Sciences and Research, Gwalior, MP, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Aditya Dilipkumar Patil
- Founder, Tech Hom Research Solutions (THRS), Plot no. 38, 1st floor, opposite to biroba mandir, near ST stand, Satara, Maharashtra 415110, India
| |
Collapse
|
13
|
Hadler MD, Alle H, Geiger JRP. Parvalbumin interneuron cell-to-network plasticity: mechanisms and therapeutic avenues. Trends Pharmacol Sci 2024; 45:586-601. [PMID: 38763836 DOI: 10.1016/j.tips.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) and schizophrenia (SCZ) represent two major neuropathological conditions with a high disease burden. Despite their distinct etiologies, patients suffering from AD or SCZ share a common burden of disrupted memory functions unattended by current therapies. Recent preclinical analyses highlight cell-type-specific contributions of parvalbumin interneurons (PVIs), particularly the plasticity of their cellular excitability, towards intact neuronal network function (cell-to-network plasticity) and memory performance. Here we argue that deficits of PVI cell-to-network plasticity may underlie memory deficits in AD and SCZ, and we explore two therapeutic avenues: the targeting of PVI-specific neuromodulation, including by neuropeptides, and the recruitment of network synchrony in the gamma frequency range (40 Hz) by external stimulation. We finally propose that these approaches be merged under consideration of recent insights into human brain physiology.
Collapse
Affiliation(s)
- Michael D Hadler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R P Geiger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
14
|
George K, Hoang HT, Tibbs T, Nagaraja RY, Li G, Troyano-Rodriguez E, Ahmad M. Robust GRK2/3/6-dependent desensitization of oxytocin receptor in neurons. iScience 2024; 27:110047. [PMID: 38883814 PMCID: PMC11179071 DOI: 10.1016/j.isci.2024.110047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/22/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Oxytocin plays critical roles in the brain as a neuromodulator, regulating social and other affective behavior. However, the regulatory mechanisms controlling oxytocin receptor (OXTR) signaling in neurons remain unexplored. In this study, we have identified robust and rapid-onset desensitization of OXTR response in multiple regions of the mouse brain. Both cell autonomous spiking response and presynaptic activation undergo similar agonist-induced desensitization. G-protein-coupled receptor kinases (GRK) GRK2, GRK3, and GRK6 are recruited to the activated OXTR in neurons, followed by recruitment of β-arrestin-1 and -2. Neuronal OXTR desensitization was impaired by suppression of GRK2/3/6 kinase activity but remained unaltered with double knockout of β-arrestin-1 and -2. Additionally, we observed robust agonist-induced internalization of neuronal OXTR and its Rab5-dependent recruitment to early endosomes, which was impaired by GRK2/3/6 inhibition. This work defines distinctive aspects of the mechanisms governing OXTR desensitization and internalization in neurons compared to prior studies in heterologous cells.
Collapse
Affiliation(s)
- Kiran George
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hanh T.M. Hoang
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Taryn Tibbs
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Raghavendra Y. Nagaraja
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Guangpu Li
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eva Troyano-Rodriguez
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mohiuddin Ahmad
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Zhang Y, Karadas M, Liu J, Gu X, Vöröslakos M, Li Y, Tsien RW, Buzsáki G. Interaction of acetylcholine and oxytocin neuromodulation in the hippocampus. Neuron 2024; 112:1862-1875.e5. [PMID: 38537642 PMCID: PMC11156550 DOI: 10.1016/j.neuron.2024.02.021] [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: 09/11/2023] [Revised: 01/17/2024] [Accepted: 02/29/2024] [Indexed: 06/09/2024]
Abstract
A postulated role of subcortical neuromodulators is to control brain states. Mechanisms by which different neuromodulators compete or cooperate at various temporal scales remain an open question. We investigated the interaction of acetylcholine (ACh) and oxytocin (OXT) at slow and fast timescales during various brain states. Although these neuromodulators fluctuated in parallel during NREM packets, transitions from NREM to REM were characterized by a surge of ACh but a continued decrease of OXT. OXT signaling lagged behind ACh. High ACh was correlated with population synchrony and gamma oscillations during active waking, whereas minimum ACh predicts sharp-wave ripples (SPW-Rs). Optogenetic control of ACh and OXT neurons confirmed the active role of these neuromodulators in the observed correlations. Synchronous hippocampal activity consistently reduced OXT activity, whereas inactivation of the lateral septum-hypothalamus path attenuated this effect. Our findings demonstrate how cooperative actions of these neuromodulators allow target circuits to perform specific functions.
Collapse
Affiliation(s)
| | | | | | - Xinyi Gu
- Neuroscience Institute, New York, NY, USA
| | | | - Yulong Li
- School of Life Science, Peking University, Beijing, China
| | - Richard W Tsien
- Neuroscience Institute, New York, NY, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - György Buzsáki
- Neuroscience Institute, New York, NY, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
16
|
Siegler PN, Shaughnessy EK, Horman B, Vierling TT, King DH, Patisaul HB, Huhman KL, Alexander GM, Dudek SM. Identification of hippocampal area CA2 in hamster and vole brain. J Comp Neurol 2024; 532:e25603. [PMID: 38497661 PMCID: PMC10950058 DOI: 10.1002/cne.25603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/24/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
Prairie voles (Microtus ochrogaster) and Syrian, or golden, hamsters (Mesocricetus auratus) are closely related to mice (Mus musculus) and are commonly used in studies of social behavior including social interaction, social memory, and aggression. Hippocampal area CA2 is known to play a key role in these behaviors in mice and responds to social stimuli in rats, but CA2 has yet to be characterized in hamsters or voles, which are also used in studies of social behaviors. Here, we used immunofluorescence to determine whether CA2 could be molecularly identified in tissue from voles and hamsters. We found that staining for many CA2 markers was similar in these three species, with labeling seen in neurons at the distal end of the mossy fibers . In contrast, although perineuronal nets (PNNs) surround CA2 cells in mice, PNN staining differed across species. In voles, both CA2 and CA3 were labeled, whereas in hamsters, labeling was seen primarily in CA3. These results demonstrate that CA2 can be molecularly distinguished from neighboring CA1 and CA3 areas in voles and hamsters with several antibodies commonly used in mice. However, PNN staining is not useful for identifying CA2 in voles or hamsters, suggestive of differing roles for either PNNs or for the hippocampal subregions in social behavior. These findings reveal commonalities across species in the molecular profile of CA2 and should facilitate future studies of CA2 in these species.
Collapse
Affiliation(s)
- Preston Nicole Siegler
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC
| | | | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Tia T. Vierling
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Darron H. King
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Heather B. Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Kim L. Huhman
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | - Georgia M. Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| |
Collapse
|
17
|
Castagno AN, Spaiardi P, Trucco A, Maniezzi C, Raffin F, Mancini M, Nicois A, Cazzola J, Pedrinazzi M, Del Papa P, Pisani A, Talpo F, Biella GR. Oxytocin Modifies the Excitability and the Action Potential Shape of the Hippocampal CA1 GABAergic Interneurons. Int J Mol Sci 2024; 25:2613. [PMID: 38473860 DOI: 10.3390/ijms25052613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Oxytocin (OT) is a neuropeptide that modulates social-related behavior and cognition in the central nervous system of mammals. In the CA1 area of the hippocampus, the indirect effects of the OT on the pyramidal neurons and their role in information processing have been elucidated. However, limited data are available concerning the direct modulation exerted by OT on the CA1 interneurons (INs) expressing the oxytocin receptor (OTR). Here, we demonstrated that TGOT (Thr4,Gly7-oxytocin), a selective OTR agonist, affects not only the membrane potential and the firing frequency but also the neuronal excitability and the shape of the action potentials (APs) of these INs in mice. Furthermore, we constructed linear mixed-effects models (LMMs) to unravel the dependencies between the AP parameters and the firing frequency, also considering how TGOT can interact with them to strengthen or weaken these influences. Our analyses indicate that OT regulates the functionality of the CA1 GABAergic INs through different and independent mechanisms. Specifically, the increase in neuronal firing rate can be attributed to the depolarizing effect on the membrane potential and the related enhancement in cellular excitability by the peptide. In contrast, the significant changes in the AP shape are directly linked to oxytocinergic modulation. Importantly, these alterations in AP shape are not associated with the TGOT-induced increase in neuronal firing rate, being themselves critical for signal processing.
