1
|
Ren S, Wang S, Lv S, Gao J, Mao Y, Liu Y, Xie Q, Zhang T, Zhao L, Shi J. The nociceptive inputs of the paraventricular hypothalamic nucleus in formalin stimulated mice. Neurosci Lett 2024; 841:137948. [PMID: 39179131 DOI: 10.1016/j.neulet.2024.137948] [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: 03/18/2024] [Revised: 08/03/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
The paraventricular hypothalamic nucleus (PVH) is an important neuroendocrine center involved in pain regulation, but the nociceptive afferent routes for the nucleus are still unclear. We examined the profile of PVH receiving injurious information by a combination of retrograde tracing with Fluoro-Gold (FG) and FOS expression induced by formalin stimuli. The result showed that formalin injection induced significantly increased expression of FOS in the PVH, among which oxytocin containing neurons are one neuronal phenotype. Immunofluorescent staining of FG and FOS revealed that double labeled neurons were strikingly distributed in the area 2 of the cingulate cortex (Cg2), the lateral septal nucleus (LS), the periaqueductal gray (PAG), the posterior hypothalamic area (PH), and the lateral parabrachial nucleus (LPB). In the five regions, LPB had the biggest number and the highest ratio of FOS expression in FG labeled neurons, with main subnuclei distribution in the external, superior, dorsal, and central parts. Further immunofluorescent triple staining disclosed that about one third of FG and FOS double labeled neurons in the LPB were immunoreactive for calcitonin gene related peptide (CGRP). In conclusion, the present study demonstrates the nociceptive input profile of the PVH area under inflammatory pain and suggests that neurons in the LPB may play essential roles in transmitting noxious information to the PVH.
Collapse
Affiliation(s)
- Shuting Ren
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Shumin Wang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Siting Lv
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Jiaying Gao
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Yajie Mao
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Yuankun Liu
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Medical School of Yan'an University, Yan'an 716000, China
| | - Qiongyao Xie
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Ting Zhang
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Lin Zhao
- Medical School of Yan'an University, Yan'an 716000, China.
| | - Juan Shi
- Department of Human Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Preclinical School of Medicine, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
2
|
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 2024:101781. [PMID: 39423809 DOI: 10.1016/j.xcrm.2024.101781] [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: 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
|
3
|
Fricker BA, Murugan M, Seifert AW, Kelly AM. Cingulate to septal circuitry facilitates the preference to affiliate with large peer groups. Curr Biol 2024; 34:4452-4463.e4. [PMID: 39265570 PMCID: PMC11486304 DOI: 10.1016/j.cub.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Despite the prevalence of large-group living across the animal kingdom, no studies have examined the neural mechanisms that make group living possible. Spiny mice, Acomys, have evolved to live in large groups and exhibit a preference to affiliate with large over small groups. Here, we determine the neural circuitry that facilitates the drive to affiliate with large groups. We first identify an anterior cingulate cortex (ACC) to lateral septum (LS) circuit that is more responsive to large than small groups of novel same-sex peers. Using chemogenetics, we then demonstrate that this circuit is necessary for both male and female group investigation preferences but only males' preference to affiliate with larger peer groups. Furthermore, inhibition of the ACC-LS circuit specifically impairs social, but not nonsocial, affiliative grouping preferences. These findings reveal a key circuit for the regulation of mammalian peer group affiliation.
Collapse
Affiliation(s)
- Brandon A Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Malavika Murugan
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 211 Thomas Hunt Morgan Building, Lexington, KY 40506, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA.
| |
Collapse
|
4
|
Chen G, Lai S, Jiang S, Li F, Sun K, Wu X, Zhou K, Liu Y, Deng X, Chen Z, Xu F, Xu Y, Wang K, Cao G, Xu F, Bi GQ, Zhu Y. Cellular and circuit architecture of the lateral septum for reward processing. Neuron 2024; 112:2783-2798.e9. [PMID: 38959892 DOI: 10.1016/j.neuron.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
The lateral septum (LS) is composed of heterogeneous cell types that are important for various motivated behaviors. However, the transcriptional profiles, spatial arrangement, function, and connectivity of these cell types have not been systematically studied. Using single-nucleus RNA sequencing, we delineated diverse genetically defined cell types in the LS that play distinct roles in reward processing. Notably, we found that estrogen receptor 1 (Esr1)-expressing neurons in the ventral LS (LSEsr1) are key drivers of reward seeking via projections to the ventral tegmental area, and these neurons play an essential role in methamphetamine (METH) reward and METH-seeking behavior. Extended exposure to METH increases the excitability of LSEsr1 neurons by upregulating hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, thereby contributing to METH-induced locomotor sensitization. These insights not only elucidate the intricate molecular, circuit, and functional architecture of the septal region in reward processing but also reveal a neural pathway critical for METH reward and behavioral sensitization.
