1
|
Qi S, Ritchie JL, Soto DA, Pruitt AY, Reeves DA, Artimenia LM, Fuchs RA. Sex-dependent role of the dorsolateral septum in shaping contextual cocaine memory strength. Neuropharmacology 2025; 273:110459. [PMID: 40204057 DOI: 10.1016/j.neuropharm.2025.110459] [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: 02/14/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Established memories can be destabilized, updated, and reconsolidated into long-term memory stores. Memory updating and reconsolidation can alter the strength of maladaptive contextual drug memories and consequently context-induced drug craving and relapse. The dorsolateral septum (dlS) is a GABAergic nucleus that receives dense direct input from the cornu ammonis 3 regions of the dorsal hippocampus, a brain region that is critical for the maintenance of contextual cocaine memories. Accordingly, we tested the hypothesis that neuronal activity in the dlS regulates the strength of cocaine-predictive contextual memories prior to reconsolidation. Male and female Sprague-Dawley rats received cocaine self-administration training followed by extinction training in two different environmental contexts. After the last extinction training session, the rats were placed back into the cocaine-predictive context to retrieve and destabilize their cocaine-related contextual memories. Immediately or 6 h after memory retrieval, the rats received intra-dlS vehicle or baclofen/muscimol (B/M; GABAB/A agonists) infusions to inhibit neuronal activity during or after memory updating/reconsolidation, respectively. Resulting changes in cocaine and extinction memory strength were assessed based on the magnitude of unreinforced lever responding in the two contexts. Intra-dlS B/M infusion immediately after memory retrieval increased subsequent context-induced cocaine seeking behaviors in male rats, but not in female rats, whereas delayed B/M treatment had no effects in male rats. Together these findings suggest that the dlS is selectively engaged during memory updating/reconsolidation to reduce the strength of cocaine memories in males, possibly contributing to sex differences in the progression of cocaine use disorder.
Collapse
Affiliation(s)
- S Qi
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - J L Ritchie
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - D A Soto
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - A Y Pruitt
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - D A Reeves
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - L M Artimenia
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - R A Fuchs
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA; Washington State University Alcohol and Drug Abuse Research Program, Pullman, WA, USA.
| |
Collapse
|
2
|
Simon RC, Fleming WT, Briones BA, Trzeciak M, Senthilkumar P, Ishii KK, Hjort MM, Martin MM, Hashikawa K, Sanders AD, Golden SA, Stuber GD. Opioid-driven disruption of the septum reveals a role for neurotensin-expressing neurons in withdrawal. Neuron 2025:S0896-6273(25)00307-1. [PMID: 40378834 DOI: 10.1016/j.neuron.2025.04.024] [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: 03/14/2024] [Revised: 03/14/2025] [Accepted: 04/25/2025] [Indexed: 05/19/2025]
Abstract
Opioid withdrawal is an intensively aversive experience and often drives relapse. The lateral septum (LS) is a forebrain structure that is important in aversion processing and has been linked to substance use disorders, but which LS cell types contribute to the maladaptive state of withdrawal is unknown. We used single-nucleus RNA sequencing to interrogate cell-type-specific gene expression changes induced by chronic morphine exposure and discovered that morphine globally disrupts LS cell types, but neurotensin-expressing neurons (LS-Nts) are selectively activated by naloxone. Using two-photon calcium imaging and ex vivo electrophysiology, we next demonstrate that LS-Nts neurons receive elevated glutamatergic drive in morphine-dependent mice and remain hyperactivated during withdrawal. Finally, we show that manipulating LS-Nts neurons during opioid withdrawal regulates pain coping and sociability. Together, these results suggest that LS-Nts neurons are a key neural substrate involved in opioid withdrawal and establish the LS as a crucial regulator of adaptive behaviors.
Collapse
Affiliation(s)
- Rhiana C Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Weston T Fleming
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Brandy A Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Marta Trzeciak
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Pranav Senthilkumar
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Kentaro K Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madelyn M Hjort
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Madison M Martin
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Andrea D Sanders
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA
| | - Sam A Golden
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA 98195, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
3
|
Haun H, Hernandez R, Yan L, Flanigan M, Hon O, Lee S, Méndez H, Roland A, Taxier L, Kash T. Septo-hypothalamic regulation of binge-like alcohol consumption by the nociceptin system. Cell Rep 2025; 44:115482. [PMID: 40153436 DOI: 10.1016/j.celrep.2025.115482] [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: 03/13/2024] [Revised: 10/25/2024] [Accepted: 03/07/2025] [Indexed: 03/30/2025] Open
Abstract
High-intensity alcohol drinking during binge episodes contributes to the socioeconomic burden created by alcohol use disorders (AUDs), and nociceptin receptor (NOP) antagonists have emerged as a promising intervention. To better understand the contribution of the NOP system to binge drinking, we found that nociceptin-containing neurons of the lateral septum (LSPnoc) displayed increased excitability during withdrawal from binge-like alcohol drinking. LSPnoc activation promoted active avoidance and potentiated binge-like drinking behavior, whereas silencing of this population reduced alcohol drinking. LSPnoc form robust monosynaptic inputs locally within the LS and genetic deletion of NOP or microinjection of a NOP antagonist into the LS decreased alcohol intake. LSPnoc also project to the lateral hypothalamus and supramammillary nucleus of the hypothalamus, and genetic deletion of NOP from each site reduced alcohol drinking. Together, these findings implicate the septo-hypothalamic nociceptin system in excessive alcohol consumption and support NOP antagonist development for the treatment of AUD.
Collapse
Affiliation(s)
- Harold Haun
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Raul Hernandez
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Morgan Community College, Fort Morgan, CO 80701, USA
| | - Luzi Yan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Meghan Flanigan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Olivia Hon
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sophia Lee
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Hernán Méndez
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Alison Roland
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Lisa Taxier
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Thomas Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Chen M, Li J, Shan W, Yang J, Zuo Z. Auditory fear memory retrieval requires BLA-LS and LS-VMH circuitries via GABAergic and dopaminergic neurons. EMBO Rep 2025; 26:1816-1834. [PMID: 40055468 PMCID: PMC11977213 DOI: 10.1038/s44319-025-00403-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 04/09/2025] Open
Abstract
Fear and associated learning and memory are critical for developing defensive behavior. Excessive fear and anxiety are important components of post-traumatic stress disorder. However, the neurobiology of fear conditioning, especially tone-related fear memory retrieval, has not been clearly defined, which limits specific intervention development for patients with excessive fear and anxiety. Here, we show that auditory fear memory retrieval stimuli activate multiple brain regions including the lateral septum (LS). Inhibition of the LS and the connection between basolateral amygdala (BLA) and LS or between LS and ventromedial nucleus of the hypothalamus (VMH) attenuates tone-related fear conditioning and memory retrieval. Inhibiting GABAergic neurons or dopaminergic neurons in the LS also attenuates tone-related fear conditioning. Our data further show that fear conditioning is inhibited by blocking orexin B signaling in the LS. Our results indicate that the neural circuitries BLA-LS and LS-VMH are critical for tone-related fear conditioning and memory retrieval, and that GABAergic neurons, dopaminergic neurons and orexin signaling in the LS participate in this auditory fear conditioning.
Collapse
Affiliation(s)
- Miao Chen
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Weiran Shan
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jianjun Yang
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
5
|
Bredewold R, Washington C, Veenema AH. Vasopressin regulates social play behavior in sex-specific ways through glutamate modulation in the lateral septum. Neuropsychopharmacology 2025; 50:630-639. [PMID: 39304743 PMCID: PMC11845679 DOI: 10.1038/s41386-024-01987-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/23/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
Understanding the neural basis of social play in juvenile rats may ultimately help restore social play deficits in autistic children. We previously found that administration of a vasopressin (AVP) V1a receptor (V1aR) antagonist into the lateral septum (LS) increased social play behavior in male juvenile rats and decreased it in females. Here, we demonstrate that glutamate, but not GABA, is involved in this sex-specific regulation. First, we found a sex difference in extracellular LS glutamate/GABA ratio (lower in females) that was eliminated by V1aR antagonist infusion in the LS that caused an increase in glutamate release in females only. Second, infusion of the glutamate receptor agonist L-glutamic acid into the LS mimicked the V1aR antagonist-induced decrease in female social play while preventing the increase in male social play. Third, infusion of the glutamate receptor antagonists AP-5 and CNQX into the LS prevented the V1aR antagonist-induced decrease in female social play. Fourth, there were no sex differences in extracellular GABA release in the LS upon either V1aR antagonist infusion or in social play expression upon infusion of the GABA-A receptor agonist muscimol into the LS, suggesting that GABA is not involved in the sex-specific regulation of social play by the LS-AVP system. Last, we found no sex differences in the type (GAD1/2, somatostatin, calbindin 1, Sox9) of V1aR-expressing LS cells, suggesting other cellular mechanisms mediating the sex-specific effects on glutamate release in the LS by the LS-AVP system. In conclusion, we demonstrate that the LS-AVP system regulates social play sex-specifically via glutamatergic neurotransmission. These findings have relevance for potential sex-specific effects of AVP-based treatment of social deficits in children.
Collapse
Affiliation(s)
- Remco Bredewold
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Catherine Washington
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alexa H Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
6
|
Bono BS, Negishi K, Dumiaty Y, Ponce‐Ruiz MS, Akinbode TC, Baker KS, Spencer CDP, Mejia E, Guirguis M, Hebert AJ, Khan AM, Chee MJ. Brainwide Projections of Mouse Dopaminergic Zona Incerta Neurons. J Comp Neurol 2025; 533:e70039. [PMID: 40090880 PMCID: PMC11911292 DOI: 10.1002/cne.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/27/2025] [Accepted: 02/24/2025] [Indexed: 03/18/2025]
Abstract
The zona incerta (ZI) supports diverse behaviors including binge feeding, sleep-wake cycles, nociception, and hunting. Diverse ZI functions can be attributed to its heterogeneous neurochemical characterization, cytoarchitecture, and efferent connections. The ZI is predominantly GABAergic, but we recently identified a subset of medial ZI GABA cells that are marked by the enzyme tyrosine hydroxylase (TH) and produce dopamine (DA). While the role of GABA within the ZI is well studied, less is known about the functions of ZI DA cells. To identify potential roles of ZI DA cells, we further phenotyped them and mapped their efferent fiber projections. We showed that wild-type TH-immunoreactive (-ir) ZI cells did not express somatostatin or calretinin immunoreactivity. We next validated a Th-cre;L10-Egfp mouse line and found that medial Egfp ZI cells were more likely to be TH-ir. We therefore delivered a Cre-dependent virus into the medial ZI of Th-cre or Th-cre;L10-Egfp mice and selected two injection cases for full brain mapping, namely, cases with the lowest and highest colocalization between TH-ir and virally transduced, DsRed-labeled cells, to identify common target sites. Overall, DsRed-labeled fibers were distributed brainwide and were most prominent within the motor-related midbrain (MBmot), notably the periaqueductal gray area and superior colliculus. We also observed numerous DsRed-labeled fibers within the polymodal association cortex-related thalamus (DORpm), like paraventricular thalamic nucleus and nucleus of reunions, that processes external and internal sensory input. Overall, ZI DA cells displayed a similar fiber profile to ZI GABA cells and may integrate sensory input to coordinate motor output at their target sites.