Collapse
Affiliation(s)
- Antonio Nicolas Castagno
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Paolo Spaiardi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
- INFN-Pavia Section, 27100 Pavia, Italy
| | - Arianna Trucco
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Claudia Maniezzi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy
| | - Francesca Raffin
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Maria Mancini
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Alessandro Nicois
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), 80078 Pozzuoli, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Jessica Cazzola
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Matilda Pedrinazzi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Paola Del Papa
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
- IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Francesca Talpo
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Gerardo Rosario Biella
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, 27100 Pavia, Italy
- INFN-Pavia Section, 27100 Pavia, Italy
| |
Collapse
|
18
|
Siegler PN, Shaughnessy EK, Horman B, Vierling TT, King DH, Patisaul HB, Huhman KL, Alexander GM, Dudek SM. Identification of hippocampal area CA2 in hamster and vole brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579957. [PMID: 38405991 PMCID: PMC10888814 DOI: 10.1101/2024.02.12.579957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Prairie voles (Microtus ochrogaster) and Syrian, or golden, hamsters (Mesocricetus auratus) are closely related to mice (Mus musculus) and rats (Rattus norvegicus, for example) and are commonly used in studies of social behavior including social interaction, social memory, and aggression. The CA2 region of the hippocampus is known to play a key role in social memory and aggression in mice and responds to social stimuli in rats, likely owing to its high expression of oxytocin and vasopressin 1b receptors. However, CA2 has yet to be identified and characterized in hamsters or voles. In this study, we sought to determine whether CA2 could be identified molecularly in vole and hamster. To do this, we used immunofluorescence with primary antibodies raised against known molecular markers of CA2 in mice and rats to stain hippocampal sections from voles and hamsters in parallel with those from mice. Here, we report that, like in mouse and rat, staining for many CA2 proteins in vole and hamster hippocampus reveals a population of neurons that express regulator of G protein signaling 14 (RGS14), Purkinje cell protein 4 (PCP4) and striatal-enriched protein tyrosine phosphatase (STEP), which together delineate the borders with CA3 and CA1. These cells were located at the distal end of the mossy fiber projections, marked by the presence of Zinc Transporter 3 (ZnT-3) and calbindin in all three species. In addition to staining the mossy fibers, calbindin also labeled a layer of CA1 pyramidal cells in mouse and hamster but not in vole. However, Wolframin ER transmembrane glycoprotein (WFS1) immunofluorescence, which marks all CA1 neurons, was present in all three species and abutted the distal end of CA2, marked by RGS14 immunofluorescence. Staining for two stress hormone receptors-the glucocorticoid (GR) and mineralocorticoid (MR) receptors-was also similar in all three species, with GR staining found primarily in CA1 and MR staining enriched in CA2. Interestingly, although perineuronal nets (PNNs) are known to surround CA2 cells in mouse and rat, we found that staining for PNNs differed across species in that both CA2 and CA3 showed staining in voles and primarily CA3 in hamsters with only some neurons in proximal CA2 showing staining. These results demonstrate that, like in mouse, CA2 in voles and hamsters can be molecularly distinguished from neighboring CA1 and CA3 areas, but PNN staining is less useful for identifying CA2 in the latter two species. These findings reveal commonalities across species in molecular profile of CA2, which will facilitate future studies of CA2 in these species. Yet to be determined is how differences in PNNs might relate to differences in social behavior across species.
Collapse
Affiliation(s)
- Preston Nicole Siegler
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC
| | | | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Tia T. Vierling
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Darron H. King
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Heather B. Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Kim L. Huhman
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | - Georgia M. Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| |
Collapse
|
19
|
Wei X, Wang J, Yang E, Zhang Y, Qian Q, Li X, Huang F, Sun B. Efr3b is essential for social recognition by modulating the excitability of CA2 pyramidal neurons. Proc Natl Acad Sci U S A 2024; 121:e2314557121. [PMID: 38190534 PMCID: PMC10801834 DOI: 10.1073/pnas.2314557121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
CA2 pyramidal neurons (PNs) are associated with social behaviors. The mechanisms, however, remain to be fully investigated. Here, we report that Efr3b, a protein essential for phospholipid metabolism at the plasma membrane, is widely expressed in the brain, especially in the hippocampal CA2/CA3 areas. To assess the functional significance of Efr3b in the brain, we generated Efr3bf/f mice and crossed them with Nestin-cre mice to delete Efr3b specifically in the brain. We find that Efr3b deficiency in the brain leads to deficits of social novelty recognition and hypoexcitability of CA2 PNs. We then knocked down the expression of Efr3b specifically in CA2 PNs of C57BL/6J mice, and our results showed that reducing Efr3b in CA2 PNs also resulted in deficits of social novelty recognition and hypoexcitability of CA2 PNs. More interestingly, restoring the expression of Efr3b in CA2 PNs enhances their excitability and improves social novelty recognition in Efr3b-deficient mice. Furthermore, direct activation of CA2 PNs with chemogenetics improves social behaviors in Efr3b-deficient mice. Together, our data suggest that Efr3b is essential for social novelty by modulating the excitability of CA2 PNs.
Collapse
Affiliation(s)
- Xiaojie Wei
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
- Children’s Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310020, China
| | - Jing Wang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
- School of Medicine, Shaoxing University, Shaoxing312000, China
| | - Enlu Yang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Yiping Zhang
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Qi Qian
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| | - Xuekun Li
- Children’s Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou310020, China
| | - Fude Huang
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing100190, China
- Nuo-Beta Pharmaceutical Technology, Co. Ltd., Shanghai201210, China
| | - Binggui Sun
- Department of Anesthesiology of the Children’s Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Zhejiang University, Hangzhou310058, China
- National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou310058, China
| |
Collapse
|
20
|
Ngodup T, Irie T, Elkins SP, Trussell LO. The Na + leak channel NALCN controls spontaneous activity and mediates synaptic modulation by α2-adrenergic receptors in auditory neurons. eLife 2024; 12:RP89520. [PMID: 38197879 PMCID: PMC10945507 DOI: 10.7554/elife.89520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Cartwheel interneurons of the dorsal cochlear nucleus (DCN) potently suppress multisensory signals that converge with primary auditory afferent input, and thus regulate auditory processing. Noradrenergic fibers from locus coeruleus project to the DCN, and α2-adrenergic receptors inhibit spontaneous spike activity but simultaneously enhance synaptic strength in cartwheel cells, a dual effect leading to enhanced signal-to-noise for inhibition. However, the ionic mechanism of this striking modulation is unknown. We generated a glycinergic neuron-specific knockout of the Na+ leak channel NALCN in mice and found that its presence was required for spontaneous firing in cartwheel cells. Activation of α2-adrenergic receptors inhibited both NALCN and spike generation, and this modulation was absent in the NALCN knockout. Moreover, α2-dependent enhancement of synaptic strength was also absent in the knockout. GABAB receptors mediated inhibition through NALCN as well, acting on the same population of channels as α2 receptors, suggesting close apposition of both receptor subtypes with NALCN. Thus, multiple neuromodulatory systems determine the impact of synaptic inhibition by suppressing the excitatory leak channel, NALCN.
Collapse
Affiliation(s)
- Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Tomohiko Irie
- Department of Physiology, Kitasato University School of MedicineSagamiharaJapan
| | - Seán P Elkins
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
21
|
Menon R, Neumann ID. Detection, processing and reinforcement of social cues: regulation by the oxytocin system. Nat Rev Neurosci 2023; 24:761-777. [PMID: 37891399 DOI: 10.1038/s41583-023-00759-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/29/2023]
Abstract
Many social behaviours are evolutionarily conserved and are essential for the healthy development of an individual. The neuropeptide oxytocin (OXT) is crucial for the fine-tuned regulation of social interactions in mammals. The advent and application of state-of-the-art methodological approaches that allow the activity of neuronal circuits involving OXT to be monitored and functionally manipulated in laboratory mammals have deepened our understanding of the roles of OXT in these behaviours. In this Review, we discuss how OXT promotes the sensory detection and evaluation of social cues, the subsequent approach and display of social behaviour, and the rewarding consequences of social interactions in selected reproductive and non-reproductive social behaviours. Social stressors - such as social isolation, exposure to social defeat or social trauma, and partner loss - are often paralleled by maladaptations of the OXT system, and restoring OXT system functioning can reinstate socio-emotional allostasis. Thus, the OXT system acts as a dynamic mediator of appropriate behavioural adaptations to environmental challenges by enhancing and reinforcing social salience and buffering social stress.