Collapse
Affiliation(s)
- Gaowei Chen
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shishi Lai
- Yunnan University School of Medicine, Yunnan University, Kunming 650091, China; Southwest United Graduate School, Kunming 650092, China
| | - Shaolei Jiang
- University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Fengling Li
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kaige Sun
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shandong Normal University, Jinan 250014, China
| | - Xiaocong Wu
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Kuikui Zhou
- University of Health and Rehabilitation Sciences, Qingdao 266000, China
| | - Yutong Liu
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaofei Deng
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zijun Chen
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fang Xu
- Interdisciplinary Center for Brain Information, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yu Xu
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming 650091, China
| | - Kunhua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming 650091, China
| | - Gang Cao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fuqiang Xu
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guo-Qiang Bi
- Interdisciplinary Center for Brain Information, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shenzhen Neher Neural Plasticity Laboratory, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
5
|
Gomes-Ribeiro J, Martins J, Sereno J, Deslauriers-Gauthier S, Summavielle T, Coelho JE, Remondes M, Castelo-Branco M, Lopes LV. Mapping functional traces of opioid memories in the rat brain. Brain Commun 2024; 6:fcae281. [PMID: 39229487 PMCID: PMC11369824 DOI: 10.1093/braincomms/fcae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 07/04/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
Addiction to psychoactive substances is a maladaptive learned behaviour. Contexts surrounding drug use integrate this aberrant mnemonic process and hold strong relapse-triggering ability. Here, we asked where context and salience might be concurrently represented in the brain during retrieval of drug-context paired associations. For this, we developed a morphine-conditioned place preference protocol that allows contextual stimuli presentation inside a magnetic resonance imaging scanner and investigated differences in activity and connectivity at context recall. We found context-specific responses to stimulus onset in multiple brain regions, namely, limbic, sensory and striatal. Differences in functional interconnectivity were found among amygdala, lateral habenula, and lateral septum. We also investigated alterations to resting-state functional connectivity and found increased centrality of the lateral septum in a proposed limbic network, as well as increased functional connectivity of the lateral habenula and hippocampal 'cornu ammonis' 1 region, after a protocol of associative drug-context. Finally, we found that pre- conditioned place preference resting-state connectivity of the lateral habenula and amygdala was predictive of inter-individual conditioned place preference score differences. Overall, our findings show that drug and saline-paired contexts establish distinct memory traces in overlapping functional brain microcircuits and that intrinsic connectivity of the habenula, septum, and amygdala likely underlies the individual maladaptive contextual learning to opioid exposure. We have identified functional maps of acquisition and retrieval of drug-related memory that may support the relapse-triggering ability of opioid-associated sensory and contextual cues. These findings may clarify the inter-individual sensitivity and vulnerability seen in addiction to opioids found in humans.
Collapse
Affiliation(s)
- Joana Gomes-Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - José Sereno
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- CQC, Chemistry Department, University of Coimbra, 3004-535 Coimbra, Portugal
| | | | - Teresa Summavielle
- Addiction Biology Group, i3S- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- ESS, Polytechnic of Porto, 4200-072 Porto, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Miguel Remondes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Faculdade de Medicina Veterinária, Universidade Lusófona, 1749-024 Lisboa, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Luísa V Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Raudales R, Kim G, Kelly SM, Hatfield J, Guan W, Zhao S, Paul A, Qian Y, Li B, Huang ZJ. Specific and comprehensive genetic targeting reveals brain-wide distribution and synaptic input patterns of GABAergic axo-axonic interneurons. eLife 2024; 13:RP93481. [PMID: 39012795 PMCID: PMC11251723 DOI: 10.7554/elife.93481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024] Open
Abstract
Axo-axonic cells (AACs), also called chandelier cells (ChCs) in the cerebral cortex, are the most distinctive type of GABAergic interneurons described in the neocortex, hippocampus, and basolateral amygdala (BLA). AACs selectively innervate glutamatergic projection neurons (PNs) at their axon initial segment (AIS), thus may exert decisive control over PN spiking and regulate PN functional ensembles. However, the brain-wide distribution, synaptic connectivity, and circuit function of AACs remain poorly understood, largely due to the lack of specific and reliable experimental tools. Here, we have established an intersectional genetic strategy that achieves specific and comprehensive targeting of AACs throughout the mouse brain based on their lineage (Nkx2.1) and molecular (Unc5b, Pthlh) markers. We discovered that AACs are deployed across essentially all the pallium-derived brain structures, including not only the dorsal pallium-derived neocortex and medial pallium-derived hippocampal formation, but also the lateral pallium-derived claustrum-insular complex, and the ventral pallium-derived extended amygdaloid complex and olfactory centers. AACs are also abundant in anterior olfactory nucleus, taenia tecta, and lateral septum. AACs show characteristic variations in density across neocortical areas and layers and across subregions of the hippocampal formation. Neocortical AACs comprise multiple laminar subtypes with distinct dendritic and axonal arborization patterns. Retrograde monosynaptic tracing from AACs across neocortical, hippocampal, and BLA regions reveal shared as well as distinct patterns of synaptic input. Specific and comprehensive targeting of AACs facilitates the study of their developmental genetic program and circuit function across brain structures, providing a ground truth platform for understanding the conservation and variation of a bona fide cell type across brain regions and species.
Collapse
Affiliation(s)
- Ricardo Raudales
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Program in Neurobiology, Stony Brook UniversityStony BrookUnited States
| | - Gukhan Kim
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Sean M Kelly
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Program in Neurobiology, Stony Brook UniversityStony BrookUnited States
| | - Joshua Hatfield
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Department of Neurobiology, Duke UniversityDurhamUnited States
| | - Wuqiang Guan
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
| | - Shengli Zhao
- Department of Neurobiology, Duke UniversityDurhamUnited States
| | - Anirban Paul
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Department of Neural and Behavioral Sciences, Penn State College of MedicineHersheyUnited States
| | - Yongjun Qian
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Department of Neurobiology, Duke UniversityDurhamUnited States
| | - Bo Li
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Department of Neurobiology, Duke UniversityDurhamUnited States
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
| | - Z Josh Huang
- Cold Spring Harbor LaboratoryCold Spring HarborUnited States
- Department of Neurobiology, Duke UniversityDurhamUnited States
- Department of Biomedical Engineering, Duke UniversityDurhamUnited States
| |
Collapse
|
9
|
Zhang YD, Shi DD, Wang Z. Neurobiology of Obsessive-Compulsive Disorder from Genes to Circuits: Insights from Animal Models. Neurosci Bull 2024:10.1007/s12264-024-01252-9. [PMID: 38982026 DOI: 10.1007/s12264-024-01252-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/27/2024] [Indexed: 07/11/2024] Open
Abstract
Obsessive-compulsive disorder (OCD) is a chronic, severe psychiatric disorder that has been ranked by the World Health Organization as one of the leading causes of illness-related disability, and first-line interventions are limited in efficacy and have side-effect issues. However, the exact pathophysiology underlying this complex, heterogeneous disorder remains unknown. This scenario is now rapidly changing due to the advancement of powerful technologies that can be used to verify the function of the specific gene and dissect the neural circuits underlying the neurobiology of OCD in rodents. Genetic and circuit-specific manipulation in rodents has provided important insights into the neurobiology of OCD by identifying the molecular, cellular, and circuit events that induce OCD-like behaviors. This review will highlight recent progress specifically toward classic genetic animal models and advanced neural circuit findings, which provide theoretical evidence for targeted intervention on specific molecular, cellular, and neural circuit events.