Collapse
Affiliation(s)
- Bianca S. Bono
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Kenichiro Negishi
- Department of Biological SciencesThe University of Texas at El PasoEl PasoTexasUSA
| | - Yasmina Dumiaty
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Monica S. Ponce‐Ruiz
- Department of Biological SciencesThe University of Texas at El PasoEl PasoTexasUSA
| | | | - Kayla S. Baker
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | | | - Elizabeth Mejia
- Department of Biological SciencesThe University of Texas at El PasoEl PasoTexasUSA
| | - Marina Guirguis
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Alex J. Hebert
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Arshad M. Khan
- Department of Biological SciencesThe University of Texas at El PasoEl PasoTexasUSA
- Border Biomedical Research CenterThe University of Texas at El PasoEl PasoTexasUSA
| | - Melissa J. Chee
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| |
Collapse
|
7
|
Vincent CJ, Aguilar-Alvarez R, Vanderhoof SO, Mott DD, Jasnow AM. An amygdala-cortical circuit for encoding generalized fear memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633190. [PMID: 39868237 PMCID: PMC11761744 DOI: 10.1101/2025.01.15.633190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Generalized learning is a fundamental process observed across species, contexts, and sensory modalities that enables animals to use past experiences to adapt to changing conditions. Evidence suggests that the prefrontal cortex (PFC) extracts general features of an experience that can be used across multiple situations. The anterior cingulate cortex (ACC), a region of the PFC, is implicated in generalized fear responses to novel contexts. However, the ACC's role in encoding contextual information is poorly understood, especially under increased threat intensity that promotes generalization. Here, we show that synaptic plasticity within the ACC and signaling from amygdala inputs during fear learning are necessary for generalized fear responses to novel encountered contexts. The ACC did not encode specific fear to the training context, suggesting this region extracts general features of a threatening experience rather than specific contextual information. Together with our previous work, our results demonstrate that generalized learning about threatening contexts is encoded, in part, within an ascending amygdala-cortical circuit, whereas descending ACC projections to the amygdala drive generalized fear responses during exposure to novel contexts. Our results further demonstrate that schematic learning can occur in the PFC after single-trial learning, a process typically attributed to learning over many repeated learning episodes.
Collapse
|
8
|
Seo K, Won S, Lee HY, Sin Y, Lee S, Park H, Kim YG, Yang SY, Kim DJ, Suk K, Koo JW, Baek M, Choi SY, Lee H. Astrocytic inhibition of lateral septal neurons promotes diverse stress responses. Nat Commun 2024; 15:10091. [PMID: 39572547 PMCID: PMC11582824 DOI: 10.1038/s41467-024-54376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
Inhibitory neuronal circuits within the lateral septum (LS) play a key role in regulating mood and stress responses. Even though glial cells can modulate these circuits, the impact of astrocytes on LS neural circuits and their functional interactions remains largely unexplored. Here, we demonstrate that astrocytes exhibit increased intracellular Ca²⁺ levels in response to aversive sensory and social stimuli in both male and female mice. This astrocytic Ca²⁺ elevation inhibits neighboring LS neurons by reducing excitatory synaptic transmissions through A1R-mediated signaling in both the dorsal (LSd) and intermediate LS (LSi) and enhancing inhibitory synaptic transmission via A2AR-mediated signaling in the LSi. At the same time, astrocytes reduce inhibitory tone on distant LS neurons. In the LSd, astrocytes promote social avoidance and anxiety, as well as increased heart rate in socially stressed male mice. In contrast, astrocytes in the LSi contribute to elevated heart rate and heightened blood corticosterone levels in unstressed male mice. These results suggest that the dynamic interactions between astrocytes and neurons within the LS modulate physiological and behavioral responses to stressful experiences.
Collapse
Affiliation(s)
- Kain Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Sanghyun Won
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Hee-Yoon Lee
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Yeonju Sin
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Sangho Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Hyejin Park
- Laboratory Animal Resource Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Yong Geon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Seo Young Yang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Dong-Jae Kim
- Laboratory Animal Resource Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Myungin Baek
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea
| | - Se-Young Choi
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.
| | - Hyosang Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea.
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, Republic of Korea.
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.
| |
Collapse
|
9
|
Borie AM, Dromard Y, Chakraborty P, Fontanaud P, Andre EM, François A, Colson P, Muscatelli F, Guillon G, Desarménien MG, Jeanneteau F. Neuropeptide therapeutics to repress lateral septum neurons that disable sociability in an autism mouse model. Cell Rep Med 2024; 5:101781. [PMID: 39423809 PMCID: PMC11604546 DOI: 10.1016/j.xcrm.2024.101781] [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: 02/15/2024] [Revised: 06/20/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Confronting oxytocin and vasopressin deficits in autism spectrum disorders and rare syndromes brought promises and disappointments for the treatment of social disabilities. We searched downstream of oxytocin and vasopressin for targets alleviating social deficits in a mouse model of Prader-Willi syndrome and Schaaf-Yang syndrome, both associated with high prevalence of autism. We found a population of neurons in the lateral septum-activated on termination of social contacts-which oxytocin and vasopressin inhibit as per degree of peer affiliation. These are somatostatin neurons expressing oxytocin receptors coupled to GABA-B signaling, which are inhibited via GABA-A channels by vasopressin-excited GABA neurons. Loss of oxytocin or vasopressin signaling recapitulated the disease phenotype. By contrast, deactivation of somatostatin neurons or receptor signaling alleviated social deficits of disease models by increasing the duration of contacts with mates and strangers. These findings provide new insights into the treatment framework of social disabilities in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Amélie M Borie
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Yann Dromard
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Prabahan Chakraborty
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Pierre Fontanaud
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Emilie M Andre
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France; Département de Maieutique, University of Montpellier, 34090 Montpellier, France
| | - Amaury François
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Pascal Colson
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France; Department of Anesthesiology and Critical Care Medicine, Arnaud de Villeneuve Academic Hospital, Montpellier 34090 Montpellier, France
| | - Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée, INSERM, University of Aix-Marseille, 13273 Marseille, France
| | - Gilles Guillon
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Michel G Desarménien
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, Department of Neuroscience, Stress Hormones and Plasticity Unit, University of Montpellier, INSERM, CNRS, 34090 Montpellier, France.
| |
Collapse
|
10
|
Rahimi S, Joyce L, Fenzl T, Drexel M. Crosstalk between the subiculum and sleep-wake regulation: A review. J Sleep Res 2024; 33:e14134. [PMID: 38196146 DOI: 10.1111/jsr.14134] [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: 09/25/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 01/11/2024]
Abstract
The circuitry underlying the initiation, maintenance, and coordination of wakefulness, rapid eye movement sleep, and non-rapid eye movement sleep is not thoroughly understood. Sleep is thought to arise due to decreased activity in the ascending reticular arousal system, which originates in the brainstem and awakens the thalamus and cortex during wakefulness. Despite the conventional association of sleep-wake states with hippocampal rhythms, the mutual influence of the hippocampal formation in regulating vigilance states has been largely neglected. Here, we focus on the subiculum, the main output region of the hippocampal formation. The subiculum, particulary the ventral part, sends extensive monosynaptic projections to crucial regions implicated in sleep-wake regulation, including the thalamus, lateral hypothalamus, tuberomammillary nucleus, basal forebrain, ventrolateral preoptic nucleus, ventrolateral tegmental area, and suprachiasmatic nucleus. Additionally, second-order projections from the subiculum are received by the laterodorsal tegmental nucleus, locus coeruleus, and median raphe nucleus, suggesting the potential involvement of the subiculum in the regulation of the sleep-wake cycle. We also discuss alterations in the subiculum observed in individuals with sleep disorders and in sleep-deprived mice, underscoring the significance of investigating neuronal communication between the subiculum and pathways promoting both sleep and wakefulness.
Collapse
Affiliation(s)
- Sadegh Rahimi
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Leesa Joyce
- Clinic of Anesthesiology and Intensive Care, School of Medicine, Technical University of Munich, München, Germany
| | - Thomas Fenzl
- Clinic of Anesthesiology and Intensive Care, School of Medicine, Technical University of Munich, München, Germany
| | - Meinrad Drexel
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Bono BS, Negishi K, Dumiaty Y, Ponce MS, Akinbode TC, Baker KS, Spencer CDP, Mejia E, Guirguis M, Hebert AJ, Khan AM, Chee MJ. Brain-wide projections of mouse dopaminergic zona incerta neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611701. [PMID: 40093135 PMCID: PMC11908183 DOI: 10.1101/2024.09.06.611701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The zona incerta (ZI) supports diverse behaviors including binge feeding, sleep/wake cycles, nociception, and hunting. This diversity of functions can be attributed to the heterogenous neurochemicals, cytoarchitecture, and efferent connections that characterize the ZI. The ZI is predominantly GABAergic, but we recently identified a subset of medial ZI GABA cells that co-express dopamine (DA), as marked by the enzyme tyrosine hydroxylase (TH). While the role of GABA within the ZI is well studied, little is understood about the function of ZI DA cells. To identify potential roles of ZI DA cells we mapped the efferent fiber projections from Th-cre ZI cells. We first validated a Th-cre;L10-Egfp mouse line and found that medial Egfp ZI cells were more likely to co-express TH-immunoreactivity (TH-ir). We thus delivered a cre-dependent virus into the medial ZI of Th-cre or Th-cre;L10-Egfp mice and selected two injection cases for full brain mapping. We selected the cases with the lowest (17%) and highest (53%) percentage of colocalization between TH-ir and virus transfected cells labelled with DsRed. Overall, DsRed-labelled fibers were observed throughout the brain and were most prominent within motor-related regions of the midbrain (MBmot), notably the periaqueductal grey area and superior colliculus. We also observed considerable DsRed-labelled fibers within the polymodal cortex associated regions of the thalamus (DORpm), including the paraventricular thalamic nucleus and nucleus of reunions. Overall, ZI DA cells displayed a similar connectivity profile to ZI GABA cells, suggesting that ZI DA cells may perform synergistic or opposing functions at the same target sites.
Collapse
Affiliation(s)
- Bianca S. Bono
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Kenichiro Negishi
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Yasmina Dumiaty
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Monica S. Ponce
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| | | | - Kayla S. Baker
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | | | - Elizabeth Mejia
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Marina Guirguis
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Alex J. Hebert
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Arshad M. Khan
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Melissa J. Chee
- Department of Neuroscience, Carleton University, Ottawa, ON, K1S 5B6, Canada
| |
Collapse
|
12
|
Ramos-Prats A, Matulewicz P, Edenhofer ML, Wang KY, Yeh CW, Fajardo-Serrano A, Kress M, Kummer K, Lien CC, Ferraguti F. Loss of mGlu 5 receptors in somatostatin-expressing neurons alters negative emotional states. Mol Psychiatry 2024; 29:2774-2786. [PMID: 38575807 PMCID: PMC11420089 DOI: 10.1038/s41380-024-02541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024]
Abstract
Subtype 5 metabotropic glutamate receptors (mGlu5) are known to play an important role in regulating cognitive, social and valence systems. However, it remains largely unknown at which circuits and neuronal types mGlu5 act to influence these behavioral domains. Altered tissue- or cell-specific expression or function of mGlu5 has been proposed to contribute to the exacerbation of neuropsychiatric disorders. Here, we examined how these receptors regulate the activity of somatostatin-expressing (SST+) neurons, as well as their influence on behavior and brain rhythmic activity. Loss of mGlu5 in SST+ neurons elicited excitatory synaptic dysfunction in a region and sex-specific manner together with a range of emotional imbalances including diminished social novelty preference, reduced anxiety-like behavior and decreased freezing during retrieval of fear memories. In addition, the absence of mGlu5 in SST+ neurons during fear processing impaired theta frequency oscillatory activity in the medial prefrontal cortex and ventral hippocampus. These findings reveal a critical role of mGlu5 in controlling SST+ neurons excitability necessary for regulating negative emotional states.