Collapse
Affiliation(s)
- Rohit Menon
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
22
|
Ngodup T, Irie T, Elkins S, Trussell LO. The Na + leak channel NALCN controls spontaneous activity and mediates synaptic modulation by α2-adrenergic receptors in auditory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.23.546323. [PMID: 37987013 PMCID: PMC10659375 DOI: 10.1101/2023.06.23.546323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Cartwheel interneurons of the dorsal cochlear nucleus (DCN) potently suppress multisensory signals that converge with primary auditory afferent input, and thus regulate auditory processing. Noradrenergic fibers from locus coeruleus project to the DCN, and α2-adrenergic receptors inhibit spontaneous spike activity but simultaneously enhance synaptic strength in cartwheel cells, a dual effect leading to enhanced signal-to-noise for inhibition. However, the ionic mechanism of this striking modulation is unknown. We generated a glycinergic neuron-specific knockout of the Na+ leak channel NALCN, and found that its presence was required for spontaneous firing in cartwheel cells. Activation of α2-adrenergic receptors inhibited both NALCN and spike generation, and this modulation was absent in the NALCN knockout. Moreover, α2-dependent enhancement of synaptic strength was also absent in the knockout. GABAB receptors mediated inhibition through NALCN as well, acting on the same population of channels as α2 receptors, suggesting close apposition of both receptor subtypes with NALCN. Thus, multiple neuromodulatory systems determine the impact of synaptic inhibition by suppressing the excitatory leak channel, NALCN.
Collapse
Affiliation(s)
- Tenzin Ngodup
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| | - Tomohiko Irie
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Sean Elkins
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland OR USA
| |
Collapse
|
23
|
Hung YC, Wu YJ, Chien ME, Lin YT, Tsai CF, Hsu KS. Loss of oxytocin receptors in hilar mossy cells impairs social discrimination. Neurobiol Dis 2023; 187:106311. [PMID: 37769745 DOI: 10.1016/j.nbd.2023.106311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023] Open
Abstract
Hippocampal oxytocin receptor (OXTR) signaling is crucial for discrimination of social stimuli to guide social recognition, but circuit mechanisms and cell types involved remain incompletely understood. Here, we report a role for OXTR-expressing hilar mossy cells (MCs) of the dentate gyrus in social stimulus discrimination by regulating granule cell (GC) activity. Using a Cre-loxP recombination approach, we found that ablation of Oxtr from MCs impairs discrimination of social, but not object, stimuli in adult male mice. Ablation of MC Oxtr increases spontaneous firing rate of GCs, synaptic excitation to inhibition ratio of MC-to-GC circuit, and GC firing when temporally associated with the lateral perforant path inputs. Using mouse hippocampal slices, we found that bath application of OXTR agonist [Thr4,Gly7]-oxytocin causes membrane depolarization and increases MC firing activity. Optogenetic activation of MC-to-GC circuit ameliorates social discrimination deficit in MC OXTR deficient mice. Together, our results uncover a previously unknown role of MC OXTR signaling for discrimination of social stimuli and delineate a MC-to-GC circuit responsible for social information processing.
Collapse
Affiliation(s)
- Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Jen Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan; Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Miao-Er Chien
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan
| | - Yu-Ting Lin
- Institute of Systems Neuroscience, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Fang Tsai
- Department of Physical Medicine and Rehabilitation, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
24
|
Althammer F. Heralding a new era of oxytocinergic research: New tools, new problems? J Neuroendocrinol 2023; 35:e13333. [PMID: 37621199 DOI: 10.1111/jne.13333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
According to classic neuroendocrinology, hypothalamic oxytocin cells can be categorized into parvo- and magnocellular neurons. However, research in the last decade provided ample evidence that this black-and-white model of oxytocin neurons is most likely oversimplified. Novel genetic, functional and morphological studies indicate that oxytocin neurons might be organized in functional modules and suggest the existence of five or more distinct oxytocinergic subpopulations. However, many of these novel, automated high-throughput techniques might be inherently biased and interpretation of acquired data needs to be approached with caution to enable drawing sound and reliable conclusions. In addition, the recent finding that astrocytes in various brain regions express functional oxytocin receptors represents a paradigm shift and challenges the view that oxytocin primarily acts as a direct peptidergic neurotransmitter. This review highlights the latest technical advances in oxytocinergic research, puts recent studies on the oxytocin system into context and formulates various provocative ideas based on novel findings that challenges various prevailing hypotheses and dogmas about oxytocinergic modulation.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
25
|
Shen LP, Li W, Pei LZ, Yin J, Xie ST, Li HZ, Yan C, Wang JJ, Zhang Q, Zhang XY, Zhu JN. Oxytocin Receptor in Cerebellar Purkinje Cells Does Not Engage in Autism-Related Behaviors. CEREBELLUM (LONDON, ENGLAND) 2023; 22:888-904. [PMID: 36040660 DOI: 10.1007/s12311-022-01466-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
The classical motor center cerebellum is one of the most consistent structures of abnormality in autism spectrum disorders (ASD), and neuropeptide oxytocin is increasingly explored as a potential pharmacotherapy for ASD. However, whether oxytocin targets the cerebellum for therapeutic effects remains unclear. Here, we report a localization of oxytocin receptor (OXTR) in Purkinje cells (PCs) of cerebellar lobule Crus I, which is functionally connected with ASD-implicated circuits. OXTR activation neither affects firing activities, intrinsic excitability, and synaptic transmission of normal PCs nor improves abnormal intrinsic excitability and synaptic transmission of PCs in maternal immune activation (MIA) mouse model of autism. Furthermore, blockage of OXTR in Crus I in wild-type mice does not induce autistic-like social, stereotypic, cognitive, and anxiety-like behaviors. These results suggest that oxytocin signaling in Crus I PCs seems to be uninvolved in ASD pathophysiology, and contribute to understanding of targets and mechanisms of oxytocin in ASD treatment.
Collapse
Affiliation(s)
- Li-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wei Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ling-Zhu Pei
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jun Yin
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shu-Tao Xie
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hong-Zhao Li
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jian-Jun Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
| | - Qipeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Xiao-Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| | - Jing-Ning Zhu
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Physiology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute for Brain Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
26
|
Hassan SI, Bigler S, Siegelbaum SA. Social odor discrimination and its enhancement by associative learning in the hippocampal CA2 region. Neuron 2023; 111:2232-2246.e5. [PMID: 37192623 PMCID: PMC10524117 DOI: 10.1016/j.neuron.2023.04.026] [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] [Received: 09/28/2021] [Revised: 10/25/2022] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Although the hippocampus is crucial for social memory, how social sensory information is combined with contextual information to form episodic social memories remains unknown. Here, we investigated the mechanisms for social sensory information processing using two-photon calcium imaging from hippocampal CA2 pyramidal neurons (PNs)-which are crucial for social memory-in awake head-fixed mice exposed to social and non-social odors. We found that CA2 PNs represent social odors of individual conspecifics and that these representations are refined during associative social odor-reward learning to enhance the discrimination of rewarded compared with unrewarded odors. Moreover, the structure of the CA2 PN population activity enables CA2 to generalize along categories of rewarded versus unrewarded and social versus non-social odor stimuli. Finally, we found that CA2 is important for learning social but not non-social odor-reward associations. These properties of CA2 odor representations provide a likely substrate for the encoding of episodic social memory.
Collapse
Affiliation(s)
- Sami I Hassan
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Shivani Bigler
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, The Kavli Institute for Brain Science, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
27
|
Wang Z, Yueh H, Chau M, Veenstra-VanderWeele J, O'Reilly KC. Circuits underlying social function and dysfunction. Autism Res 2023; 16:1268-1288. [PMID: 37458578 DOI: 10.1002/aur.2978] [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: 01/27/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023]
Abstract
Substantial advances have been made toward understanding the genetic and environmental risk factors for autism, a neurodevelopmental disorder with social impairment as a core feature. In combination with optogenetic and chemogenetic tools to manipulate neural circuits in vivo, it is now possible to use model systems to test how specific neural circuits underlie social function and dysfunction. Here, we review the literature that has identified circuits associated with social interest (sociability), social reward, social memory, dominance, and aggression, and we outline a preliminary roadmap of the neural circuits driving these social behaviors. We highlight the neural circuitry underlying each behavioral domain, as well as develop an interactive map of how these circuits overlap across domains. We find that some of the circuits underlying social behavior are general and are involved in the control of multiple behavioral aspects, whereas other circuits appear to be specialized for specific aspects of social behavior. Our overlapping circuit map therefore helps to delineate the circuits involved in the various domains of social behavior and to identify gaps in knowledge.