Collapse
Affiliation(s)
- Ying-Dan Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Dong-Dong Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
- Shanghai Intelligent Psychological Evaluation and Intervention Engineering Technology Research Center, Shanghai, 200030, China.
| |
Collapse
|
10
|
Peng S, Yang X, Meng S, Liu F, Lv Y, Yang H, Kong Y, Xie W, Li M. Dual circuits originating from the ventral hippocampus independently facilitate affective empathy. Cell Rep 2024; 43:114277. [PMID: 38805397 DOI: 10.1016/j.celrep.2024.114277] [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: 07/26/2023] [Revised: 03/24/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024] Open
Abstract
Affective empathy enables social mammals to learn and transfer emotion to conspecifics, but an understanding of the neural circuitry and genetics underlying affective empathy is still very limited. Here, using the naive observational fear between cagemates as a paradigm similar to human affective empathy and chemo/optogenetic neuroactivity manipulation in mouse brain, we investigate the roles of multiple brain regions in mouse affective empathy. Remarkably, two neural circuits originating from the ventral hippocampus, previously unknown to function in empathy, are revealed to regulate naive observational fear. One is from ventral hippocampal pyramidal neurons to lateral septum GABAergic neurons, and the other is from ventral hippocampus pyramidal neurons to nucleus accumbens dopamine-receptor-expressing neurons. Furthermore, we identify the naive observational-fear-encoding neurons in the ventral hippocampus. Our findings highlight the potentially diverse regulatory pathways of empathy in social animals, shedding light on the mechanisms underlying empathy circuity and its disorders.
Collapse
Affiliation(s)
- Siqi Peng
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Xiuqi Yang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Sibie Meng
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Fuyuan Liu
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China
| | - Yaochen Lv
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Huiquan Yang
- School of Medicine, Southeast University, Nanjing 210096, China
| | - Youyong Kong
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China
| | - Wei Xie
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China; Jiangsu Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Moyi Li
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China; Jiangsu Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
11
|
van Velthoven CTJ, Gao Y, Kunst M, Lee C, McMillen D, Chakka AB, Casper T, Clark M, Chakrabarty R, Daniel S, Dolbeare T, Ferrer R, Gloe J, Goldy J, Guzman J, Halterman C, Ho W, Huang M, James K, Nguy B, Pham T, Ronellenfitch K, Thomas ED, Torkelson A, Pagan CM, Kruse L, Dee N, Ng L, Waters J, Smith KA, Tasic B, Yao Z, Zeng H. The transcriptomic and spatial organization of telencephalic GABAergic neuronal types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599583. [PMID: 38948843 PMCID: PMC11212977 DOI: 10.1101/2024.06.18.599583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The telencephalon of the mammalian brain comprises multiple regions and circuit pathways that play adaptive and integrative roles in a variety of brain functions. There is a wide array of GABAergic neurons in the telencephalon; they play a multitude of circuit functions, and dysfunction of these neurons has been implicated in diverse brain disorders. In this study, we conducted a systematic and in-depth analysis of the transcriptomic and spatial organization of GABAergic neuronal types in all regions of the mouse telencephalon and their developmental origins. This was accomplished by utilizing 611,423 single-cell transcriptomes from the comprehensive and high-resolution transcriptomic and spatial cell type atlas for the adult whole mouse brain we have generated, supplemented with an additional single-cell RNA-sequencing dataset containing 99,438 high-quality single-cell transcriptomes collected from the pre- and postnatal developing mouse brain. We present a hierarchically organized adult telencephalic GABAergic neuronal cell type taxonomy of 7 classes, 52 subclasses, 284 supertypes, and 1,051 clusters, as well as a corresponding developmental taxonomy of 450 clusters across different ages. Detailed charting efforts reveal extraordinary complexity where relationships among cell types reflect both spatial locations and developmental origins. Transcriptomically and developmentally related cell types can often be found in distant and diverse brain regions indicating that long-distance migration and dispersion is a common characteristic of nearly all classes of telencephalic GABAergic neurons. Additionally, we find various spatial dimensions of both discrete and continuous variations among related cell types that are correlated with gene expression gradients. Lastly, we find that cortical, striatal and some pallidal GABAergic neurons undergo extensive postnatal diversification, whereas septal and most pallidal GABAergic neuronal types emerge simultaneously during the embryonic stage with limited postnatal diversification. Overall, the telencephalic GABAergic cell type taxonomy can serve as a foundational reference for molecular, structural and functional studies of cell types and circuits by the entire community.