Collapse
Affiliation(s)
- Arnau Ramos-Prats
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Pawel Matulewicz
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Kai-Yi Wang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Wei Yeh
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ana Fajardo-Serrano
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Francesco Ferraguti
- Institute of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
13
|
Guo SS, Gong Y, Zhang TT, Su XY, Wu YJ, Yan YX, Cao Y, Song XL, Xie JC, Wu D, Jiang Q, Li Y, Zhao X, Zhu MX, Xu TL, Liu MG. A thalamic nucleus reuniens-lateral septum-lateral hypothalamus circuit for comorbid anxiety-like behaviors in chronic itch. SCIENCE ADVANCES 2024; 10:eadn6272. [PMID: 39150998 PMCID: PMC11328909 DOI: 10.1126/sciadv.adn6272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/10/2024] [Indexed: 08/18/2024]
Abstract
Chronic itch often clinically coexists with anxiety symptoms, creating a vicious cycle of itch-anxiety comorbidities that are difficult to treat. However, the neuronal circuit mechanisms underlying the comorbidity of anxiety in chronic itch remain elusive. Here, we report anxiety-like behaviors in mouse models of chronic itch and identify γ-aminobutyric acid-releasing (GABAergic) neurons in the lateral septum (LS) as the key player in chronic itch-induced anxiety. In addition, chronic itch is accompanied with enhanced activity and synaptic plasticity of excitatory projections from the thalamic nucleus reuniens (Re) onto LS GABAergic neurons. Selective chemogenetic inhibition of the Re → LS circuit notably alleviated chronic itch-induced anxiety, with no impact on anxiety induced by restraint stress. Last, GABAergic neurons in lateral hypothalamus (LH) receive monosynaptic inhibition from LS GABAergic neurons to mediate chronic itch-induced anxiety. These findings underscore the potential significance of the Re → LS → LH pathway in regulating anxiety-like comorbid symptoms associated with chronic itch.
Collapse
Affiliation(s)
- Su-Shan Guo
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Gong
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ting-Ting Zhang
- Department of Anesthesiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Xin-Yu Su
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yan-Jiao Wu
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi-Xiao Yan
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yue Cao
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xing-Lei Song
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian-Cheng Xie
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Dehua Wu
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
| | - Qin Jiang
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Li
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuan Zhao
- Department of Anesthesiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tian-Le Xu
- Department of Anesthesiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| | - Ming-Gang Liu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Mental Health and Drug Discovery, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| |
Collapse
|
14
|
Goode TD, Alipio JB, Besnard A, Pathak D, Kritzer-Cheren MD, Chung A, Duan X, Sahay A. A dorsal hippocampus-prodynorphinergic dorsolateral septum-to-lateral hypothalamus circuit mediates contextual gating of feeding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606427. [PMID: 39149322 PMCID: PMC11326193 DOI: 10.1101/2024.08.02.606427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Adaptive regulation of feeding depends on linkage of internal states and food outcomes with contextual cues. Human brain imaging has identified dysregulation of a hippocampal-lateral hypothalamic area (LHA) network in binge eating, but mechanistic instantiation of underlying cell-types and circuitry is lacking. Here, we identify an evolutionary conserved and discrete Prodynorphin (Pdyn)-expressing subpopulation of Somatostatin (Sst)-expressing inhibitory neurons in the dorsolateral septum (DLS) that receives primarily dorsal, but not ventral, hippocampal inputs. DLS(Pdyn) neurons inhibit LHA GABAergic neurons and confer context- and internal state-dependent calibration of feeding. Viral deletion of Pdyn in the DLS mimicked effects seen with optogenetic silencing of DLS Pdyn INs, suggesting a potential role for DYNORPHIN-KAPPA OPIOID RECEPTOR signaling in contextual regulation of food-seeking. Together, our findings illustrate how the dorsal hippocampus has evolved to recruit an ancient LHA feeding circuit module through Pdyn DLS inhibitory neurons to link contextual information with regulation of food consumption.
Collapse
Affiliation(s)
- Travis D Goode
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Jason Bondoc Alipio
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Antoine Besnard
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Devesh Pathak
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Michael D Kritzer-Cheren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Ain Chung
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| | - Xin Duan
- Department of Ophthalmology, University of California, San Francisco, CA
- Department of Physiology, University of California, San Francisco, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, CA
| | - Amar Sahay
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- BROAD Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
15
|
Li XY, Zhang SY, Hong YZ, Chen ZG, Long Y, Yuan DH, Zhao JJ, Tang SS, Wang H, Hong H. TGR5-mediated lateral hypothalamus-dCA3-dorsolateral septum circuit regulates depressive-like behavior in male mice. Neuron 2024; 112:1795-1814.e10. [PMID: 38518778 DOI: 10.1016/j.neuron.2024.02.019] [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: 07/17/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
Although bile acids play a notable role in depression, the pathological significance of the bile acid TGR5 membrane-type receptor in this disorder remains elusive. Using depression models of chronic social defeat stress and chronic restraint stress in male mice, we found that TGR5 in the lateral hypothalamic area (LHA) predominantly decreased in GABAergic neurons, the excitability of which increased in depressive-like mice. Upregulation of TGR5 or inhibition of GABAergic excitability in LHA markedly alleviated depressive-like behavior, whereas down-regulation of TGR5 or enhancement of GABAergic excitability facilitated stress-induced depressive-like behavior. TGR5 also bidirectionally regulated excitability of LHA GABAergic neurons via extracellular regulated protein kinases-dependent Kv4.2 channels. Notably, LHA GABAergic neurons specifically innervated dorsal CA3 (dCA3) CaMKIIα neurons for mediation of depressive-like behavior. LHA GABAergic TGR5 exerted antidepressant-like effects by disinhibiting dCA3 CaMKIIα neurons projecting to the dorsolateral septum (DLS). These findings advance our understanding of TGR5 and the LHAGABA→dCA3CaMKIIα→DLSGABA circuit for the development of potential therapeutic strategies in depression.
Collapse
Affiliation(s)
- Xu-Yi Li
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi-Ya Zhang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi-Zhou Hong
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhi-Gang Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dan-Hua Yuan
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Jia Zhao
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Su-Su Tang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine/Nanhu Brain-Computer Interface Institute, Hangzhou 310013, China.
| | - Hao Hong
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
16
|
Jeong M, Jang JH, Oh SJ, Park J, Lee J, Hwang S, Oh YS. Maladaptation of dentate gyrus mossy cells mediates contextual discrimination deficit after traumatic stress. Cell Rep 2024; 43:114000. [PMID: 38527063 DOI: 10.1016/j.celrep.2024.114000] [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: 02/02/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 03/27/2024] Open
Abstract
Fear overgeneralization is a maladaptive response to traumatic stress that is associated with the inability to discriminate between threat and safety contexts, a hallmark feature of post-traumatic stress disorder (PTSD). However, the neural mechanisms underlying this deficit remain unclear. Here, we show that traumatic stress exposure impairs contextual discrimination between threat and safety contexts in the learned helplessness (LH) model. Mossy cells (MCs) in the dorsal hippocampus are suppressed in response to traumatic stress. Bidirectional manipulation of MC activity in the LH model reveals that MC inhibition is causally linked to impaired contextual discrimination. Mechanistically, MC inhibition increases the number of active granule cells in a given context, significantly overlapping context-specific ensembles. Our study demonstrates that maladaptive inhibition of MCs after traumatic stress is a substantial mechanism underlying fear overgeneralization with contextual discrimination deficit, suggesting a potential therapeutic target for cognitive symptoms of PTSD.
Collapse
Affiliation(s)
- Minseok Jeong
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jin-Hyeok Jang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Seo-Jin Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Jeongrak Park
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Junseop Lee
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Sehyeon Hwang
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yong-Seok Oh
- Department of Brain Sciences, Daegu-Gyeongbuk Institute of Science and Technology, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea; Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu 41062, Republic of Korea.
| |
Collapse
|
17
|
Almeida VN. Somatostatin and the pathophysiology of Alzheimer's disease. Ageing Res Rev 2024; 96:102270. [PMID: 38484981 DOI: 10.1016/j.arr.2024.102270] [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: 07/18/2023] [Revised: 03/09/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-β plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aβ production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aβ to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aβ release, fibrillation and plaque formation. Aβ plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.
Collapse
Affiliation(s)
- Victor N Almeida
- Institute of Psychiatry, Faculty of Medicine, University of São Paulo (USP), Brazil; Faculty of Languages, Federal University of Minas Gerais (UFMG), Brazil.
| |
Collapse
|
18
|
Li H, Kawatake-Kuno A, Inaba H, Miyake Y, Itoh Y, Ueki T, Oishi N, Murai T, Suzuki T, Uchida S. Discrete prefrontal neuronal circuits determine repeated stress-induced behavioral phenotypes in male mice. Neuron 2024; 112:786-804.e8. [PMID: 38228137 DOI: 10.1016/j.neuron.2023.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/26/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024]
Abstract
Chronic stress is a major risk factor for psychiatric disorders, including depression. Although depression is a highly heterogeneous syndrome, it remains unclear how chronic stress drives individual differences in behavioral responses. In this study, we developed a subtyping-based approach wherein stressed male mice were divided into four subtypes based on their behavioral patterns of social interaction deficits and anhedonia, the core symptoms of psychiatric disorders. We identified three prefrontal cortical neuronal projections that regulate repeated stress-induced behavioral phenotypes. Among them, the medial prefrontal cortex (mPFC)→anterior paraventricular thalamus (aPVT) pathway determines the specific behavioral subtype that exhibits both social deficits and anhedonia. Additionally, we identified the circuit-level molecular mechanism underlying this subtype: KDM5C-mediated epigenetic repression of Shisa2 transcription in aPVT projectors in the mPFC led to social deficits and anhedonia. Thus, we provide a set of biological aspects at the cellular, molecular, and epigenetic levels that determine distinctive stress-induced behavioral phenotypes.
Collapse
Affiliation(s)
- Haiyan Li
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ayako Kawatake-Kuno
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiromichi Inaba
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuka Miyake
- SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Yukihiro Itoh
- SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Takatoshi Ueki
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Naoya Oishi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshiya Murai
- Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takayoshi Suzuki
- SANKEN, Osaka University, 8-1 Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Shusaku Uchida
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan; Kyoto University Medical Science and Business Liaison Organization, Medical Innovation Center, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Division of Neuropsychiatry, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan; Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| |
Collapse
|
19
|
Zhang Q, Xue Y, Wei K, Wang H, Ma Y, Wei Y, Fan Y, Gao L, Yao H, Wu F, Ding X, Zhang Q, Ding J, Fan Y, Lu M, Hu G. Locus Coeruleus-Dorsolateral Septum Projections Modulate Depression-Like Behaviors via BDNF But Not Norepinephrine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303503. [PMID: 38155473 PMCID: PMC10933643 DOI: 10.1002/advs.202303503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Locus coeruleus (LC) dysfunction is involved in the pathophysiology of depression; however, the neural circuits and specific molecular mechanisms responsible for this dysfunction remain unclear. Here, it is shown that activation of tyrosine hydroxylase (TH) neurons in the LC alleviates depression-like behaviors in susceptible mice. The dorsolateral septum (dLS) is the most physiologically relevant output from the LC under stress. Stimulation of the LCTH -dLSSST innervation with optogenetic and chemogenetic tools bidirectionally can regulate depression-like behaviors in both male and female mice. Mechanistically, it is found that brain-derived neurotrophic factor (BDNF), but not norepinephrine, is required for the circuit to produce antidepressant-like effects. Genetic overexpression of BDNF in the circuit or supplementation with BDNF protein in the dLS is sufficient to produce antidepressant-like effects. Furthermore, viral knockdown of BDNF in this circuit abolishes the antidepressant-like effect of ketamine, but not fluoxetine. Collectively, these findings underscore the notable antidepressant-like role of the LCTH -dLSSST pathway in depression via BDNF-TrkB signaling.