Collapse
Affiliation(s)
- Ziwen Wang
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hannah Yueh
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Mirabella Chau
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Kally C O'Reilly
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
28
|
Liu Y, Li A, Bair-Marshall C, Xu H, Jee HJ, Zhu E, Sun M, Zhang Q, Lefevre A, Chen ZS, Grinevich V, Froemke RC, Wang J. Oxytocin promotes prefrontal population activity via the PVN-PFC pathway to regulate pain. Neuron 2023; 111:1795-1811.e7. [PMID: 37023755 PMCID: PMC10272109 DOI: 10.1016/j.neuron.2023.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/02/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023]
Abstract
Neurons in the prefrontal cortex (PFC) can provide top-down regulation of sensory-affective experiences such as pain. Bottom-up modulation of sensory coding in the PFC, however, remains poorly understood. Here, we examined how oxytocin (OT) signaling from the hypothalamus regulates nociceptive coding in the PFC. In vivo time-lapse endoscopic calcium imaging in freely behaving rats showed that OT selectively enhanced population activity in the prelimbic PFC in response to nociceptive inputs. This population response resulted from the reduction of evoked GABAergic inhibition and manifested as elevated functional connectivity involving pain-responsive neurons. Direct inputs from OT-releasing neurons in the paraventricular nucleus (PVN) of the hypothalamus are crucial to maintaining this prefrontal nociceptive response. Activation of the prelimbic PFC by OT or direct optogenetic stimulation of oxytocinergic PVN projections reduced acute and chronic pain. These results suggest that oxytocinergic signaling in the PVN-PFC circuit constitutes a key mechanism to regulate cortical sensory processing.
Collapse
Affiliation(s)
- Yaling Liu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Anna Li
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA
| | - Chloe Bair-Marshall
- Skirball Institute for Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Helen Xu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA
| | - Hyun Jung Jee
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA
| | - Elaine Zhu
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA
| | - Mengqi Sun
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Qiaosheng Zhang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Zhe Sage Chen
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Robert C Froemke
- Skirball Institute for Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, USA
| | - Jing Wang
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of Medicine, New York, NY, USA; Interdisciplinary Pain Research Program, New York University Langone Health, New York, NY, USA; Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA; Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Oubraim S, Shen RY, Haj-Dahmane S. Oxytocin excites dorsal raphe serotonin neurons and bidirectionally gates their glutamate synapses. iScience 2023; 26:106707. [PMID: 37250336 PMCID: PMC10214716 DOI: 10.1016/j.isci.2023.106707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Oxytocin (OXT) modulates wide spectrum of social and emotional behaviors via modulation of numerous neurotransmitter systems, including serotonin (5-HT). However, how OXT controls the function of dorsal raphe nucleus (DRN) 5-HT neurons remains unknown. Here, we reveal that OXT excites and alters the firing pattern of 5-HT neurons via activation of postsynaptic OXT receptors (OXTRs). In addition, OXT induces cell-type-specific depression and potentiation of DRN glutamate synapses by two retrograde lipid messengers, 2-arachidonoylglycerol (2-AG) and arachidonic acid (AA), respectively. Neuronal mapping demonstrates that OXT preferentially potentiates glutamate synapses of 5-HT neurons projecting to medial prefrontal cortex (mPFC) and depresses glutamatergic inputs to 5-HT neurons projecting to lateral habenula (LHb) and central amygdala (CeA). Thus, by engaging distinct retrograde lipid messengers, OXT exerts a target-specific gating of glutamate synapses on the DRN. As such, our data uncovers the neuronal mechanisms by which OXT modulates the function of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
30
|
Kozlakidis Z, Shi P, Abarbanel G, Klein C, Sfera A. Recent Developments in Protein Lactylation in PTSD and CVD: Novel Strategies and Targets. BIOTECH 2023; 12:38. [PMID: 37218755 PMCID: PMC10204439 DOI: 10.3390/biotech12020038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/24/2023] Open
Abstract
In 1938, Corneille Heymans received the Nobel Prize in physiology for discovering that oxygen sensing in the aortic arch and carotid sinus was mediated by the nervous system. The genetics of this process remained unclear until 1991 when Gregg Semenza while studying erythropoietin, came upon hypoxia-inducible factor 1, for which he obtained the Nobel Prize in 2019. The same year, Yingming Zhao found protein lactylation, a posttranslational modification that can alter the function of hypoxia-inducible factor 1, the master regulator of cellular senescence, a pathology implicated in both post-traumatic stress disorder (PTSD) and cardiovascular disease (CVD). The genetic correlation between PTSD and CVD has been demonstrated by many studies, of which the most recent one utilizes large-scale genetics to estimate the risk factors for these conditions. This study focuses on the role of hypertension and dysfunctional interleukin 7 in PTSD and CVD, the former caused by stress-induced sympathetic arousal and elevated angiotensin II, while the latter links stress to premature endothelial cell senescence and early vascular aging. This review summarizes the recent developments and highlights several novel PTSD and CVD pharmacological targets. They include lactylation of histone and non-histone proteins, along with the related biomolecular actors such as hypoxia-inducible factor 1α, erythropoietin, acid-sensing ion channels, basigin, and Interleukin 7, as well as strategies to delay premature cellular senescence by telomere lengthening and resetting the epigenetic clock.
Collapse
Affiliation(s)
- Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization (IARC/WHO), 69372 Lyon, France
| | - Patricia Shi
- Department of Psychiatry, Loma Linda University, Loma Linda, CA 92350, USA
| | - Ganna Abarbanel
- Patton State Hospital, University of California, Riverside, CA 92521, USA
| | | | - Adonis Sfera
- Patton State Hospital, University of California, Riverside, CA 92521, USA
- Department of Psychiatry, University of California, Riverside, CA 92521, USA
| |
Collapse
|
31
|
Chamberland S, Nebet ER, Valero M, Hanani M, Egger R, Larsen SB, Eyring KW, Buzsáki G, Tsien RW. Brief synaptic inhibition persistently interrupts firing of fast-spiking interneurons. Neuron 2023; 111:1264-1281.e5. [PMID: 36787751 PMCID: PMC10121938 DOI: 10.1016/j.neuron.2023.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/06/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Neurons perform input-output operations that integrate synaptic inputs with intrinsic electrical properties; these operations are generally constrained by the brevity of synaptic events. Here, we report that sustained firing of CA1 hippocampal fast-spiking parvalbumin-expressing interneurons (PV-INs) can be persistently interrupted for several hundred milliseconds following brief GABAAR-mediated inhibition in vitro and in vivo. A single presynaptic neuron could interrupt PV-IN firing, occasionally with a single action potential (AP), and reliably with AP bursts. Experiments and computational modeling reveal that the persistent interruption of firing maintains neurons in a depolarized, quiescent state through a cell-autonomous mechanism. Interrupted PV-INs are strikingly responsive to Schaffer collateral inputs. The persistent interruption of firing provides a disinhibitory circuit mechanism favoring spike generation in CA1 pyramidal cells. Overall, our results demonstrate that neuronal silencing can far outlast brief synaptic inhibition owing to the well-tuned interplay between neurotransmitter release and postsynaptic membrane dynamics, a phenomenon impacting microcircuit function.
Collapse
Affiliation(s)
- Simon Chamberland
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA.
| | - Erica R Nebet
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Manuel Valero
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Monica Hanani
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Robert Egger
- NYU Neuroscience Institute and Department of Otolaryngology, NYU Langone Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - Samantha B Larsen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - Katherine W Eyring
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA
| | - György Buzsáki
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY, USA
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
32
|
Ahmed IA, Liu JJ, Gieniec KA, Bair-Marshall CJ, Adewakun AB, Hetzler BE, Arp CJ, Khatri L, Vanwalleghem GC, Seidenberg AT, Cowin P, Trauner D, Chao MV, Davis FM, Tsien RW, Froemke RC. Optopharmacological tools for precise spatiotemporal control of oxytocin signaling in the central nervous system and periphery. RESEARCH SQUARE 2023:rs.3.rs-2715993. [PMID: 37034806 PMCID: PMC10081362 DOI: 10.21203/rs.3.rs-2715993/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Oxytocin is a neuropeptide critical for maternal physiology and social behavior, and is thought to be dysregulated in several neuropsychiatric disorders. Despite the biological and neurocognitive importance of oxytocin signaling, methods are lacking to activate oxytocin receptors with high spatiotemporal precision in the brain and peripheral mammalian tissues. Here we developed and validated caged analogs of oxytocin which are functionally inert until cage release is triggered by ultraviolet light. We examined how focal versus global oxytocin application affected oxytocin-driven Ca2+ wave propagation in mouse mammary tissue. We also validated the application of caged oxytocin in the hippocampus and auditory cortex with electrophysiological recordings in vitro, and demonstrated that oxytocin uncaging can accelerate the onset of mouse maternal behavior in vivo. Together, these results demonstrate that optopharmacological control of caged peptides is a robust tool with spatiotemporal precision for modulating neuropeptide signaling throughout the brain and body.