Collapse
Affiliation(s)
| | - Yuan Gao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Scott Daniel
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mike Huang
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Beagan Nguy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Lauren Kruse
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
12
|
Puska G, Szendi V, Dobolyi A. Lateral septum as a possible regulatory center of maternal behaviors. Neurosci Biobehav Rev 2024; 161:105683. [PMID: 38649125 DOI: 10.1016/j.neubiorev.2024.105683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
The lateral septum (LS) is involved in controlling anxiety, aggression, feeding, and other motivated behaviors. Lesion studies have also implicated the LS in various forms of caring behaviors. Recently, novel experimental tools have provided a more detailed insight into the function of the LS, including the specific role of distinct cell types and their neuronal connections in behavioral regulations, in which the LS participates. This article discusses the regulation of different types of maternal behavioral alterations using the distributions of established maternal hormones such as prolactin, estrogens, and the neuropeptide oxytocin. It also considers the distribution of neurons activated in mothers in response to pups and other maternal activities, as well as gene expressional alterations in the maternal LS. Finally, this paper proposes further research directions to keep up with the rapidly developing knowledge on maternal behavioral control in other maternal brain regions.
Collapse
Affiliation(s)
- Gina Puska
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Department of Zoology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Vivien Szendi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Arpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
13
|
Shemesh Y, Benjamin A, Shoshani-Haye K, Yizhar O, Chen A. Studying dominance and aggression requires ethologically relevant paradigms. Curr Opin Neurobiol 2024; 86:102879. [PMID: 38692167 DOI: 10.1016/j.conb.2024.102879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/13/2024] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
Although aggression is associated with several psychiatric disorders, there is no effective treatment nor a rigorous definition for "pathological aggression". Mice make a valuable model for studying aggression. They have a dynamic social structure that depends on the habitat and includes reciprocal interactions between the mice's aggression levels, social dominance hierarchy (SDH), and resource allocation. Nevertheless, the classical behavioral tests for territorial aggression and SDH in mice are reductive and have limited ethological and translational relevance. Recent work has explored the use of semi-natural environments to simultaneously study dominance-related behaviors, resource allocation, and aggressive behavior. Semi-natural setups allow experimental control of the environment combined with manipulations of neural activity. We argue that these setups can help bridge the translational gap in aggression research toward discovering neuronal mechanisms underlying maladaptive aggression.
Collapse
Affiliation(s)
- Yair Shemesh
- Department of Brain Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Asaf Benjamin
- Department of Brain Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel. https://twitter.com/AsafBenj
| | - Keren Shoshani-Haye
- Department of Brain Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ofer Yizhar
- Department of Brain Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel. https://twitter.com/OferYizhar
| | - Alon Chen
- Department of Brain Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
14
|
Rigney N, Campos-Lira E, Kirchner MK, Wei W, Belkasim S, Beaumont R, Singh S, Suarez SG, Hartswick D, Stern JE, de Vries GJ, Petrulis A. A vasopressin circuit that modulates mouse social investigation and anxiety-like behavior in a sex-specific manner. Proc Natl Acad Sci U S A 2024; 121:e2319641121. [PMID: 38709918 PMCID: PMC11098102 DOI: 10.1073/pnas.2319641121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
One of the largest sex differences in brain neurochemistry is the expression of the neuropeptide arginine vasopressin (AVP) within the vertebrate brain, with males having more AVP cells in the bed nucleus of the stria terminalis (BNST) than females. Despite the long-standing implication of AVP in social and anxiety-like behaviors, the circuitry underlying AVP's control of these behaviors is still not well defined. Using optogenetic approaches, we show that inhibiting AVP BNST cells reduces social investigation in males, but not in females, whereas stimulating these cells increases social investigation in both sexes, but more so in males. These cells may facilitate male social investigation through their projections to the lateral septum (LS), an area with the highest density of sexually differentiated AVP innervation in the brain, as optogenetic stimulation of BNST AVP → LS increased social investigation and anxiety-like behavior in males but not in females; the same stimulation also caused a biphasic response of LS cells ex vivo. Blocking the vasopressin 1a receptor (V1aR) in the LS eliminated all these responses. Together, these findings establish a sexually differentiated role for BNST AVP cells in the control of social investigation and anxiety-like behavior, likely mediated by their projections to the LS.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | - Elba Campos-Lira
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | | | - Wei Wei
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | - Selma Belkasim
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | - Rachael Beaumont
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | - Sumeet Singh
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | | | - Delenn Hartswick
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | - Javier E. Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| | | | - Aras Petrulis
- Neuroscience Institute, Georgia State University, Atlanta, GA30302
| |
Collapse
|
15
|
Liu YJ, Wang Y, Wu JW, Zhou J, Song BL, Jiang Y, Li LF. GABAergic synapses from the ventral lateral septum to the paraventricular nucleus of hypothalamus modulate anxiety. Front Neurosci 2024; 18:1337207. [PMID: 38567287 PMCID: PMC10985145 DOI: 10.3389/fnins.2024.1337207] [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: 11/12/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