Collapse
Affiliation(s)
- Qian Zhang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - You Xue
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Ke Wei
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Hao Wang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yuan Ma
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yao Wei
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Yi Fan
- Department of NeurologyAffiliated Nanjing Brain HospitalNanjing Medical UniversityNanjing210024China
| | - Lei Gao
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Hang Yao
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Fangfang Wu
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Xin Ding
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Qingyu Zhang
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
| | - Jianhua Ding
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Yi Fan
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Ming Lu
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| | - Gang Hu
- Department of PharmacologySchool of MedicineNanjing University of Chinese MedicineNanjing210023China
- Jiangsu Key Laboratory of NeurodegenerationDepartment of PharmacologyNanjing Medical UniversityNanjing211166China
| |
Collapse
|
20
|
Rodriguez LA, Tran MN, Garcia-Flores R, Oh S, Phillips RA, Pattie EA, Divecha HR, Kim SH, Shin JH, Lee YK, Montoya C, Jaffe AE, Collado-Torres L, Page SC, Martinowich K. TrkB-dependent regulation of molecular signaling across septal cell types. Transl Psychiatry 2024; 14:52. [PMID: 38263132 PMCID: PMC10805920 DOI: 10.1038/s41398-024-02758-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
The lateral septum (LS), a GABAergic structure located in the basal forebrain, is implicated in social behavior, learning, and memory. We previously demonstrated that expression of tropomyosin kinase receptor B (TrkB) in LS neurons is required for social novelty recognition. To better understand molecular mechanisms by which TrkB signaling controls behavior, we locally knocked down TrkB in LS and used bulk RNA-sequencing to identify changes in gene expression downstream of TrkB. TrkB knockdown induces upregulation of genes associated with inflammation and immune responses, and downregulation of genes associated with synaptic signaling and plasticity. Next, we generated one of the first atlases of molecular profiles for LS cell types using single nucleus RNA-sequencing (snRNA-seq). We identified markers for the septum broadly, and the LS specifically, as well as for all neuronal cell types. We then investigated whether the differentially expressed genes (DEGs) induced by TrkB knockdown map to specific LS cell types. Enrichment testing identified that downregulated DEGs are broadly expressed across neuronal clusters. Enrichment analyses of these DEGs demonstrated that downregulated genes are uniquely expressed in the LS, and associated with either synaptic plasticity or neurodevelopmental disorders. Upregulated genes are enriched in LS microglia, associated with immune response and inflammation, and linked to both neurodegenerative disease and neuropsychiatric disorders. In addition, many of these genes are implicated in regulating social behaviors. In summary, the findings implicate TrkB signaling in the LS as a critical regulator of gene networks associated with psychiatric disorders that display social deficits, including schizophrenia and autism, and with neurodegenerative diseases, including Alzheimer's.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Renee Garcia-Flores
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Seyun Oh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Robert A Phillips
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Yong Kyu Lee
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Carly Montoya
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
21
|
Park K, Kohl MM, Kwag J. Memory encoding and retrieval by retrosplenial parvalbumin interneurons are impaired in Alzheimer's disease model mice. Curr Biol 2024; 34:434-443.e4. [PMID: 38157861 DOI: 10.1016/j.cub.2023.12.014] [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: 07/07/2023] [Revised: 10/23/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Memory deficits in Alzheimer's disease (AD) show a strong link with GABAergic interneuron dysfunctions.1,2,3,4,5,6,7 The ensemble dynamics of GABAergic interneurons represent memory encoding and retrieval,8,9,10,11,12 but how GABAergic interneuron dysfunction affects inhibitory ensemble dynamics in AD is unknown. As the retrosplenial cortex (RSC) is critical for episodic memory13,14,15,16 and is affected by β-amyloid accumulation in early AD,17,18,19,20,21 we address this question by performing Ca2+ imaging in RSC parvalbumin (PV)-expressing interneurons during a contextual fear memory task in healthy control mice and the 5XFAD mouse model of AD. We found that populations of PV interneurons responsive to aversive electric foot shocks during contextual fear conditioning (shock-responsive) significantly decreased in the 5XFAD mice, indicating dysfunctions in the recruitment of memory-encoding PV interneurons. In the control mice, ensemble activities of shock-responsive PV interneurons were selectively upregulated during the freezing epoch of the contextual fear memory retrieval, manifested by synaptic potentiation of PV interneuron-mediated inhibition. However, such changes in ensemble dynamics during memory retrieval and synaptic plasticity were both absent in the 5XFAD mice. Optogenetic silencing of PV interneurons during contextual fear conditioning in the control mice mimicked the memory deficits in the 5XFAD mice, while optogenetic activation of PV interneurons in the 5XFAD mice restored memory retrieval. These results demonstrate the critical roles of contextual fear memory-encoding PV interneurons for memory retrieval. Furthermore, synaptic dysfunction of PV interneurons may disrupt the recruitment of PV interneurons and their ensemble dynamics underlying contextual fear memory retrieval, subsequently leading to memory deficits in AD.
Collapse
Affiliation(s)
- Kyerl Park
- Department of Brain and Cognitive Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Korea; Department of Brain and Cognitive Engineering, Korea University, Anam-ro 145, Seongbuk-gu, Seoul 02841, Korea
| | - Michael M Kohl
- School of Psychology and Neuroscience, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Jeehyun Kwag
- Department of Brain and Cognitive Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
22
|
Simon RC, Fleming WT, Senthilkumar P, Briones BA, Ishii KK, Hjort MM, Martin MM, Hashikawa K, Sanders AD, Golden SA, Stuber GD. Opioid-driven disruption of the septal complex reveals a role for neurotensin-expressing neurons in withdrawal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575766. [PMID: 38293241 PMCID: PMC10827099 DOI: 10.1101/2024.01.15.575766] [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: 02/01/2024]
Abstract
Because opioid withdrawal is an intensely aversive experience, persons with opioid use disorder (OUD) often relapse to avoid it. The lateral septum (LS) is a forebrain structure that is important in aversion processing, and previous studies have linked the lateral septum (LS) to substance use disorders. It is unclear, however, which precise LS cell types might contribute to the maladaptive state of withdrawal. To address this, we used single-nucleus RNA-sequencing to interrogate cell type specific gene expression changes induced by chronic morphine and withdrawal. We discovered that morphine globally disrupted the transcriptional profile of LS cell types, but Neurotensin-expressing neurons (Nts; LS-Nts neurons) were selectively activated by naloxone. Using two-photon calcium imaging and ex vivo electrophysiology, we next demonstrate that LS-Nts neurons receive enhanced glutamatergic drive in morphine-dependent mice and remain hyperactivated during opioid withdrawal. Finally, we showed that activating and silencing LS-Nts neurons during opioid withdrawal regulates pain coping behaviors and sociability. Together, these results suggest that LS-Nts neurons are a key neural substrate involved in opioid withdrawal and establish the LS as a crucial regulator of adaptive behaviors, specifically pertaining to OUD.
Collapse
Affiliation(s)
- Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Weston T. Fleming
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Pranav Senthilkumar
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Kentaro K. Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Madelyn M. Hjort
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Madison M. Martin
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Andrea D. Sanders
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
| | - Sam A. Golden
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA, 98195, USA
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
23
|
Dromard Y, Borie AM, Chakraborty P, Muscatelli F, Guillon G, Desarménien MG, Jeanneteau F. Disengagement of somatostatin neurons from lateral septum circuitry by oxytocin and vasopressin restores social-fear extinction and suppresses aggression outbursts in Prader-Willi syndrome model. Biol Psychiatry 2023; 95:S0006-3223(23)01661-X. [PMID: 39491230 PMCID: PMC11216544 DOI: 10.1016/j.biopsych.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 07/03/2024]
Abstract
BACKGROUND Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. METHODS Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. RESULTS OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. CONCLUSIONS SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.
Collapse
Affiliation(s)
- Yann Dromard
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Amélie M Borie
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Prabahan Chakraborty
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée, INSERM, University of Aix-Marseille, Marseille, France
| | - Gilles Guillon
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Michel G Desarménien
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France.
| |
Collapse
|
24
|
García MT, Tran DN, Peterson RE, Stegmann SK, Hanson SM, Reid CM, Xie Y, Vu S, Harwell CC. A developmentally defined population of neurons in the lateral septum controls responses to aversive stimuli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.24.559205. [PMID: 37873286 PMCID: PMC10592641 DOI: 10.1101/2023.09.24.559205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
When interacting with their environment, animals must balance exploratory and defensive behavior to evaluate and respond to potential threats. The lateral septum (LS) is a structure in the ventral forebrain that calibrates the magnitude of behavioral responses to stress-related external stimuli, including the regulation of threat avoidance. The complex connectivity between the LS and other parts of the brain, together with its largely unexplored neuronal diversity, makes it difficult to understand how defined LS circuits control specific behaviors. Here, we describe a mouse model in which a population of neurons with a common developmental origin (Nkx2.1-lineage neurons) are absent from the LS. Using a combination of circuit tracing and behavioral analyses, we found that these neurons receive inputs from the perifornical area of the anterior hypothalamus (PeFAH) and are specifically activated in stressful contexts. Mice lacking Nkx2.1-lineage LS neurons display increased exploratory behavior even under stressful conditions. Our study extends the current knowledge about how defined neuronal populations within the LS can evaluate contextual information to select appropriate behavioral responses. This is a necessary step towards understanding the crucial role that the LS plays in neuropsychiatric conditions where defensive behavior is dysregulated, such as anxiety and aggression disorders.