Collapse
Affiliation(s)
- Ismail A. Ahmed
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jing-Jing Liu
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Krystyna A. Gieniec
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Chloe J. Bair-Marshall
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ayomiposi B. Adewakun
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Belinda E. Hetzler
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Christopher J. Arp
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Latika Khatri
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Gilles C. Vanwalleghem
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine. Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Alec T. Seidenberg
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Pamela Cowin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Dirk Trauner
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses V. Chao
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - Felicity M. Davis
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Richard W. Tsien
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Robert C. Froemke
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10003, USA
| |
Collapse
|
33
|
Oliva A, Fernandez-Ruiz A, Karaba LA. CA2 orchestrates hippocampal network dynamics. Hippocampus 2023; 33:241-251. [PMID: 36575880 PMCID: PMC9974898 DOI: 10.1002/hipo.23495] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/25/2022] [Accepted: 12/11/2022] [Indexed: 12/29/2022]
Abstract
The hippocampus is composed of various subregions: CA1, CA2, CA3, and the dentate gyrus (DG). Despite the abundant hippocampal research literature, until recently, CA2 received little attention. The development of new genetic and physiological tools allowed recent studies characterizing the unique properties and functional roles of this hippocampal subregion. Despite its small size, the cellular content of CA2 is heterogeneous at the molecular and physiological levels. CA2 has been heavily implicated in social behaviors, including social memory. More generally, the mechanisms by which the hippocampus is involved in memory include the reactivation of neuronal ensembles following experience. This process is coordinated by synchronous network events known as sharp-wave ripples (SWRs). Recent evidence suggests that CA2 plays an important role in the generation of SWRs. The unique connectivity and physiological properties of CA2 pyramidal cells make this region a computational hub at the core of hippocampal information processing. Here, we review recent findings that support the role of CA2 in coordinating hippocampal network dynamics from a systems neuroscience perspective.
Collapse
Affiliation(s)
- Azahara Oliva
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | | | - Lindsay A Karaba
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| |
Collapse
|
34
|
Radzicki D, Chong S, Dudek SM. Morphological and molecular markers of mouse area CA2 along the proximodistal and dorsoventral hippocampal axes. Hippocampus 2023; 33:133-149. [PMID: 36762588 PMCID: PMC10443601 DOI: 10.1002/hipo.23509] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/11/2023]
Abstract
Hippocampal area CA2 is a molecularly and functionally distinct region of the hippocampus that has classically been defined as the area with large pyramidal neurons lacking input from the dentate gyrus and the thorny excrescences (TEs) characteristic of CA3 neurons. A modern definition of CA2, however, makes use of the expression of several molecular markers that distinguish it from neighboring CA3 and CA1. Using immunohistochemistry, we sought to characterize the staining patterns of commonly used CA2 markers along the dorsal-ventral hippocampal axis and determine how these markers align along the proximodistal axis. We used a region of CA2 that stained for both Regulator of G-protein Signaling 14 (RGS14) and Purkinje Cell Protein 4 (PCP4; "double-labeled zone" [DLZ]) as a reference. Here, we report that certain commonly used CA2 molecular markers may be better suited for drawing distinct boundaries between CA2/3 and CA2/1. For example, RGS14+ and STEP+ neurons showed minimal to no extension into area CA1 while areas stained with VGluT2 and Wisteria Floribunda agglutinin were consistently smaller than the DLZ/CA2 borders by ~100 μ on the CA1 or CA3 sides respectively. In addition, these patterns are dependent on position along the dorsal-ventral hippocampal axis such that PCP4 labeling often extended beyond the distal border of the DLZ into CA1. Finally, we found that, consistent with previous findings, mossy fibers innervate a subset of RGS14 positive neurons (~65%-70%) and that mossy fiber bouton number and relative size in CA2 are less than that of boutons in CA3. Unexpectedly, we did find evidence of some complex spines on apical dendrites in CA2, though much fewer in number than in CA3. Our results indicate that certain molecular markers may be better suited than others when defining the proximal and distal borders of area CA2 and that the presence or absence of complex spines alone may not be suitable as a distinguishing feature differentiating CA3 from CA2 neurons.
Collapse
Affiliation(s)
- Daniel Radzicki
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| | - Sarah Chong
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health SciencesNational Institute of HealthResearch Triangle ParkNorth CarolinaUSA
| |
Collapse
|
35
|
Shinohara Y, Kohara K. Projections of hippocampal CA2 pyramidal neurons: Distinct innervation patterns of CA2 compared to CA3 in rodents. Hippocampus 2023; 33:691-699. [PMID: 36855258 DOI: 10.1002/hipo.23519] [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: 09/01/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
The hippocampus is a center for spatial and episodic memory formation in rodents. Understanding the composition of subregions and circuitry maps of the hippocampus is essential for elucidating the mechanism of memory formation and recall. For decades, the trisynaptic circuit (entorhinal cortex layer II-dentate gyrus - CA3-CA1) has been considered the neural network substrate responsible for learning and memory. Recently, CA2 has emerged as an important area in the hippocampal circuitry, with distinct functions from those of CA3. In this article, we review the historical definition of the hippocampal area CA2 and the differential projection patterns between CA2 and CA3 pyramidal neurons. We provide a concise and comprehensive map of CA2 outputs by comparing (1) ipsi versus contra projections, (2) septal versus temporal projections, and (3) lamellar structures of CA2 and CA3 pyramidal neurons.
Collapse
Affiliation(s)
- Yoshiaki Shinohara
- Department of Anatomy and Cell Biology, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keigo Kohara
- KMU Biobank Center, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
36
|
Baudon A, Clauss Creusot E, Charlet A. [Emergent role of astrocytes in oxytocin-mediated modulatory control of neuronal circuits and brain functions]. Biol Aujourdhui 2023; 216:155-165. [PMID: 36744981 DOI: 10.1051/jbio/2022022] [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: 09/22/2022] [Indexed: 02/07/2023]
Abstract
The neuropeptide oxytocin has been in the focus of scientists for decades due to its profound and pleiotropic effects on physiology, activity of neuronal circuits and behaviors. Until recently, it was believed that oxytocinergic action exclusively occurs through direct activation of neuronal oxytocin receptors. However, several studies demonstrated the existence and functional relevance of astroglial oxytocin receptors in various brain regions in the mouse and rat brain. Astrocytic signaling and activity are critical for many important physiological processes including metabolism, neurotransmitter clearance from the synaptic cleft and integrated brain functions. While it can be speculated that oxytocinergic action on astrocytes predominantly facilitates neuromodulation via the release of gliotransmitters, the precise role of astrocytic oxytocin receptors remains elusive. In this review, we discuss the latest studies on the interaction between the oxytocinergic system and astrocytes, and give details of underlying intracellular cascades.
Collapse
Affiliation(s)
- Angel Baudon
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Etienne Clauss Creusot
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique et Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, 8 allée du Général Rouvillois, 67000 Strasbourg, France
| |
Collapse
|
37
|
Talpo F, Spaiardi P, Castagno AN, Maniezzi C, Raffin F, Terribile G, Sancini G, Pisani A, Biella GR. Neuromodulatory functions exerted by oxytocin on different populations of hippocampal neurons in rodents. Front Cell Neurosci 2023; 17:1082010. [PMID: 36816855 PMCID: PMC9932910 DOI: 10.3389/fncel.2023.1082010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Oxytocin (OT) is a neuropeptide widely known for its peripheral hormonal effects (i.e., parturition and lactation) and central neuromodulatory functions, related especially to social behavior and social, spatial, and episodic memory. The hippocampus is a key structure for these functions, it is innervated by oxytocinergic fibers, and contains OT receptors (OTRs). The hippocampal OTR distribution is not homogeneous among its subregions and types of neuronal cells, reflecting the specificity of oxytocin's modulatory action. In this review, we describe the most recent discoveries in OT/OTR signaling in the hippocampus, focusing primarily on the electrophysiological oxytocinergic modulation of the OTR-expressing hippocampal neurons. We then look at the effect this modulation has on the balance of excitation/inhibition and synaptic plasticity in each hippocampal subregion. Additionally, we review OTR downstream signaling, which underlies the OT effects observed in different types of hippocampal neuron. Overall, this review comprehensively summarizes the advancements in unraveling the neuromodulatory functions exerted by OT on specific hippocampal networks.