Emotional disorders, such as anxiety and depression, represent a major societal problem; however, the underlying neurological mechanism remains unknown. The ventral lateral septum (LSv) is implicated in regulating processes related to mood and motivation. In this study, we found that LSv GABAergic neurons were significantly activated in mice experiencing chronic social defeat stress (CSDS) after exposure to a social stressor. We then controlled LSv GABAergic neuron activity using a chemogenetic approach. The results showed that although manipulation of LSv GABAergic neurons had little effect on anxiety-like behavioral performances, the activation of LSv GABAergic neurons during CSDS worsened social anxiety during a social interaction (SI) test. Moreover, LSv GABAergic neurons showed strong projections to the paraventricular nucleus (PVN) of the hypothalamus, which is a central hub for stress reactions. Remarkably, while activation of GABAergic LSv-PVN projections induced social anxiety under basal conditions, activation of this pathway during CSDS alleviated social anxiety during the SI test. On the other hand, the chemogenetic manipulation of LSv GABAergic neurons or LSvGABA-PVN projections had no significant effect on despair-like behavioral performance in the tail suspension test. Overall, LS GABAergic neurons, particularly the LSv GABAergic-PVN circuit, has a regulatory role in pathological anxiety and is thus a potential therapeutic target for the treatment of emotional disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lai-Fu Li
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, China
| |
Collapse
|
16
|
Gonye EC, Dagli AV, Kumar NN, Clements RT, Xu W, Bayliss DA. Expression of endogenous epitope-tagged GPR4 in the mouse brain. eNeuro 2024; 11:ENEURO.0002-24.2024. [PMID: 38408869 DOI: 10.1523/eneuro.0002-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 02/28/2024] Open
Abstract
GPR4 is a proton-sensing G protein-coupled receptor implicated in many peripheral and central physiological processes. GPR4 expression has previously been assessed only via detection of the cognate transcript or indirectly, by use of fluorescent reporters. In this work, CRISPR/Cas9 knock-in technology was used to encode a hemagglutinin (HA) epitope tag within the endogenous locus of Gpr4 and visualize GPR4-HA in the mouse central nervous system using a specific, well characterized HA antibody; GPR4 expression was further verified by complementary Gpr4 mRNA detection. HA immunoreactivity was found in a limited set of brain regions, including in the retrotrapezoid nucleus (RTN), serotonergic raphe nuclei, medial habenula, lateral septum, and several thalamic nuclei. GPR4 expression was not restricted to cells of a specific neurochemical identity as it was observed in excitatory, inhibitory, and aminergic neuronal cell groups. HA immunoreactivity was not detected in brain vascular endothelium, despite clear expression of Gpr4 mRNA in endothelial cells. In the RTN, GPR4 expression was detected at the soma and in proximal dendrites along blood vessels and the ventral surface of the brainstem; HA immunoreactivity was not detected in RTN projections to two known target regions. This localization of GPR4 protein in mouse brain neurons corroborates putative sites of expression where its function has been previously implicated (e.g., CO2-regulated breathing by RTN), and provides a guide for where GPR4 could contribute to other CO2/H+ modulated brain functions. Finally, GPR4-HA animals provide a useful reagent for further study of GPR4 in other physiological processes outside of the brain.Significance Statement GPR4 is a proton-sensing G-protein coupled receptor whose expression is necessary for a number of diverse physiological processes including acid-base sensing in the kidney, immune function, and cancer progression. In the brain, GPR4 has been implicated in the hypercapnic ventilatory response mediated by brainstem neurons. While knockout studies in animals have clearly demonstrated its necessity for normal physiology, descriptions of GPR4 expression have been limited due to a lack of specific antibodies for use in mouse models. In this paper, we implemented a CRISPR/Cas9 knock-in approach to incorporate the coding sequence for a small epitope tag into the locus of GPR4. Using these mice, we were able to describe GPR4 protein expression directly for the first time.
Collapse
Affiliation(s)
- Elizabeth C Gonye
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Alexandra V Dagli
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Natasha N Kumar
- University of New South Wales Sydney, School of Biomedical Sciences, New South Wales, Australia
| | - Rachel T Clements
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| | - Wenhao Xu
- University of Virginia, Genetically Engineered Mouse Model Core, Charlottesville, VA, USA
| | - Douglas A Bayliss
- University of Virginia, Department of Pharmacology, Charlottesville, VA, USA
| |
Collapse
|
17
|
Crespi BJ. The Roots of Science, Technology, Engineering, and Mathematics: What Are the Evolutionary and Neural Bases of Human Mathematics and Technology? BRAIN, BEHAVIOR AND EVOLUTION 2024; 99:1-12. [PMID: 38368855 DOI: 10.1159/000537908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Neural exaptations represent descent via transitions to novel neural functions. A primary transition in human cognitive and neural evolution was from a predominantly socially oriented primate brain to a brain that also instantiates and subserves science, technology, and engineering, all of which depend on mathematics. Upon what neural substrates and upon what evolved cognitive mechanisms did human capacities for science, technology, engineering, and mathematics (STEM), and especially its mathematical underpinnings, emerge? Previous theory focuses on roles for tools, language, and arithmetic in the cognitive origins of STEM, but none of these factors appears sufficient to support the transition. METHODS In this article, I describe and evaluate a novel hypothesis for the neural origins and substrates of STEM-based cognition: that they are based in human kinship systems and human maximizing of inclusive fitness. RESULTS The main evidence for this hypothesis is threefold. First, as demonstrated by anthropologists, human kinship systems exhibit complex mathematical and geometrical structures that function under sets of explicit rules, and such systems and rules pervade and organize all human cultures. Second, human kinship underlies the core algebraic mechanism of evolution, maximization of inclusive fitness, quantified as personal reproduction plus the sum of all effects on reproduction of others, each multiplied by their coefficient of relatedness to self. This is the only "natural" equation expected to be represented in the human brain. Third, functional imaging studies show that kinship-related cognition activates frontal-parietal regions that are also activated in STEM-related tasks. In turn, the decision-making that integrates kinship levels with costs and benefits from alternative behaviors has recently been shown to recruit the lateral septum, a hub region that combines internal (from the prefrontal cortex, amygdala, and other regions) and external information relevant to social behavior, using a dedicated subsystem of neurons specific to kinship. CONCLUSIONS Taken together, these lines of evidence suggest that kinship systems and kin-associated behaviors may represent exaptations for the origin of human STEM.