Collapse
Affiliation(s)
- Miguel Turrero García
- Department of Neurology, University of California, San Francisco; San Francisco, CA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; San Francisco, CA
| | - Diana N. Tran
- Department of Neurobiology, Harvard Medical School; Boston, MA
| | | | | | - Sarah M. Hanson
- Department of Neurology, University of California, San Francisco; San Francisco, CA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; San Francisco, CA
| | - Christopher M. Reid
- Department of Neurology, University of California, San Francisco; San Francisco, CA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; San Francisco, CA
- Ph.D. Program in Neuroscience, Harvard University; Boston, MA
| | - Yajun Xie
- Department of Neurology, University of California, San Francisco; San Francisco, CA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; San Francisco, CA
| | - Steve Vu
- Department of Neurobiology, Harvard Medical School; Boston, MA
| | - Corey C. Harwell
- Department of Neurology, University of California, San Francisco; San Francisco, CA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research; San Francisco, CA
- Chan Zuckerberg Biohub San Francisco; San Francisco, CA
- Lead contact
| |
Collapse
|
25
|
Dos Santos WO, Juliano VAL, Chaves FM, Vieira HR, Frazao R, List EO, Kopchick JJ, Munhoz CD, Donato J. Growth Hormone Action in Somatostatin Neurons Regulates Anxiety and Fear Memory. J Neurosci 2023; 43:6816-6829. [PMID: 37625855 PMCID: PMC10552943 DOI: 10.1523/jneurosci.0254-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/28/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023] Open
Abstract
Dysfunctions in growth hormone (GH) secretion increase the prevalence of anxiety and other neuropsychiatric diseases. GH receptor (GHR) signaling in the amygdala has been associated with fear memory, a key feature of posttraumatic stress disorder. However, it is currently unknown which neuronal population is targeted by GH action to influence the development of neuropsychiatric diseases. Here, we showed that approximately 60% of somatostatin (SST)-expressing neurons in the extended amygdala are directly responsive to GH. GHR ablation in SST-expressing cells (SSTΔGHR mice) caused no alterations in energy or glucose metabolism. Notably, SSTΔGHR male mice exhibited increased anxiety-like behavior in the light-dark box and elevated plus maze tests, whereas SSTΔGHR females showed no changes in anxiety. Using auditory Pavlovian fear conditioning, both male and female SSTΔGHR mice exhibited a significant reduction in fear memory. Conversely, GHR ablation in SST neurons did not affect memory in the novel object recognition test. Gene expression was analyzed in a micro punch comprising the central nucleus of the amygdala (CEA) and basolateral (BLA) complex. GHR ablation in SST neurons caused sex-dependent changes in the expression of factors involved in synaptic plasticity and function. In conclusion, GHR expression in SST neurons is necessary to regulate anxiety in males, but not female mice. GHR ablation in SST neurons also decreases fear memory and affects gene expression in the amygdala, although marked sex differences were observed. Our findings identified for the first time a neurochemically-defined neuronal population responsible for mediating the effects of GH on behavioral aspects associated with neuropsychiatric diseases.SIGNIFICANCE STATEMENT Hormone action in the brain regulates different neurological aspects, affecting the predisposition to neuropsychiatric disorders, like depression, anxiety, and posttraumatic stress disorder. Growth hormone (GH) receptor is widely expressed in the brain, but the exact function of neuronal GH action is not fully understood. Here, we showed that mice lacking the GH receptor in a group of neurons that express the neuropeptide somatostatin exhibit increased anxiety. However, this effect is only observed in male mice. In contrast, the absence of the GH receptor in somatostatin-expressing neurons decreases fear memory, a key feature of posttraumatic stress disorder, in males and females. Thus, our study identified a specific group of neurons in which GH acts to affect the predisposition to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Willian O Dos Santos
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Vitor A L Juliano
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Fernanda M Chaves
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Henrique R Vieira
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Renata Frazao
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens 45701, Ohio
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens 45701, Ohio
| | - Carolina D Munhoz
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
26
|
Rodriguez LA, Tran MN, Garcia-Flores R, Pattie EA, Divecha HR, Kim SH, Shin JH, Lee YK, Montoya C, Jaffe AE, Collado-Torres L, Page SC, Martinowich K. TrkB-dependent regulation of molecular signaling across septal cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547069. [PMID: 37425939 PMCID: PMC10327212 DOI: 10.1101/2023.06.29.547069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The lateral septum (LS), a GABAergic structure located in the basal forebrain, is implicated in social behavior, learning and memory. We previously demonstrated that expression of tropomyosin kinase receptor B (TrkB) in LS neurons is required for social novelty recognition. To better understand molecular mechanisms by which TrkB signaling controls behavior, we locally knocked down TrkB in LS and used bulk RNA-sequencing to identify changes in gene expression downstream of TrkB. TrkB knockdown induces upregulation of genes associated with inflammation and immune responses, and downregulation of genes associated with synaptic signaling and plasticity. Next, we generated one of the first atlases of molecular profiles for LS cell types using single nucleus RNA-sequencing (snRNA-seq). We identified markers for the septum broadly, and the LS specifically, as well as for all neuronal cell types. We then investigated whether the differentially expressed genes (DEGs) induced by TrkB knockdown map to specific LS cell types. Enrichment testing identified that downregulated DEGs are broadly expressed across neuronal clusters. Enrichment analyses of these DEGs demonstrated that downregulated genes are uniquely expressed in the LS, and associated with either synaptic plasticity or neurodevelopmental disorders. Upregulated genes are enriched in LS microglia, associated with immune response and inflammation, and linked to both neurodegenerative disease and neuropsychiatric disorders. In addition, many of these genes are implicated in regulating social behaviors. In summary, the findings implicate TrkB signaling in the LS as a critical regulator of gene networks associated with psychiatric disorders that display social deficits, including schizophrenia and autism, and with neurodegenerative diseases, including Alzheimer's.
Collapse
Affiliation(s)
- Lionel A. Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Renee Garcia-Flores
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A. Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Yong Kyu Lee
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Carly Montoya
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Andrew E. Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205, USA
| |
Collapse
|
27
|
Shi HJ, Wang S, Wang XP, Zhang RX, Zhu LJ. Hippocampus: Molecular, Cellular, and Circuit Features in Anxiety. Neurosci Bull 2023; 39:1009-1026. [PMID: 36680709 PMCID: PMC10264315 DOI: 10.1007/s12264-023-01020-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/13/2022] [Indexed: 01/22/2023] Open
Abstract
Anxiety disorders are currently a major psychiatric and social problem, the mechanisms of which have been only partially elucidated. The hippocampus serves as a major target of stress mediators and is closely related to anxiety modulation. Yet so far, its complex anatomy has been a challenge for research on the mechanisms of anxiety regulation. Recent advances in imaging, virus tracking, and optogenetics/chemogenetics have permitted elucidation of the activity, connectivity, and function of specific cell types within the hippocampus and its connected brain regions, providing mechanistic insights into the elaborate organization of the hippocampal circuitry underlying anxiety. Studies of hippocampal neurotransmitter systems, including glutamatergic, GABAergic, cholinergic, dopaminergic, and serotonergic systems, have contributed to the interpretation of the underlying neural mechanisms of anxiety. Neuropeptides and neuroinflammatory factors are also involved in anxiety modulation. This review comprehensively summarizes the hippocampal mechanisms associated with anxiety modulation, based on molecular, cellular, and circuit properties, to provide tailored targets for future anxiety treatment.
Collapse
Affiliation(s)
- Hu-Jiang Shi
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Shuang Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xin-Ping Wang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui-Xin Zhang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, 210009, China.
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| |
Collapse
|
28
|
Wang M, Li P, Li Z, da Silva BS, Zheng W, Xiang Z, He Y, Xu T, Cordeiro C, Deng L, Dai Y, Ye M, Lin Z, Zhou J, Zhou X, Ye F, Cunha RA, Chen J, Guo W. Lateral septum adenosine A 2A receptors control stress-induced depressive-like behaviors via signaling to the hypothalamus and habenula. Nat Commun 2023; 14:1880. [PMID: 37019936 PMCID: PMC10076302 DOI: 10.1038/s41467-023-37601-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Major depressive disorder ranks as a major burden of disease worldwide, yet the current antidepressant medications are limited by frequent non-responsiveness and significant side effects. The lateral septum (LS) is thought to control of depression, however, the cellular and circuit substrates are largely unknown. Here, we identified a subpopulation of LS GABAergic adenosine A2A receptors (A2AR)-positive neurons mediating depressive symptoms via direct projects to the lateral habenula (LHb) and the dorsomedial hypothalamus (DMH). Activation of A2AR in the LS augmented the spiking frequency of A2AR-positive neurons leading to a decreased activation of surrounding neurons and the bi-directional manipulation of LS-A2AR activity demonstrated that LS-A2ARs are necessary and sufficient to trigger depressive phenotypes. Thus, the optogenetic modulation (stimulation or inhibition) of LS-A2AR-positive neuronal activity or LS-A2AR-positive neurons projection terminals to the LHb or DMH, phenocopied depressive behaviors. Moreover, A2AR are upregulated in the LS in two male mouse models of repeated stress-induced depression. This identification that aberrantly increased A2AR signaling in the LS is a critical upstream regulator of repeated stress-induced depressive-like behaviors provides a neurophysiological and circuit-based justification of the antidepressant potential of A2AR antagonists, prompting their clinical translation.
Collapse
Affiliation(s)
- Muran Wang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Peijun Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
| | - Zewen Li
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Beatriz S da Silva
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- Portuguese National Institute of Legal Medicine and Forensic Sciences (INMLCF, IP), Coimbra, Portugal
| | - Wu Zheng
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Zhenghua Xiang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology, Ministry of Education, Naval Medical University, Shanghai, China
| | - Yan He
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Tao Xu
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Cristina Cordeiro
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- Portuguese National Institute of Legal Medicine and Forensic Sciences (INMLCF, IP), Coimbra, Portugal
| | - Lu Deng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
| | - Yuwei Dai
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Mengqian Ye
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Zhiqing Lin
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jianhong Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xuzhao Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Fenfen Ye
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jiangfan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
29
|
Li YD, Luo YJ, Song J. Optimizing memory performance and emotional states: multi-level enhancement of adult hippocampal neurogenesis. Curr Opin Neurobiol 2023; 79:102693. [PMID: 36822141 PMCID: PMC10023407 DOI: 10.1016/j.conb.2023.102693] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/03/2023] [Accepted: 01/12/2023] [Indexed: 02/25/2023]
Abstract
Adult hippocampal neurogenesis (AHN) plays a key role in modulating memory and emotion processing. A fundamental question remains on how to effectively modulate AHN to improve hippocampal function. Here, we review recent work on how distinct aspects of hippocampal neurogenesis, including the number, maturation state, and activity of adult-born neurons (ABNs), contribute to overall hippocampal function. We propose multi-level enhancement of hippocampal neurogenesis with the combination of increased number, elevated activity, and enhanced maturation of ABNs as a potential strategy to optimize overall hippocampal performance. In addition, integration of ABNs induces significant remodeling of the local hippocampal circuits, which may in turn modulates brain-wide network dynamics. We discuss recent progress on how integration of ABNs contributes to local hippocampal circuit and brain-wide network dynamics during behavior.
Collapse
Affiliation(s)
- Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. https://twitter.com/yadlee2
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
30
|
Bredewold R, Washington C, Veenema AH. Vasopressin regulates social play behavior in sex-specific ways through glutamate modulation in the lateral septum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535148. [PMID: 37034639 PMCID: PMC10081315 DOI: 10.1101/2023.03.31.535148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Social play is a highly rewarding behavior that is essential for the development of social skills. Social play is impaired in children diagnosed with autism, a disorder with a strong sex bias in prevalence. We recently showed that the arginine vasopressin (AVP) system in the lateral septum (LS) regulates social play behavior sex-specifically in juvenile rats: Administration of a AVP 1a receptor (V1aR) antagonist increased social play behavior in males and decreased it in females. Here, we demonstrate that glutamate, but not GABA, is involved in the sex-specific regulation of social play by the LS-AVP system. First, males show higher extracellular glutamate concentrations in the LS than females while they show similar extracellular GABA concentrations. This resulted in a baseline sex difference in excitatory/inhibitory balance, which was eliminated by V1aR antagonist administration into the LS: V1aR antagonist increased extracellular glutamate release in females but not in males. Second, administration of the glutamate receptor agonist L-glutamic acid into the LS prevented the V1aR antagonist-induced increase in social play behavior in males while mimicking the V1aR antagonist-induced decrease in social play behavior in females. Third, administration of the glutamate receptor antagonists AP-5 and CNQX into the LS prevented the V1aR antagonist-induced decrease in social play behavior in females. Last, both sexes showed increases in extracellular LS-GABA release upon V1aR antagonist administration into the LS and decreases in social play behavior upon administration of the GABA-A receptor agonist muscimol into the LS, suggesting that GABA is not involved in the sex-specific regulation of social play by the LS-AVP system. Finally, to start identifying the cellular mechanism mediating the sex-specific effects of the LS-AVP system on LS-glutamate, we determined the presence of potential sex differences in the type of LS cells expressing V1aR. However, no sex differences were found in the percentage of Avpr1a+ LS cells expressing markers for either GABAergic neurons, somatostatin-expressing neurons, calbindin 1-expressing neurons, or astrocytes. In conclusion, these findings demonstrate that the LS-AVP system regulates social play sex-specifically via differential local glutamatergic neurotransmission in male and female juvenile rats. Further research is required to uncover the underlying cellular mechanism.