Collapse
Affiliation(s)
- Francesca Talpo
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Paolo Spaiardi
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy,Istituto Nazionale di Fisica Nucleare, Sezione di Pavia, Pavia, Italy
| | - Antonio Nicolas Castagno
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Claudia Maniezzi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesca Raffin
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Giulia Terribile
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Giulio Sancini
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy,Nanomedicine Center, Neuroscience Center, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy,Neurological Institute Foundation Casimiro Mondino (IRCCS), Pavia, Italy
| | - Gerardo Rosario Biella
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy,Istituto Nazionale di Fisica Nucleare, Sezione di Pavia, Pavia, Italy,*Correspondence: Gerardo Rosario Biella,
| |
Collapse
|
38
|
Cymerblit-Sabba A, Walsh C, Duan KZ, Song J, Holmes O, Young WS. Simultaneous Knockouts of the Oxytocin and Vasopressin 1b Receptors in Hippocampal CA2 Impair Social Memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526271. [PMID: 36789441 PMCID: PMC9928026 DOI: 10.1101/2023.01.30.526271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Oxytocin (Oxt) and vasopressin (Avp) are two neuropeptides with many central actions related to social cognition. The oxytocin (Oxtr) and vasopressin 1b (Avpr1b) receptors are co-expressed in the pyramidal neurons of the hippocampal subfield CA2 and are known to play a critical role in social memory formation. How the neuropeptides perform this function in this region is not fully understood. Here, we report the behavioral effects of a life-long conditional removal (knockout, KO) of either the Oxtr alone or both Avpr1b and Oxtr from the pyramidal neurons of CA2 as well as the resultant changes in synaptic transmission within the different fields of the hippocampus. Surprisingly, the removal of both receptors results in mice that are unable to habituate to a familiar female presented for short duration over short intervals but are able to recognize and discriminate females when presented for a longer duration over a longer interval. Importantly, these double KO mice were unable to discriminate between a male littermate and a novel male. Synaptic transmission between CA3 and CA2 is enhanced in these mice, suggesting a compensatory mechanism is activated to make up for the loss of the receptors. Overall, our results demonstrate that co-expression of the receptors in CA2 is necessary to allow intact social memory processing.
Collapse
Affiliation(s)
- Adi Cymerblit-Sabba
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| | - Caroline Walsh
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| | - Kai-Zheng Duan
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| | - June Song
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| | - Oliver Holmes
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| | - W Scott Young
- Section on Neural Gene Expression, National Institute of Mental Health (NIMH), National Institute of Health, Bethesda, MD, United States
| |
Collapse
|
39
|
Černotová D, Hrůzová K, Levčík D, Svoboda J, Stuchlík A. Linking Social Cognition, Parvalbumin Interneurons, and Oxytocin in Alzheimer's Disease: An Update. J Alzheimers Dis 2023; 96:861-875. [PMID: 37980658 PMCID: PMC10741376 DOI: 10.3233/jad-230333] [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] [Accepted: 09/11/2023] [Indexed: 11/21/2023]
Abstract
Finding a cure for Alzheimer's disease (AD) has been notoriously challenging for many decades. Therefore, the current focus is mainly on prevention, timely intervention, and slowing the progression in the earliest stages. A better understanding of underlying mechanisms at the beginning of the disease could aid in early diagnosis and intervention, including alleviating symptoms or slowing down the disease progression. Changes in social cognition and progressive parvalbumin (PV) interneuron dysfunction are among the earliest observable effects of AD. Various AD rodent models mimic these early alterations, but only a narrow field of study has considered their mutual relationship. In this review, we discuss current knowledge about PV interneuron dysfunction in AD and emphasize their importance in social cognition and memory. Next, we propose oxytocin (OT) as a potent modulator of PV interneurons and as a promising treatment for managing some of the early symptoms. We further discuss the supporting evidence on its beneficial effects on AD-related pathology. Clinical trials have employed the use of OT in various neuropsychiatric diseases with promising results, but little is known about its prospective impacts on AD. On the other hand, the modulatory effects of OT in specific structures and local circuits need to be clarified in future studies. This review highlights the connection between PV interneurons and social cognition impairment in the early stages of AD and considers OT as a promising therapeutic agent for addressing these early deficits.
Collapse
Affiliation(s)
- Daniela Černotová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolína Hrůzová
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Levčík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Stuchlík
- Laboratory of Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
40
|
Walsh JJ, Christoffel DJ, Malenka RC. Neural circuits regulating prosocial behaviors. Neuropsychopharmacology 2023; 48:79-89. [PMID: 35701550 PMCID: PMC9700801 DOI: 10.1038/s41386-022-01348-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 11/09/2022]
Abstract
Positive, prosocial interactions are essential for survival, development, and well-being. These intricate and complex behaviors are mediated by an amalgamation of neural circuit mechanisms working in concert. Impairments in prosocial behaviors, which occur in a large number of neuropsychiatric disorders, result from disruption of the coordinated activity of these neural circuits. In this review, we focus our discussion on recent findings that utilize modern approaches in rodents to map, monitor, and manipulate neural circuits implicated in a variety of prosocial behaviors. We highlight how modulation by oxytocin, serotonin, and dopamine of excitatory and inhibitory synaptic transmission in specific brain regions is critical for regulation of adaptive prosocial interactions. We then describe how recent findings have helped elucidate pathophysiological mechanisms underlying the social deficits that accompany neuropsychiatric disorders. We conclude by discussing approaches for the development of more efficacious and targeted therapeutic interventions to ameliorate aberrant prosocial behaviors.
Collapse
Affiliation(s)
- Jessica J Walsh
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 27514, USA.
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA.
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27514, USA.
| | - Daniel J Christoffel
- Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27514, USA
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305-5453, USA.
| |
Collapse
|
41
|
Piskorowski RA, Chevaleyre V. Hippocampal area CA2: interneuron disfunction during pathological states. Front Neural Circuits 2023; 17:1181032. [PMID: 37180763 PMCID: PMC10174260 DOI: 10.3389/fncir.2023.1181032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Hippocampal area CA2 plays a critical role in social recognition memory and has unique cellular and molecular properties that distinguish it from areas CA1 and CA3. In addition to having a particularly high density of interneurons, the inhibitory transmission in this region displays two distinct forms of long-term synaptic plasticity. Early studies on human hippocampal tissue have reported unique alteration in area CA2 with several pathologies and psychiatric disorders. In this review, we present recent studies revealing changes in inhibitory transmission and plasticity of area CA2 in mouse models of multiple sclerosis, autism spectrum disorder, Alzheimer's disease, schizophrenia and the 22q11.2 deletion syndrome and propose how these changes could underly deficits in social cognition observed during these pathologies.
Collapse
Affiliation(s)
- Rebecca A. Piskorowski
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
- *Correspondence: Rebecca A. Piskorowski,
| | - Vivien Chevaleyre
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
| |
Collapse
|
42
|
Althammer F, Wimmer MC, Krabichler Q, Küppers S, Schimmer J, Fröhlich H, Dötsch L, Gruber T, Wunsch S, Schubert T, Kirchner MK, Stern JE, Charlet A, Grinevich V, Schaaf CP. Analysis of the hypothalamic oxytocin system and oxytocin receptor-expressing astrocytes in a mouse model of Prader-Willi syndrome. J Neuroendocrinol 2022; 34:e13217. [PMID: 36458331 DOI: 10.1111/jne.13217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Prader-Willi syndrome (PWS) is a neurodevelopmental disorder characterized by hyperphagia, obesity, developmental delay and intellectual disability. Studies suggest dysfunctional signaling of the neuropeptide oxytocin as one of the key mechanisms in PWS, and administration of oxytocin via intranasal or systemic routes yielded promising results in both humans and mouse models. However, a detailed assessment of the oxytocin system in mouse models of PWS such as the Magel2-deficient Magel2tm1.Stw mouse, is lacking. In the present study, we performed an automated counting of oxytocin cells in the entire paraventricular nucleus of the hypothalamus of Magel2tm1.Stw and wild-type control mice and found a significant reduction in the caudal part, which represents the parvocellular subdivision. In addition, based on the recent discovery that some astrocytes express the oxytocin receptor (OTR), we performed detailed analysis of astrocyte numbers and morphology in various brain regions, and assessed expression levels of the astrocyte marker glial fibrillary acidic protein, which was significantly decreased in the hypothalamus, but not other brain regions in Magel2tm1.Stw mice. Finally, we analyzed the number of OTR-expressing astrocytes in various brain regions and found a significant reduction in the nucleus accumbens of Magel2tm1.Stw mice, as well as a sex-specific difference in the lateral septum. This study suggests a role for caudal paraventricular nucleus oxytocin neurons as well as OTR-expressing astrocytes in a mouse model of PWS, provides novel information about sex-specific expression of astrocytic OTRs, and presents several new brain regions containing OTR-expressing astrocytes in the mouse brain.