Collapse
Affiliation(s)
- Bernard J Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
18
|
Denney KA, Wu MV, Sun SED, Moon S, Tollkuhn J. Comparative analysis of gonadal hormone receptor expression in the postnatal house mouse, meadow vole, and prairie vole brain. Horm Behav 2024; 158:105463. [PMID: 37995608 PMCID: PMC11145901 DOI: 10.1016/j.yhbeh.2023.105463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
The socially monogamous prairie vole (Microtus ochrogaster) and promiscuous meadow vole (Microtus pennsylvanicus) are closely related, but only prairie voles display long-lasting pair bonds, biparental care, and selective aggression towards unfamiliar individuals after pair bonding. These social behaviors in mammals are largely mediated by steroid hormone signaling in the social behavior network (SBN) of the brain. Hormone receptors are reproducible markers of sex differences that can provide more information than anatomy alone and can even be at odds with anatomical dimorphisms. We reasoned that behaviors associated with social monogamy in prairie voles may emerge in part from unique expression patterns of steroid hormone receptors in this species, and that these expression patterns would be more similar across males and females in prairie than in meadow voles or the laboratory mouse. To obtain insight into steroid hormone signaling in the developing prairie vole brain, we assessed expression of estrogen receptor alpha (Esr1), estrogen receptor beta (Esr2), and androgen receptor (Ar) within the SBN, using in situ hybridization at postnatal day 14 in mice, meadow, and prairie voles. We found species-specific patterns of hormone receptor expression in the hippocampus and ventromedial hypothalamus, as well as species differences in the sex bias of these markers in the principal nucleus of the bed nucleus of the stria terminalis. These findings suggest the observed differences in gonadal hormone receptor expression may underlie species differences in the display of social behaviors.
Collapse
Affiliation(s)
- Katherine A Denney
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA; Program in Neuroscience, Stony Brook University, Stony Brook, NY 11790, USA
| | - Melody V Wu
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Simón E D Sun
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Soyoun Moon
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
19
|
Hasunuma K, Murakawa T, Takenawa S, Mitsui K, Hatsukano T, Sano K, Nakata M, Ogawa S. Estrogen Receptor β in the Lateral Septum Mediates Estrogen Regulation of Social Anxiety-like Behavior in Male Mice. Neuroscience 2024; 537:126-140. [PMID: 38042251 DOI: 10.1016/j.neuroscience.2023.11.019] [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: 10/01/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 12/04/2023]
Abstract
17β-estradiol (E2) regulates various forms of social behavior through the activation of two types of estrogen receptors, ERα and ERβ. The lateral septum (LS) is thought to be one of the potential target sites of E2, but the role played by ERα and ERβ in this brain area remains largely unknown. In the present study, we first analyzed the distribution of ERα and ERβ with double fluorescent immunohistochemistry in a transgenic mouse line in which red fluorescent protein (RFP) signal has been a reliable marker of ERβ expression. The overall number of ERβ-RFP-expressing cells was significantly higher (about 2.5 times) compared to ERα-expressing cells. The distribution of the two types of ERs was different, with co-expression only seen in about 1.2% of total ER-positive cells. Given these distinctive distribution patterns, we examined the behavioral effects of site-specific knockdown of each ER using viral vector-mediated small interference RNA (siRNA) techniques in male mice. We found ERβ-specific behavioral alterations during a social interaction test, suggesting involvement of ERβ-expressing LS neurons in the regulation of social anxiety and social interest. Further, we investigated the neuronal projections of ERα- and ERβ-expressing LS cells by injecting an anterograde viral tracer in ERα-Cre and ERβ-iCre mice. Dense expression of green fluorescence protein (GFP) in synaptic terminals was observed in ERβ-iCre mice in areas known to be related to the modulation of anxiety. These findings collectively suggest that ERβ expressed in the LS plays a major role in the estrogenic control of social anxiety-like behavior.
Collapse
Affiliation(s)
- Kansuke Hasunuma
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Tomoaki Murakawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Satoshi Takenawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Koshiro Mitsui
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Tetsu Hatsukano
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Kazuhiro Sano
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Mariko Nakata
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba 305-8577, Japan.
| |
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
|
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] [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
|
22
|
Song BL, Zhou J, Jiang Y, Li LF, Liu YJ. Dopamine D2 receptor within the intermediate region of the lateral septum modulate social hierarchy in male mice. Neuropharmacology 2023; 241:109735. [PMID: 37788799 DOI: 10.1016/j.neuropharm.2023.109735] [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: 07/05/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
The dopamine (DA) system has long been involved in social hierarchies; however, the specific mechanisms have not been elucidated. The lateral septum (LS) is a limbic brain structure that regulates various emotional, motivational, and social behaviors. DA receptors are abundantly expressed in the LS, modulating its functions. In this study, we evaluated the functions of DA receptors within different subregions of the LS in social dominance using a confrontation tube test in male mice. The results showed that mice living in social groups formed linear dominance hierarchies after a few days of cohousing, and the subordinates showed increased anxiety. Fos expressions was elevated in the entire LS after a confrontation tube test in the subordinates. However, DA neurons were more activated in the dominates within the ventral tegmental area and the dorsal raphe nucleus. Quantitative real-time polymerase chain reaction results showed that D2 receptor (D2R) within the intermediate region of the LS (LSi) were elevated in the subordinate. In the following pharmacological studies, we found simultaneous D2R activation in the dominants and D2R inhibition in the subordinates switched the original dominant-subordinate relationship. The aforementioned results suggested that D2R within the LSi plays an important role in social dominance in male mice. These findings improve our understanding of the neural mechanisms underlying the social hierarchy, which is closely related to our social life and happiness.