Collapse
Affiliation(s)
- Remco Bredewold
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Catherine Washington
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Alexa H Veenema
- Neurobiology of Social Behavior Laboratory, Department of Psychology and Neuroscience Program, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
31
|
Li L, Durand-de Cuttoli R, Aubry AV, Burnett CJ, Cathomas F, Parise LF, Chan KL, Morel C, Yuan C, Shimo Y, Lin HY, Wang J, Russo SJ. Social trauma engages lateral septum circuitry to occlude social reward. Nature 2023; 613:696-703. [PMID: 36450985 PMCID: PMC9876792 DOI: 10.1038/s41586-022-05484-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/25/2022] [Indexed: 12/05/2022]
Abstract
In humans, traumatic social experiences can contribute to psychiatric disorders1. It is suggested that social trauma impairs brain reward function such that social behaviour is no longer rewarding, leading to severe social avoidance2,3. In rodents, the chronic social defeat stress (CSDS) model has been used to understand the neurobiology underlying stress susceptibility versus resilience following social trauma, yet little is known regarding its impact on social reward4,5. Here we show that, following CSDS, a subset of male and female mice, termed susceptible (SUS), avoid social interaction with non-aggressive, same-sex juvenile C57BL/6J mice and do not develop context-dependent social reward following encounters with them. Non-social stressors have no effect on social reward in either sex. Next, using whole-brain Fos mapping, in vivo Ca2+ imaging and whole-cell recordings, we identified a population of stress/threat-responsive lateral septum neurotensin (NTLS) neurons that are activated by juvenile social interactions only in SUS mice, but not in resilient or unstressed control mice. Optogenetic or chemogenetic manipulation of NTLS neurons and their downstream connections modulates social interaction and social reward. Together, these data suggest that previously rewarding social targets are possibly perceived as social threats in SUS mice, resulting from hyperactive NTLS neurons that occlude social reward processing.
Collapse
Affiliation(s)
- Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio V Aubry
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Joseph Burnett
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L Chan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carole Morel
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chongzhen Yuan
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J Peters VA Medical Center, Research & Development, New York, NY, USA
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsiao-Yun Lin
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J Peters VA Medical Center, Research & Development, New York, NY, USA
| | - Jun Wang
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J Peters VA Medical Center, Research & Development, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
32
|
An M, Kim HK, Park H, Kim K, Heo G, Park HE, Chung C, Kim SY. Lateral Septum Somatostatin Neurons are Activated by Diverse Stressors. Exp Neurobiol 2022; 31:376-389. [PMID: 36631846 PMCID: PMC9841747 DOI: 10.5607/en22024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/31/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023] Open
Abstract
The lateral septum (LS) is a forebrain structure that has been implicated in a wide range of behavioral and physiological responses to stress. However, the specific populations of neurons in the LS that mediate stress responses remain incompletely understood. Here, we show that neurons in the dorsal lateral septum (LSd) that express the somatostatin gene (hereafter, LSdSst neurons) are activated by diverse stressors. Retrograde tracing from LSdSst neurons revealed that these neurons are directly innervated by neurons in the locus coeruleus (LC), the primary source of norepinephrine well-known to mediate diverse stress-related functions in the brain. Consistently, we found that norepinephrine increased excitatory synaptic transmission onto LSdSst neurons, suggesting the functional connectivity between LSdSst neurons and LC noradrenergic neurons. However, optogenetic stimulation of LSdSst neurons did not affect stress-related behaviors or autonomic functions, likely owing to the functional heterogeneity within this population. Together, our findings show that LSdSst neurons are activated by diverse stressors and suggest that norepinephrine released from the LC may modulate the activity of LSdSst neurons under stressful circumstances.
Collapse
Affiliation(s)
- Myungmo An
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea,Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Hyun-Kyung Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea,Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea
| | - Kyunghoe Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea,Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Gyuryang Heo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - Han-Eol Park
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea,
ChiHye Chung, TEL: 82-2-450-0432, e-mail:
| | - Sung-Yon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul 08826, Korea,Department of Chemistry, Seoul National University, Seoul 08826, Korea,To whom correspondence should be addressed. Sung-Yon Kim, TEL: 82-2-880-4994, e-mail:
| |
Collapse
|
33
|
Terstege DJ, Epp JR. Network Neuroscience Untethered: Brain-Wide Immediate Early Gene Expression for the Analysis of Functional Connectivity in Freely Behaving Animals. BIOLOGY 2022; 12:34. [PMID: 36671727 PMCID: PMC9855808 DOI: 10.3390/biology12010034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022]
Abstract
Studying how spatially discrete neuroanatomical regions across the brain interact is critical to advancing our understanding of the brain. Traditional neuroimaging techniques have led to many important discoveries about the nature of these interactions, termed functional connectivity. However, in animal models these traditional neuroimaging techniques have generally been limited to anesthetized or head-fixed setups or examination of small subsets of neuroanatomical regions. Using the brain-wide expression density of immediate early genes (IEG), we can assess brain-wide functional connectivity underlying a wide variety of behavioural tasks in freely behaving animal models. Here, we provide an overview of the necessary steps required to perform IEG-based analyses of functional connectivity. We also outline important considerations when designing such experiments and demonstrate the implications of these considerations using an IEG-based network dataset generated for the purpose of this review.
Collapse
Affiliation(s)
| | - Jonathan R. Epp
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
34
|
Optogenetic Suppression of Lateral Septum Somatostatin Neurons Enhances Hippocampus Cholinergic Theta Oscillations and Local Synchrony. Brain Sci 2022; 13:brainsci13010001. [PMID: 36671983 PMCID: PMC9856160 DOI: 10.3390/brainsci13010001] [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: 11/27/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
The septal complex regulates both motivated and innate behaviors, chiefly by the action of its diverse population of long-range projection neurons. A small population of somatostatin-expressing GABAergic cells in the lateral septum projects deep into subcortical regions, yet on its way it also targets neighboring medial septum neurons that profusely innervate cortical targets by ascending synaptic pathways. Here, we used optogenetic stimulation and extracellular recordings in acutely anesthetized transgenic mice to show that lateral septum somatostatin neurons can disinhibit the cholinergic septo-hippocampal pathway, thus enhancing the amplitude and synchrony of theta oscillations while depressing sharp-wave ripple episodes in the dorsal hippocampus. These results suggest that septal somatostatin cells can recruit ascending cholinergic pathways to promote hippocampal theta oscillations.
Collapse
|
35
|
Zhong C, Wang L, Cao Y, Sun C, Huang J, Wang X, Pan S, He S, Huang K, Lu Z, Xu F, Lu Y, Wang L. A neural circuit from the dorsal CA3 to the dorsomedial hypothalamus mediates balance between risk exploration and defense. Cell Rep 2022; 41:111570. [PMID: 36323263 DOI: 10.1016/j.celrep.2022.111570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/28/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
An appropriate balance between explorative and defensive behavior is essential for the survival and reproduction of prey animals in risky environments. However, the neural circuit and mechanism that allow for such a balance remains poorly understood. Here, we use a semi-naturalistic predator threat test (PTT) to observe and quantify the defense-exploration balance, especially risk exploration behavior in mice. During the PTT, the activity of the putative dorsal CA3 glutamatergic neurons (dCA3Glu) is suppressed by predatory threat and risk exploration, whereas the neurons are activated during contextual exploration. Moreover, optogenetic excitation of these neurons induces a significant increase in risk exploration. A circuit, comprising the dorsal CA3, dorsal lateral septal, and dorsomedial hypothalamic (dCA3Glu-dLSGABA-DMH) areas, may be involved. Moreover, activation of the dCA3Glu-dLSGABA-DMH circuit promotes the switch from defense to risk exploration and suppresses threat-induced increase in arousal.
Collapse
Affiliation(s)
- Cheng Zhong
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lulu Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Yi Cao
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Chongyang Sun
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Jianyu Huang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Xufang Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Suwan Pan
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Shuyu He
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Kang Huang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Zhonghua Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Fuqiang Xu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; Center for Brain Science, Wuhan Institute of Physics and Mathematics, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yi Lu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| | - Liping Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
36
|
Hashimoto M, Brito SI, Venner A, Pasqualini AL, Yang TL, Allen D, Fuller PM, Anthony TE. Lateral septum modulates cortical state to tune responsivity to threat stimuli. Cell Rep 2022; 41:111521. [PMID: 36288710 PMCID: PMC9645245 DOI: 10.1016/j.celrep.2022.111521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/17/2022] [Accepted: 09/26/2022] [Indexed: 11/30/2022] Open
Abstract
Sudden unexpected environmental changes capture attention and, when perceived as potentially dangerous, evoke defensive behavioral states. Perturbations of the lateral septum (LS) can produce extreme hyperdefensiveness even to innocuous stimuli, but how this structure influences stimulus-evoked defensive responses and threat perception remains unclear. Here, we show that Crhr2-expressing neurons in mouse LS exhibit phasic activation upon detection of threatening but not rewarding stimuli. Threat-stimulus-driven activity predicts the probability but not vigor or type of defensive behavior evoked. Although necessary for and sufficient to potentiate stimulus-triggered defensive responses, LSCrhr2 neurons do not promote specific behaviors. Rather, their stimulation elicits negative valence and physiological arousal. Moreover, LSCrhr2 activity tracks brain state fluctuations and drives cortical activation and rapid awakening in the absence of threat. Together, our findings suggest that LS directs bottom-up modulation of cortical function to evoke preparatory defensive internal states and selectively enhance responsivity to threat-related stimuli.
Collapse
Affiliation(s)
- Mariko Hashimoto
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Salvador Ignacio Brito
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anne Venner
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda Loren Pasqualini
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Tracy Lulu Yang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - David Allen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Michael Fuller
- Department of Neurology, Beth Israel Deaconess Medical Center and Division of Sleep Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Todd Erryl Anthony
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Departments of Psychiatry and Neurology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Li H, Sung HH, Lau CG. Activation of Somatostatin-Expressing Neurons in the Lateral Septum Improves Stress-Induced Depressive-like Behaviors in Mice. Pharmaceutics 2022; 14:pharmaceutics14102253. [PMID: 36297687 PMCID: PMC9607457 DOI: 10.3390/pharmaceutics14102253] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Depression is a debilitating mood disorder with highly heterogeneous pathogenesis. The limbic system is well-linked to depression. As an important node in the limbic system, the lateral septum (LS) can modulate multiple affective and motivational behaviors. However, the role of LS in depression remains unclear. By using c-Fos expression mapping, we first screened and showed activation of the LS in various depression-related behavioral tests, including the forced swim test (FST), tail suspension test (TST), and sucrose preference test. In the LS, more than 10% of the activated neurons were somatostatin-expressing (SST) neurons. We next developed a microendoscopic calcium imaging method in freely moving mice and revealed that LSSST neural activity increased during mobility in the TST but not open field test. We hypothesize that LSSST neuronal activity is linked to stress and depression. In two mouse models of depression, repeated lipopolysaccharide (LPS) injection and chronic restraint stress (CRS), we showed that LS neuronal activation was suppressed. To examine whether the re-activation of LSSST neurons can be therapeutically beneficial, we optogenetically activated LSSST neurons and produced antidepressant-like effects in LPS-injected mice by increasing TST motility. Moreover, chemogenetic activation of LSSST neurons increased FST struggling in the CRS-exposed mice. Together, these results provide the first evidence of a role for LSSST neurons in regulating depressive-like behaviors in mice and identify them as a potential therapeutic target for neuromodulation-based intervention in depression.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Hyun Hailey Sung
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Correspondence: ; Tel.: +852-3442-4345
| |
Collapse
|
38
|
A hypothalamic dopamine locus for psychostimulant-induced hyperlocomotion in mice. Nat Commun 2022; 13:5944. [PMID: 36209152 PMCID: PMC9547883 DOI: 10.1038/s41467-022-33584-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/22/2022] [Indexed: 11/29/2022] Open
Abstract
The lateral septum (LS) has been implicated in the regulation of locomotion. Nevertheless, the neurons synchronizing LS activity with the brain’s clock in the suprachiasmatic nucleus (SCN) remain unknown. By interrogating the molecular, anatomical and physiological heterogeneity of dopamine neurons of the periventricular nucleus (PeVN; A14 catecholaminergic group), we find that Th+/Dat1+ cells from its anterior subdivision innervate the LS in mice. These dopamine neurons receive dense neuropeptidergic innervation from the SCN. Reciprocal viral tracing in combination with optogenetic stimulation ex vivo identified somatostatin-containing neurons in the LS as preferred synaptic targets of extrahypothalamic A14 efferents. In vivo chemogenetic manipulation of anterior A14 neurons impacted locomotion. Moreover, chemogenetic inhibition of dopamine output from the anterior PeVN normalized amphetamine-induced hyperlocomotion, particularly during sedentary periods. Cumulatively, our findings identify a hypothalamic locus for the diurnal control of locomotion and pinpoint a midbrain-independent cellular target of psychostimulants. The psychostimulant-sensitive neural mechanism linking the circadian clock to locomotion is unknown. Here, hypothalamic A14 neurons are shown to time diurnal activity by entraining the lateral septum, and their activity is shown to be sensitive to amphetamine.