Collapse
Affiliation(s)
| | | | - Quirin Krabichler
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Stephanie Küppers
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Jonas Schimmer
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Henning Fröhlich
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Dötsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Tim Gruber
- Van Andel Institute, Grand Rapids, MI, USA
| | - Selina Wunsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Tim Schubert
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Matthew K Kirchner
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Javier E Stern
- Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, USA
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique and University of Strasbourg, Institute of Cellular and Integrative Neuroscience, Strasbourg, France
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
43
|
Triana-Del Rio R, Ranade S, Guardado J, LeDoux J, Klann E, Shrestha P. The modulation of emotional and social behaviors by oxytocin signaling in limbic network. Front Mol Neurosci 2022; 15:1002846. [PMID: 36466805 PMCID: PMC9714608 DOI: 10.3389/fnmol.2022.1002846] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2024] Open
Abstract
Neuropeptides can exert volume modulation in neuronal networks, which account for a well-calibrated and fine-tuned regulation that depends on the sensory and behavioral contexts. For example, oxytocin (OT) and oxytocin receptor (OTR) trigger a signaling pattern encompassing intracellular cascades, synaptic plasticity, gene expression, and network regulation, that together function to increase the signal-to-noise ratio for sensory-dependent stress/threat and social responses. Activation of OTRs in emotional circuits within the limbic forebrain is necessary to acquire stress/threat responses. When emotional memories are retrieved, OTR-expressing cells act as gatekeepers of the threat response choice/discrimination. OT signaling has also been implicated in modulating social-exposure elicited responses in the neural circuits within the limbic forebrain. In this review, we describe the cellular and molecular mechanisms that underlie the neuromodulation by OT, and how OT signaling in specific neural circuits and cell populations mediate stress/threat and social behaviors. OT and downstream signaling cascades are heavily implicated in neuropsychiatric disorders characterized by emotional and social dysregulation. Thus, a mechanistic understanding of downstream cellular effects of OT in relevant cell types and neural circuits can help design effective intervention techniques for a variety of neuropsychiatric disorders.
Collapse
Affiliation(s)
| | - Sayali Ranade
- Department of Neurobiology and Behavior, School of Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Jahel Guardado
- Center for Neural Science, New York University, New York, NY, United States
| | - Joseph LeDoux
- Center for Neural Science, New York University, New York, NY, United States
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, United States
| | - Prerana Shrestha
- Department of Neurobiology and Behavior, School of Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
44
|
Inoue K, Ford CL, Horie K, Young LJ. Oxytocin receptors are widely distributed in the prairie vole (Microtus ochrogaster) brain: Relation to social behavior, genetic polymorphisms, and the dopamine system. J Comp Neurol 2022; 530:2881-2900. [PMID: 35763609 PMCID: PMC9474670 DOI: 10.1002/cne.25382] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022]
Abstract
Oxytocin regulates social behavior via direct modulation of neurons, regulation of neural network activity, and interaction with other neurotransmitter systems. The behavioral effects of oxytocin signaling are determined by the species-specific distribution of brain oxytocin receptors. The socially monogamous prairie vole has been a useful model organism for elucidating the role of oxytocin in social behaviors, including pair bonding, response to social loss, and consoling. However, there has been no comprehensive mapping of oxytocin receptor-expressing cells throughout the prairie vole brain. Here, we employed a highly sensitive in situ hybridization, RNAscope, to construct an exhaustive, brain-wide map of oxytocin receptor mRNA-expressing cells. We found that oxytocin receptor mRNA expression was widespread and diffused throughout the brain, with specific areas displaying a particularly robust expression. Comparing receptor binding with mRNA revealed that regions of the hippocampus and substantia nigra contained oxytocin receptor protein but lacked mRNA, indicating that oxytocin receptors can be transported to distal neuronal processes, consistent with presynaptic oxytocin receptor functions. In the nucleus accumbens, a region involved in oxytocin-dependent social bonding, oxytocin receptor mRNA expression was detected in both the D1 and D2 dopamine receptor-expressing subtypes of cells. Furthermore, natural genetic polymorphisms robustly influenced oxytocin receptor expression in both D1 and D2 receptor cell types in the nucleus accumbens. Collectively, our findings further elucidate the extent to which oxytocin signaling is capable of influencing brain-wide neural activity, responses to social stimuli, and social behavior. KEY POINTS: Oxytocin receptor mRNA is diffusely expressed throughout the brain, with strong expression concentrated in certain areas involved in social behavior. Oxytocin receptor mRNA expression and protein localization are misaligned in some areas, indicating that the receptor protein may be transported to distal processes. In the nucleus accumbens, oxytocin receptors are expressed on cells expressing both D1 and D2 dopamine receptor subtypes, and the majority of variation in oxytocin receptor expression between animals is attributable to polymorphisms in the oxytocin receptor gene.
Collapse
Affiliation(s)
- Kiyoshi Inoue
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta GA 30329, USA
| | - Charles L. Ford
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta GA 30329, USA
| | - Kengo Horie
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta GA 30329, USA
| | - Larry J. Young
- Center for Translational Social Neuroscience, Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta GA 30322, USA
| |
Collapse
|
45
|
Pekarek BT, Kochukov M, Lozzi B, Wu T, Hunt PJ, Tepe B, Hanson Moss E, Tantry EK, Swanson JL, Dooling SW, Patel M, Belfort BDW, Romero JM, Bao S, Hill MC, Arenkiel BR. Oxytocin signaling is necessary for synaptic maturation of adult-born neurons. Genes Dev 2022; 36:1100-1118. [PMID: 36617877 PMCID: PMC9851403 DOI: 10.1101/gad.349930.122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022]
Abstract
Neural circuit plasticity and sensory response dynamics depend on forming new synaptic connections. Despite recent advances toward understanding the consequences of circuit plasticity, the mechanisms driving circuit plasticity are unknown. Adult-born neurons within the olfactory bulb have proven to be a powerful model for studying circuit plasticity, providing a broad and accessible avenue into neuron development, migration, and circuit integration. We and others have shown that efficient adult-born neuron circuit integration hinges on presynaptic activity in the form of diverse signaling peptides. Here, we demonstrate a novel oxytocin-dependent mechanism of adult-born neuron synaptic maturation and circuit integration. We reveal spatial and temporal enrichment of oxytocin receptor expression within adult-born neurons in the murine olfactory bulb, with oxytocin receptor expression peaking during activity-dependent integration. Using viral labeling, confocal microscopy, and cell type-specific RNA-seq, we demonstrate that oxytocin receptor signaling promotes synaptic maturation of newly integrating adult-born neurons by regulating their morphological development and expression of mature synaptic AMPARs and other structural proteins.
Collapse
Affiliation(s)
- Brandon T Pekarek
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Mikhail Kochukov
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Department of Anesthesiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Brittney Lozzi
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Timothy Wu
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Patrick J Hunt
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Burak Tepe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Elizabeth Hanson Moss
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Evelyne K Tantry
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Jessica L Swanson
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Sean W Dooling
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Mayuri Patel
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Benjamin D W Belfort
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Juan M Romero
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Suyang Bao
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Matthew C Hill
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
46
|
Manjila SB, Betty R, Kim Y. Missing pieces in decoding the brain oxytocin puzzle: Functional insights from mouse brain wiring diagrams. Front Neurosci 2022; 16:1044736. [PMID: 36389241 PMCID: PMC9643707 DOI: 10.3389/fnins.2022.1044736] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/06/2022] [Indexed: 10/24/2023] Open
Abstract
The hypothalamic neuropeptide, oxytocin (Oxt), has been the focus of research for decades due to its effects on body physiology, neural circuits, and various behaviors. Oxt elicits a multitude of actions mainly through its receptor, the Oxt receptor (OxtR). Despite past research to understand the central projections of Oxt neurons and OxtR- coupled signaling pathways in different brain areas, it remains unclear how this nonapeptide exhibits such pleiotropic effects while integrating external and internal information. Most reviews in the field either focus on neuroanatomy of the Oxt-OxtR system, or on the functional effects of Oxt in specific brain areas. Here, we provide a review by integrating brain wide connectivity of Oxt neurons and their downstream circuits with OxtR expression in mice. We categorize Oxt connected brain regions into three functional modules that regulate the internal state, somatic visceral, and cognitive response. Each module contains three neural circuits that process distinct behavioral effects. Broad innervations on functional circuits (e.g., basal ganglia for motor behavior) enable Oxt signaling to exert coordinated modulation in functionally inter-connected circuits. Moreover, Oxt acts as a neuromodulator of neuromodulations to broadly control the overall state of the brain. Lastly, we discuss the mismatch between Oxt projections and OxtR expression across various regions of the mouse brain. In summary, this review brings forth functional circuit-based analysis of Oxt connectivity across the whole brain in light of Oxt release and OxtR expression and provides a perspective guide to future studies.