Collapse
Affiliation(s)
- Bai-Lin Song
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Jie Zhou
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Yi Jiang
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Lai-Fu Li
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China.
| | - Ying-Juan Liu
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China.
| |
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: 1.0] [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
|
Xie Y, Reid CM, Granados AA, Garcia MT, Dale-Huang F, Hanson SM, Mancia W, Liu J, Adam M, Mosto O, Pisco AO, Alvarez-Buylla A, Harwell CC. Developmental origin and local signals cooperate to determine septal astrocyte identity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.08.561428. [PMID: 37873089 PMCID: PMC10592657 DOI: 10.1101/2023.10.08.561428] [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
Astrocyte specification during development is influenced by both intrinsic and extrinsic factors, but the precise contribution of each remains poorly understood. Here we show that septal astrocytes from Nkx2.1 and Zic4 expressing progenitor zones are allocated into non-overlapping domains of the medial (MS) and lateral septal nuclei (LS) respectively. Astrocytes in these areas exhibit distinctive molecular and morphological features tailored to the unique cellular and synaptic circuit environment of each nucleus. Using single-nucleus (sn) RNA sequencing, we trace the developmental trajectories of cells in the septum and find that neurons and astrocytes undergo region and developmental stage-specific local cell-cell interactions. We show that expression of the classic morphogens Sonic hedgehog (Shh) and Fibroblast growth factors (Fgfs) by MS and LS neurons respectively, functions to promote the molecular specification of local astrocytes in each region. Finally, using heterotopic cell transplantation, we show that both morphological and molecular specifications of septal astrocytes are highly dependent on the local microenvironment, regardless of developmental origins. Our data highlights the complex interplay between intrinsic and extrinsic factors shaping astrocyte identities and illustrates the importance of the local environment in determining astrocyte functional specialization.
Collapse
Affiliation(s)
- Yajun Xie
- Department of Neurology, University of California, San Francisco, CA
- 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, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
- Department of Neurobiology, Harvard Medical School, Boston, MA
- Ph.D. Program in Neuroscience, Harvard University, Boston, MA
| | | | - Miguel Turrero Garcia
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Fiona Dale-Huang
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Sarah M. Hanson
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Walter Mancia
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Jonathan Liu
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
| | - Manal Adam
- Department of Neurology, University of California, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, San Francisco, CA
| | - Olivia Mosto
- Department of Neurobiology, Harvard Medical School, Boston, MA
| | | | - Arturo Alvarez-Buylla
- Department of Neurology, University of California, San Francisco, CA
- Department of Neurological Surgery, University of California, San Francisco, CA
| | - Corey C. Harwell
- Department of Neurology, University of California, San Francisco, CA
- 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
|
Fricker BA, Ho D, Seifert AW, Kelly AM. Biased brain and behavioral responses towards kin in males of a communally breeding species. Sci Rep 2023; 13:17040. [PMID: 37813903 PMCID: PMC10562393 DOI: 10.1038/s41598-023-44257-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
In complex social environments, individuals may interact with not only novel and familiar conspecifics but also kin and non-kin. The ability to distinguish between conspecific identities is crucial for most animals, yet how the brain processes conspecific type and how animals may alter behavior accordingly is not well known. We examined whether the communally breeding spiny mouse (Acomys cahirinus) responds differently to conspecifics that vary in novelty and kinship. In a group interaction test, we found that males can distinguish novel kin from novel non-kin, and preferentially spend time with novel kin over familiar kin and novel non-kin. To determine whether kinship and novelty status are differentially represented in the brain, we conducted immediate early gene tests, which revealed the dorsal, but not ventral, lateral septum differentially processes kinship. Neither region differentially processes social novelty. Further, males did not exhibit differences in prosocial behavior toward novel and familiar conspecifics but exhibited more prosocial behavior with novel kin than novel non-kin. These results suggest that communally breeding species may have evolved specialized neural circuitry to facilitate a bias to be more affiliative with kin, regardless of whether they are novel or familiar, potentially to promote prosocial behaviors, thereby facilitating group cohesion.
Collapse
Affiliation(s)
- Brandon A Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - Deborah Ho
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 101 Morgan Building, Lexington, KY, 40506, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
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
|
27
|
Müller-Komorowska D, Kuru B, Beck H, Braganza O. Phase information is conserved in sparse, synchronous population-rate-codes via phase-to-rate recoding. Nat Commun 2023; 14:6106. [PMID: 37777512 PMCID: PMC10543394 DOI: 10.1038/s41467-023-41803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/19/2023] [Indexed: 10/02/2023] Open
Abstract
Neural computation is often traced in terms of either rate- or phase-codes. However, most circuit operations will simultaneously affect information across both coding schemes. It remains unclear how phase and rate coded information is transmitted, in the face of continuous modification at consecutive processing stages. Here, we study this question in the entorhinal cortex (EC)- dentate gyrus (DG)- CA3 system using three distinct computational models. We demonstrate that DG feedback inhibition leverages EC phase information to improve rate-coding, a computation we term phase-to-rate recoding. Our results suggest that it i) supports the conservation of phase information within sparse rate-codes and ii) enhances the efficiency of plasticity in downstream CA3 via increased synchrony. Given the ubiquity of both phase-coding and feedback circuits, our results raise the question whether phase-to-rate recoding is a recurring computational motif, which supports the generation of sparse, synchronous population-rate-codes in areas beyond the DG.
Collapse
Affiliation(s)
- Daniel Müller-Komorowska
- Neural Coding and Brain Computing Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan.
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany.
| | - Baris Kuru
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Heinz Beck
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen e.V, Bonn, Germany
| | - Oliver Braganza
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany.