Collapse
|
39
|
Conte R, Zangirolame CMS, Gobbo DR, Pereira LDAS, Panfilio CE, Reginato RD, Maluf LLS, Scerni DA, Céspedes IC. Effects of moderate alcohol consumption on behavior and neural systems of Wistar rats. AN ACAD BRAS CIENC 2022; 94:e20210673. [PMID: 35857964 DOI: 10.1590/0001-3765202220210673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/09/2021] [Indexed: 11/21/2022] Open
Abstract
Chronic alcohol consumption affects various neurotransmitters, especially those implicated in the transitioning to alcohol use disorders (particularly dopaminergic and CRFergic systems). Few studies have investigated moderate alcohol consumption and its harmful consequences. The objective of this work was to analyze behavioral and neurochemical (dopaminergic and CRFergic systems) alterations during chronic moderate alcohol consumption. Twelve male Wistar rats were submitted to an intermittent alcohol ingestion protocol (alcohol group) for four weeks. The control group consisted of six rats. Open Field and Elevated Plus Maze tests were used for analysis of motor and anxiety-like behaviors. Immunohistochemistry analysis was performed in dopaminergic and CRFergic systems. Animals exposed to alcohol consumed moderate doses, chronic and intermittently. Behavioral tests detected fewer fecal boli in the alcohol exposed group, and immunohistochemical analysis indicated fewer dopamine-immunoreactive cells in the ventral tegmental area, and more CRF-immunoreactive cells in the anterior cingulate cortex and dorsolateral septum in this group. Thus we concluded that Wistar rats that consumed moderate doses of alcohol voluntarily and chronically showed a discreet anxiolytic effect in behavior, and a hypodopaminergic and hyperCRFergic neurochemical condition, which together are strong inducers of alcohol consumption predisposing to the development of alcohol use disorder (AUD).
Collapse
Affiliation(s)
- Rafael Conte
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Morfologia e Genética, Rua Botucatu, 740, 1º andar do Edifício Leitão da Cunha, 04023-900 São Paulo, SP, Brazil
| | - Carolline M S Zangirolame
- Universidade Federal de São Paulo/UNIFESP, Instituto de Saúde e Sociedade, Departamento de Biociências, Rua Silva Jardim, 136, Vila Matias, 11015-020 Santos, SP, Brazil
| | - Denise R Gobbo
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Morfologia e Genética, Rua Botucatu, 740, 1º andar do Edifício Leitão da Cunha, 04023-900 São Paulo, SP, Brazil
| | - Laís DA S Pereira
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Morfologia e Genética, Rua Botucatu, 740, 1º andar do Edifício Leitão da Cunha, 04023-900 São Paulo, SP, Brazil
| | - Carlos E Panfilio
- Universidade Municipal de São Caetano do Sul/USCS, Escola da Saúde, Rua Santo Antônio, 50, Centro, 09521-160 São Caetano do Sul, SP, Brazil
| | - Rejane D Reginato
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Morfologia e Genética, Rua Botucatu, 740, 1º andar do Edifício Leitão da Cunha, 04023-900 São Paulo, SP, Brazil
| | - Luciana L S Maluf
- Universidade Federal de São Paulo/UNIFESP, Instituto de Saúde e Sociedade, Departamento de Biociências, Rua Silva Jardim, 136, Vila Matias, 11015-020 Santos, SP, Brazil
| | - Debora A Scerni
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, Rua Pedro De Toledo, 669, Vila Clementino, 04039-032 São Paulo, SP, Brazil
| | - Isabel C Céspedes
- Universidade Federal de São Paulo/UNIFESP, Escola Paulista de Medicina, Departamento de Neurologia e Neurocirurgia, Rua Pedro De Toledo, 669, Vila Clementino, 04039-032 São Paulo, SP, Brazil
| |
Collapse
|
40
|
Yeates DCM, Leavitt D, Sujanthan S, Khan N, Alushaj D, Lee ACH, Ito R. Parallel ventral hippocampus-lateral septum pathways differentially regulate approach-avoidance conflict. Nat Commun 2022; 13:3349. [PMID: 35688838 PMCID: PMC9187740 DOI: 10.1038/s41467-022-31082-0] [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: 08/09/2021] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
The ability to resolve an approach-avoidance conflict is critical to adaptive behavior. The ventral CA3 (vCA3) and CA1 (vCA1) subfields of the ventral hippocampus (vHPC) have been shown to facilitate avoidance and approach behavior, respectively, in the face of motivational conflict, but the neural circuits by which this subfield-specific regulation is implemented is unknown. We demonstrate that two distinct pathways from these subfields to lateral septum (LS) contribute to this divergent control. In Long-Evans rats, chemogenetic inhibition of the vCA3- LS caudodorsal (cd) pathway potentiated approach towards a learned conflict-eliciting stimulus, while inhibition of the vCA1-LS rostroventral (rv) pathway potentiated approach non-specifically. Additionally, vCA3-LScd inhibited animals were less hesitant to explore food during environmental uncertainty, while the vCA1- LSrv inhibited animals took longer to initiate food exploration. These findings suggest that the vHPC influences multiple behavioral systems via differential projections to the LS, which in turn send inhibitory projections to motivational centres of the brain.
Collapse
Affiliation(s)
- Dylan C M Yeates
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
| | - Dallas Leavitt
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
| | - Sajeevan Sujanthan
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
| | - Nisma Khan
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
| | - Denada Alushaj
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
| | - Andy C H Lee
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada
- Rotman Research Institute, Baycrest Centre, Toronto, ON, M6A 2E1, Canada
| | - Rutsuko Ito
- Department of Psychology (Scarborough), University of Toronto, Toronto, ON, M1C 1A4, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada.
| |
Collapse
|
41
|
Steiger A, Farfan J, Fisher N, Heller HC, Fernandez FX, Ruby NF. Reversible Suppression of Fear Memory Recall by Transient Circadian Arrhythmia. Front Integr Neurosci 2022; 16:900620. [PMID: 35694186 PMCID: PMC9184752 DOI: 10.3389/fnint.2022.900620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
We tested the hypothesis that a temporary period of circadian arrhythmia would transiently impair recall of an aversive memory in Siberian hamsters (Phodopus sungorus). Unlike mice or rats, circadian arrhythmia is easily induced in this species by a one-time manipulation of their ambient lighting [i.e., the disruptive phase shift (DPS) protocol]. Hamsters were conditioned to associate footshocks with a shock chamber (context) and with a predictive auditory tone (cue), and then exposed to the DPS protocol. Following DPS, animals either became arrhythmic (ARR), reentrained to the light-dark cycle (ENT), or became arrhythmic for < 14 days before their circadian locomotor rhythms spontaneously recovered and reentrained (ARR-ENT). Tests for contextual memory showed that freezing was decreased 9–10 days post-DPS when both ARR and ARR-ENT groups were arrhythmic. Once ARR-ENT animals reentrained (day 41), however, freezing was elevated back to Pre-DPS levels and did not differ from those observed in ENT hamsters. ENT animals maintained high levels of freezing at both time points, whereas, freezing remained low in ARR hamsters. In contrast to contextual responses, cued responses were unaffected by circadian arrhythmia; all three groups exhibited elevated levels of freezing in response to the tones. The differential impact of circadian arrhythmia on contextual versus cued associative memory suggests that arrhythmia preferentially impacts memory processes that depend on the hippocampus.
Collapse
Affiliation(s)
- Athreya Steiger
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Julia Farfan
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Nathan Fisher
- Department of Biology, Stanford University, Stanford, CA, United States
| | - H. Craig Heller
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Fabian-Xosé Fernandez
- Department of Psychology, University of Arizona College of Science, Tucson, AZ, United States
| | - Norman F. Ruby
- Department of Biology, Stanford University, Stanford, CA, United States
- *Correspondence: Norman F. Ruby,
| |
Collapse
|
42
|
Azevedo EP, Ivan VJ, Friedman JM, Stern SA. Higher-Order Inputs Involved in Appetite Control. Biol Psychiatry 2022; 91:869-878. [PMID: 34593204 PMCID: PMC9704062 DOI: 10.1016/j.biopsych.2021.07.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 01/01/2023]
Abstract
The understanding of the neural control of appetite sheds light on the pathogenesis of eating disorders such as anorexia nervosa and obesity. Both diseases are a result of maladaptive eating behaviors (overeating or undereating) and are associated with life-threatening health problems. The fine regulation of appetite involves genetic, physiological, and environmental factors, which are detected and integrated in the brain by specific neuronal populations. For centuries, the hypothalamus has been the center of attention in the scientific community as a key regulator of appetite. The hypothalamus receives and sends axonal projections to several other brain regions that are important for the integration of sensory and emotional information. These connections ensure that appropriate behavioral decisions are made depending on the individual's emotional state and environment. Thus, the mechanisms by which higher-order brain regions integrate exteroceptive information to coordinate feeding is of great importance. In this review, we will focus on the functional and anatomical projections connecting the hypothalamus to the limbic system and higher-order brain centers in the cortex. We will also address the mechanisms by which specific neuronal populations located in higher-order centers regulate appetite and how maladaptive eating behaviors might arise from altered connections among cortical and subcortical areas with the hypothalamus.
Collapse
Affiliation(s)
- Estefania P Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York.
| | - Violet J Ivan
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, New York; Howard Hughes Medical Institute, New York, New York
| | - Sarah A Stern
- Integrative Neural Circuits and Behavior Research Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida.
| |
Collapse
|
43
|
Hui Y, Zheng X, Zhang H, Li F, Yu G, Li J, Zhang J, Gong X, Guo G. Strategies for Targeting Neural Circuits: How to Manipulate Neurons Using Virus Vehicles. Front Neural Circuits 2022; 16:882366. [PMID: 35571271 PMCID: PMC9099413 DOI: 10.3389/fncir.2022.882366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/07/2022] [Indexed: 01/02/2023] Open
Abstract
Viral strategies are the leading methods for mapping neural circuits. Viral vehicles combined with genetic tools provide the possibility to visualize entire functional neural networks and monitor and manipulate neural circuit functions by high-resolution cell type- and projection-specific targeting. Optogenetics and chemogenetics drive brain research forward by exploring causal relationships among different brain regions. Viral strategies offer a fresh perspective for the analysis of the structure-function relationship of the neural circuitry. In this review, we summarize current and emerging viral strategies for targeting neural circuits and focus on adeno-associated virus (AAV) vectors.