Collapse
Affiliation(s)
| | | | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
47
|
Oxytocin-Modulated Ion Channel Ensemble Controls Depolarization, Integration and Burst Firing in CA2 Pyramidal Neurons. J Neurosci 2022; 42:7707-7720. [PMID: 36414006 PMCID: PMC9581561 DOI: 10.1523/jneurosci.0921-22.2022] [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: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/26/2022] [Indexed: 12/14/2022] Open
Abstract
Oxytocin (OXT) and OXT receptor (OXTR)-mediated signaling control excitability, firing patterns, and plasticity of hippocampal CA2 pyramidal neurons, which are pivotal in generation of brain oscillations and social memory. Nonetheless, the ionic mechanisms underlying OXTR-induced effects in CA2 neurons are not fully understood. Using slice physiology in a reporter mouse line and interleaved current-clamp and voltage-clamp experiments, we systematically identified the ion channels modulated by OXT signaling in CA2 pyramidal cells (PYRs) in mice of both sexes and explored how changes in channel conductance support altered electrical activity. Activation of OXTRs inhibits an outward potassium current mediated by inward rectifier potassium channels (I Kir) and thus favoring membrane depolarization. Concomitantly, OXT signaling also diminishes inward current mediated by hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels (I h), providing a hyperpolarizing drive. The combined reduction in both I Kir and I h synergistically elevate the membrane resistance and favor dendritic integration while the membrane potential is restrained from quickly depolarizing from rest. As a result, the responsiveness of CA2 PYRs to synaptic inputs is highly sharpened during OXTR activation. Unexpectedly, OXTR signaling also strongly enhances a tetrodotoxin-resistant (TTX-R), voltage-gated sodium current that helps drive the membrane potential to spike threshold and thus promote rhythmic firing. This novel array of OXTR-stimulated ionic mechanisms operates in close coordination and underpins OXT-induced burst firing, a key step in CA2 PYRs' contribution to hippocampal information processing and broader influence on brain circuitry. Our study deepens our understanding of underpinnings of OXT-promoted social memory and general neuropeptidergic control of cognitive states.SIGNIFICANCE STATEMENT Oxytocin (OXT) plays key roles in reproduction, parenting and social and emotional behavior, and deficiency in OXT receptor (OXTR) signaling may contribute to neuropsychiatric disorders. We identified a novel array of OXTR-modulated ion channels that operate in close coordination to retune hippocampal CA2 pyramidal neurons, enhancing responsiveness to synaptic inputs and sculpting output. OXTR signaling inhibits both potassium conductance (I Kir) and mixed cation conductance (I h), engaging opposing influences on membrane potential, stabilizing it while synergistically elevating membrane resistance and electrotonic spread. OXT signaling also facilitates a tetrodotoxin-resistant (TTX-R) Na+ current, not previously described in hippocampus (HP), engaged on further depolarization. This TTX-R current lowers the spike threshold and supports rhythmic depolarization and burst firing, a potent driver of downstream circuitry.
Collapse
|
48
|
Rigney N, de Vries GJ, Petrulis A, Young LJ. Oxytocin, Vasopressin, and Social Behavior: From Neural Circuits to Clinical Opportunities. Endocrinology 2022; 163:bqac111. [PMID: 35863332 PMCID: PMC9337272 DOI: 10.1210/endocr/bqac111] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Indexed: 11/19/2022]
Abstract
Oxytocin and vasopressin are peptide hormones secreted from the pituitary that are well known for their peripheral endocrine effects on childbirth/nursing and blood pressure/urine concentration, respectively. However, both peptides are also released in the brain, where they modulate several aspects of social behaviors. Oxytocin promotes maternal nurturing and bonding, enhances social reward, and increases the salience of social stimuli. Vasopressin modulates social communication, social investigation, territorial behavior, and aggression, predominantly in males. Both peptides facilitate social memory and pair bonding behaviors in monogamous species. Here we review the latest research delineating the neural circuitry of the brain oxytocin and vasopressin systems and summarize recent investigations into the circuit-based mechanisms modulating social behaviors. We highlight research using modern molecular genetic technologies to map, monitor activity of, or manipulate neuropeptide circuits. Species diversity in oxytocin and vasopressin effects on social behaviors are also discussed. We conclude with a discussion of the translational implications of oxytocin and vasopressin for improving social functioning in disorders with social impairments, such as autism spectrum disorder.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Geert J de Vries
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA
| | - Aras Petrulis
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Emory University, Atlanta, Georgia 30329, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
49
|
Oxytocin-based therapies for treatment of Prader-Willi and Schaaf-Yang syndromes: evidence, disappointments, and future research strategies. Transl Psychiatry 2022; 12:318. [PMID: 35941105 PMCID: PMC9360032 DOI: 10.1038/s41398-022-02054-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 11/09/2022] Open
Abstract
The prosocial neuropeptide oxytocin is being developed as a potential treatment for various neuropsychiatric disorders including autism spectrum disorder (ASD). Early studies using intranasal oxytocin in patients with ASD yielded encouraging results and for some time, scientists and affected families placed high hopes on the use of intranasal oxytocin for behavioral therapy in ASD. However, a recent Phase III trial obtained negative results using intranasal oxytocin for the treatment of behavioral symptoms in children with ASD. Given the frequently observed autism-like behavioral phenotypes in Prader-Willi and Schaaf-Yang syndromes, it is unclear whether oxytocin treatment represents a viable option to treat behavioral symptoms in these diseases. Here we review the latest findings on intranasal OT treatment, Prader-Willi and Schaaf-Yang syndromes, and propose novel research strategies for tailored oxytocin-based therapies for affected individuals. Finally, we propose the critical period theory, which could explain why oxytocin-based treatment seems to be most efficient in infants, but not adolescents.
Collapse
|
50
|
Tsai TC, Fang YS, Hung YC, Hung LC, Hsu KS. A dorsal CA2 to ventral CA1 circuit contributes to oxytocinergic modulation of long-term social recognition memory. J Biomed Sci 2022; 29:50. [PMID: 35811321 PMCID: PMC9272559 DOI: 10.1186/s12929-022-00834-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Social recognition memory (SRM) is the ability to distinguish familiar from novel conspecifics and is crucial for survival and reproductive success across social species. We previously reported that oxytocin (OXT) receptor (OXTR) signaling in the CA2/CA3a of dorsal hippocampus is essential to promote the persistence of long-term SRM, yet how the endogenous OXT system influences CA2 outputs to regulate long-term SRM formation remains unclear. METHODS To achieve a selective deletion of CA2 OXTRs, we crossed Amigo2-Cre mice with Oxtr-floxed mice to generate CA2-specific Oxtr conditional knockout (Oxtr-/-) mice. A three-chamber paradigm test was used for studying SRM in mice. Chemogenetic and optogenetic targeting strategies were employed to manipulate neuronal activity. RESULTS We show that selective ablation of Oxtr in the CA2 suffices to impair the persistence of long-term SRM but has no effect on sociability and social novelty preference in the three-chamber paradigm test. We find that cell-type specific activation of OXT neurons within the hypothalamic paraventricular nucleus enhances long-term SRM and this enhancement is blocked by local application of OXTR antagonist L-368,899 into dorsal hippocampal CA2 (dCA2) region. In addition, chemogenetic neuronal silencing in dCA2 demonstrated that neuronal activity is essential for forming long-term SRM. Moreover, chemogenetic terminal-specific inactivation reveals a crucial role for dCA2 outputs to ventral CA1 (vCA1), but not dorsal lateral septum, in long-term SRM. Finally, targeted activation of the dCA2-to-vCA1 circuit effectively ameliorates long-term SRM deficit observed in Oxtr-/- mice. CONCLUSIONS These findings highlight the importance of hippocampal CA2 OXTR signaling in governing the persistence of long-term SRM and identify a hippocampal circuit linking dCA2 to vCA1 necessary for controlling long-term SRM formation.
Collapse
Affiliation(s)
- Tsung-Chih Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Syuan Fang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan
| | - Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Ling-Chien Hung
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|