Collapse
Affiliation(s)
- Yuqing Hui
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuefeng Zheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Huijie Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Guangyin Yu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Jifeng Zhang,
| | - Xiaobing Gong
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Xiaobing Gong,
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- *Correspondence: Guoqing Guo,
| |
Collapse
|
44
|
Patel H. The role of the lateral septum in neuropsychiatric disease. J Neurosci Res 2022; 100:1422-1437. [PMID: 35443088 DOI: 10.1002/jnr.25052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 02/22/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022]
Abstract
The lateral septum (LS) is a structure in the midline of the brain that is interconnected with areas associated with stress and feeding. This review highlights the role of the LS in anxiety, depression, and eating disorders and their comorbidity. There is a prevailing view that the LS is anxiolytic. This review finds that the LS is both anxiolytic and anxiogenic. Furthermore, the LS can promote and inhibit feeding. Given these shared roles, the LS represents a common site for the comorbidity of neuropsychiatric disorders, and therefore a potential pharmacological target. This is crucial since currently available treatments are not always effective. Corticotrophin-releasing factor 2 antagonists are potential drugs for the treatment of anxiety and anorexia and require further research. Furthermore, other drugs currently in trials for binge eating, such as alpha-adrenergic agonists, may in fact promote food intake. It is hoped that the advancements in chemo- and optogenetic techniques will allow future studies to profile the specific neural connections of the LS and their function. This information could facilitate our understanding of the underlying mechanisms, and therefore pharmacological targets, of these psychiatric conditions.
Collapse
|
45
|
Somatostatin and Somatostatin-Containing Interneurons—From Plasticity to Pathology. Biomolecules 2022; 12:biom12020312. [PMID: 35204812 PMCID: PMC8869243 DOI: 10.3390/biom12020312] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/27/2022] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the obvious differences in the pathophysiology of distinct neuropsychiatric diseases or neurodegenerative disorders, some of them share some general but pivotal mechanisms, one of which is the disruption of excitation/inhibition balance. Such an imbalance can be generated by changes in the inhibitory system, very often mediated by somatostatin-containing interneurons (SOM-INs). In physiology, this group of inhibitory interneurons, as well as somatostatin itself, profoundly shapes the brain activity, thus influencing the behavior and plasticity; however, the changes in the number, density and activity of SOM-INs or levels of somatostatin are found throughout many neuropsychiatric and neurological conditions, both in patients and animal models. Here, we (1) briefly describe the brain somatostatinergic system, characterizing the neuropeptide somatostatin itself, its receptors and functions, as well the physiology and circuitry of SOM-INs; and (2) summarize the effects of the activity of somatostatin and SOM-INs in both physiological brain processes and pathological brain conditions, focusing primarily on learning-induced plasticity and encompassing selected neuropsychological and neurodegenerative disorders, respectively. The presented data indicate the somatostatinergic-system-mediated inhibition as a substantial factor in the mechanisms of neuroplasticity, often disrupted in a plethora of brain pathologies.
Collapse
|
46
|
Besnard A, Leroy F. Top-down regulation of motivated behaviors via lateral septum sub-circuits. Mol Psychiatry 2022; 27:3119-3128. [PMID: 35581296 PMCID: PMC7613864 DOI: 10.1038/s41380-022-01599-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022]
Abstract
How does cognition regulate innate behaviors? While the cognitive functions of the cortex have been extensively studied, we know much less about how cognition can regulate innate motivated behaviors to fulfill physiological, safety and social needs. Selection of appropriate motivated behaviors depends on external stimuli and past experiences that helps to scale priorities. With its abundant inputs from neocortical and allocortical regions, the lateral septum (LS) is ideally positioned to integrate perception and experience signals in order to regulate the activity of hypothalamic and midbrain nuclei that control motivated behaviors. In addition, LS receives numerous subcortical modulatory inputs, which represent the animal internal states and also participate in this regulation. In this perspective, we argue that LS sub-circuits regulate distinct motivated behaviors by integrating neural activity from neocortical, allocortical and neuromodulatory inputs. In addition, we propose that lateral inhibition between LS sub-circuits may allow the emergence of functional units that orchestrates competing motivated behaviors.
Collapse
Affiliation(s)
| | - Felix Leroy
- Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, Spain.
| |
Collapse
|
47
|
Surget A, Belzung C. Adult hippocampal neurogenesis shapes adaptation and improves stress response: a mechanistic and integrative perspective. Mol Psychiatry 2022; 27:403-421. [PMID: 33990771 PMCID: PMC8960391 DOI: 10.1038/s41380-021-01136-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 02/03/2023]
Abstract
Adult hippocampal neurogenesis (AHN) represents a remarkable form of neuroplasticity that has increasingly been linked to the stress response in recent years. However, the hippocampus does not itself support the expression of the different dimensions of the stress response. Moreover, the main hippocampal functions are essentially preserved under AHN depletion and adult-born immature neurons (abGNs) have no extrahippocampal projections, which questions the mechanisms by which abGNs influence functions supported by brain areas far from the hippocampus. Within this framework, we propose that through its computational influences AHN is pivotal in shaping adaption to environmental demands, underlying its role in stress response. The hippocampus with its high input convergence and output divergence represents a computational hub, ideally positioned in the brain (1) to detect cues and contexts linked to past, current and predicted stressful experiences, and (2) to supervise the expression of the stress response at the cognitive, affective, behavioral, and physiological levels. AHN appears to bias hippocampal computations toward enhanced conjunctive encoding and pattern separation, promoting contextual discrimination and cognitive flexibility, reducing proactive interference and generalization of stressful experiences to safe contexts. These effects result in gating downstream brain areas with more accurate and contextualized information, enabling the different dimensions of the stress response to be more appropriately set with specific contexts. Here, we first provide an integrative perspective of the functional involvement of AHN in the hippocampus and a phenomenological overview of the stress response. We then examine the mechanistic underpinning of the role of AHN in the stress response and describe its potential implications in the different dimensions accompanying this response.
Collapse
Affiliation(s)
- A Surget
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | - C Belzung
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| |
Collapse
|
48
|
Jing W, Zhang T, Liu J, Huang X, Yu Q, Yu H, Zhang Q, Li H, Deng M, Zhu LQ, Du H, Lu Y. A circuit of COCH neurons encodes social-stress-induced anxiety via MTF1 activation of Cacna1h. Cell Rep 2021; 37:110177. [PMID: 34965426 DOI: 10.1016/j.celrep.2021.110177] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 08/20/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
The hippocampus is a temporal lobe structure critical for cognition, such as learning, memory, and attention, as well as emotional responses. Hippocampal dysfunction can lead to persistent anxiety and/or depression. However, how millions of neurons in the hippocampus are molecularly and structurally organized to engage their divergent functions remains unknown. Here, we genetically target a subset of neurons expressing the coagulation factor c homolog (COCH) gene. COCH-expressing neurons or COCH neurons are topographically segregated in the distal region of the ventral CA3 hippocampus and express Mtf1 and Cacna1h. MTF1 activation of Cacna1h transcription in COCH neurons encodes the ability of COCH neurons to burst action potentials and cause social-stress-induced anxiety-like behaviors by synapsing directly with a subset of GABAergic inhibitory neurons in the lateral septum. Together, this study provides a molecular and circuitry-based framework for understanding how COCH neurons in the hippocampus are assembled to engage social behavior.
Collapse
Affiliation(s)
- Wei Jing
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongmei Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Histology and Embryology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Jiaying Liu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xian Huang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Quntao Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongyan Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qingping Zhang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Manfei Deng
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ling-Qiang Zhu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huiyun Du
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
49
|
Mahadevia D, Saha R, Manganaro A, Chuhma N, Ziolkowski-Blake A, Morgan AA, Dumitriu D, Rayport S, Ansorge MS. Dopamine promotes aggression in mice via ventral tegmental area to lateral septum projections. Nat Commun 2021; 12:6796. [PMID: 34815379 PMCID: PMC8610979 DOI: 10.1038/s41467-021-27092-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/02/2021] [Indexed: 11/29/2022] Open
Abstract
Septal-hypothalamic neuronal activity centrally mediates aggressive behavior and dopamine system hyperactivity is associated with elevated aggression. However, the causal role of dopamine in aggression and its target circuit mechanisms are largely unknown. To address this knowledge gap, we studied the modulatory role of the population- and projection-specific dopamine function in a murine model of aggressive behavior. We find that terminal activity of ventral tegmental area (VTA) dopaminergic neurons selectively projecting to the lateral septum (LS) is sufficient for promoting aggression and necessary for establishing baseline aggression. Within the LS, dopamine acts on D2-receptors to inhibit GABAergic neurons, and septal D2-signaling is necessary for VTA dopaminergic activity to promote aggression. Collectively, our data reveal a powerful modulatory influence of dopaminergic synaptic input on LS function and aggression, effectively linking the clinically pertinent hyper-dopaminergic model of aggression with the classic septal-hypothalamic aggression axis.
Collapse
Affiliation(s)
- Darshini Mahadevia
- Department of Psychiatry, Division of Developmental Neuroscience, Columbia University, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Rinki Saha
- Department of Psychiatry, Division of Developmental Neuroscience, Columbia University, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Alessia Manganaro
- Department of Pediatrics, Division for Child and Adolescent Health, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Nao Chuhma
- New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University, New York, NY, 10032, USA
| | | | - Ashlea A Morgan
- Department of Psychiatry, Division of Developmental Neuroscience, Columbia University, New York, NY, 10032, USA
- New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Dani Dumitriu
- Department of Psychiatry, Division of Developmental Neuroscience, Columbia University, New York, NY, 10032, USA
- Department of Pediatrics, Division for Child and Adolescent Health, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Stephen Rayport
- New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Psychiatry, Division of Molecular Therapeutics, Columbia University, New York, NY, 10032, USA
| | - Mark S Ansorge
- Department of Psychiatry, Division of Developmental Neuroscience, Columbia University, New York, NY, 10032, USA.
- New York State Psychiatric Institute, New York, NY, 10032, USA.
| |
Collapse
|
50
|
Putting Together Pieces of the Lateral Septum: Multifaceted Functions and Its Neural Pathways. eNeuro 2021; 8:ENEURO.0315-21.2021. [PMID: 34764187 PMCID: PMC8647703 DOI: 10.1523/eneuro.0315-21.2021] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 01/01/2023] Open
Abstract
The lateral septum (LS) is implicated as a hub that regulates a variety of affects, such as reward, feeding, anxiety, fear, sociability, and memory. However, it remains unclear how the LS, previously treated as a structure of homogeneity, exhibits such multifaceted functions. Emerging evidence suggests that different functions of the LS are mediated largely by its diverse input and output connections. It has also become clear that the LS is a heterogeneous region, where its dorsal and ventral poles play dissociable and often opposing roles. This functional heterogeneity can often be explained by distinct dorsal and ventral hippocampal inputs along the LS dorsoventral axis, as well as antagonizing connections between LS subregions. Similarly, outputs from LS subregions to respective downstream targets, such as hypothalamic, preoptic, and tegmental areas, also account for this functional heterogeneity. In this review, we provide an updated perspective on LS subregion classification, connectivity, and functions. We also identify key questions that have yet to be addressed in the field.
Collapse
|