- Institute for Socio-Economics, University of Duisburg-Essen, Duisburg, Germany.
| |
Collapse
|
28
|
de León Reyes NS, Sierra Díaz P, Nogueira R, Ruiz-Pino A, Nomura Y, de Solis CA, Schulkin J, Asok A, Leroy F. Corticotropin-releasing hormone signaling from prefrontal cortex to lateral septum suppresses interaction with familiar mice. Cell 2023; 186:4152-4171.e31. [PMID: 37669667 PMCID: PMC7615103 DOI: 10.1016/j.cell.2023.08.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/13/2023] [Accepted: 08/08/2023] [Indexed: 09/07/2023]
Abstract
Social preference, the decision to interact with one member of the same species over another, is critical to optimize social interactions. Thus, adult rodents favor interacting with novel conspecifics over familiar ones, but whether this social preference stems from neural circuits facilitating interactions with novel individuals or suppressing interactions with familiar ones remains unknown. Here, we identify neurons in the infra-limbic area (ILA) of the mouse prefrontal cortex that express the neuropeptide corticotropin-releasing hormone (CRH) and project to the dorsal region of the rostral lateral septum (rLS). We show how release of CRH during familiar encounters disinhibits rLS neurons, thereby suppressing social interactions with familiar mice and contributing to social novelty preference. We further demonstrate how the maturation of CRH expression in ILA during the first 2 post-natal weeks enables the developmental shift from a preference for littermates in juveniles to a preference for novel mice in adults.
Collapse
Affiliation(s)
- Noelia Sofia de León Reyes
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández de Elche), San Juan de Alicante, Alicante, Spain
| | - Paula Sierra Díaz
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández de Elche), San Juan de Alicante, Alicante, Spain
| | - Ramon Nogueira
- Center for Theoretical Neuroscience, Columbia University, New York, USA; Department of Neuroscience, Columbia University, New York, USA; Zuckerman Mind Brain & Behavior Institute, New York, USA
| | - Antonia Ruiz-Pino
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández de Elche), San Juan de Alicante, Alicante, Spain
| | - Yuki Nomura
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández de Elche), San Juan de Alicante, Alicante, Spain
| | - Christopher A de Solis
- Department of Neuroscience, Columbia University, New York, USA; Zuckerman Mind Brain & Behavior Institute, New York, USA
| | - Jay Schulkin
- School of Medicine, University of Washington, Seattle, USA
| | - Arun Asok
- Department of Neuroscience, Columbia University, New York, USA; Zuckerman Mind Brain & Behavior Institute, New York, USA
| | - Felix Leroy
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández de Elche), San Juan de Alicante, Alicante, Spain.
| |
Collapse
|
29
|
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
|
30
|
Pozo M, Milà-Guasch M, Haddad-Tóvolli R, Boudjadja M, Chivite I, Toledo M, Gómez-Valadés A, Eyre E, Ramírez S, Obri A, Ben-Ami Bartal I, D'Agostino G, Costa-Font J, Claret M. Negative energy balance hinders prosocial helping behavior. Proc Natl Acad Sci U S A 2023; 120:e2218142120. [PMID: 37023123 PMCID: PMC10104524 DOI: 10.1073/pnas.2218142120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/02/2023] [Indexed: 04/07/2023] Open
Abstract
The internal state of an animal, including homeostatic requirements, modulates its behavior. Negative energy balance stimulates hunger, thus promoting a range of actions aimed at obtaining food. While these survival actions are well established, the influence of the energy status on prosocial behavior remains unexplored. We developed a paradigm to assess helping behavior in which a free mouse was faced with a conspecific trapped in a restrainer. We measured the willingness of the free mouse to liberate the confined mouse under diverse metabolic conditions. Around 42% of ad libitum-fed mice exhibited a helping behavior, as evidenced by the reduction in the latencies to release the trapped cagemate. This behavior was independent of subsequent social contact reward and was associated with changes in corticosterone indicative of emotional contagion. This decision-making process was coupled with reduced blood glucose excursions and higher Adenosine triphosphate (ATP):Adenosine diphosphate (ADP) ratios in the forebrain of helper mice, suggesting that it was a highly energy-demanding process. Interestingly, chronic (food restriction and type 2 diabetes) and acute (chemogenetic activation of hunger-promoting AgRP neurons) situations mimicking organismal negative energy balance and enhanced appetite attenuated helping behavior toward a distressed conspecific. To investigate similar effects in humans, we estimated the influence of glycated hemoglobin (a surrogate of long-term glycemic control) on prosocial behavior (namely charity donation) using the Understanding Society dataset. Our results evidenced that organismal energy status markedly influences helping behavior and that hypothalamic AgRP neurons are at the interface of metabolism and prosocial behavior.
Collapse
Affiliation(s)
- Macarena Pozo
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Mehdi Boutagouga Boudjadja
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, M13 9PTManchester, United Kingdom
| | - Iñigo Chivite
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Miriam Toledo
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Alicia G. Gómez-Valadés
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
| | - Inbal Ben-Ami Bartal
- School of Psychological Sciences, Tel-Aviv University, 6997801Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, 6997801Tel Aviv, Israel
| | - Giuseppe D'Agostino
- Faculty of Biology, Medicine and Health, School of Medical Sciences, University of Manchester, M13 9PTManchester, United Kingdom
| | - Joan Costa-Font
- Department of Health Policy, London School of Economics and Political Science, WC2A 2AELondon, United Kingdom
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Spain
- School of Medicine, Universitat de Barcelona, 08036Barcelona, Spain
| |
Collapse
|
31
|
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: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
32
|
Barone A, De Simone G, Ciccarelli M, Buonaguro EF, Tomasetti C, Eramo A, Vellucci L, de Bartolomeis A. A Postsynaptic Density Immediate Early Gene-Based Connectome Analysis of Acute NMDAR Blockade and Reversal Effect of Antipsychotic Administration. Int J Mol Sci 2023; 24:ijms24054372. [PMID: 36901803 PMCID: PMC10002165 DOI: 10.3390/ijms24054372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Although antipsychotics' mechanisms of action have been thoroughly investigated, they have not been fully elucidated at the network level. We tested the hypothesis that acute pre-treatment with ketamine (KET) and administration of asenapine (ASE) would modulate the functional connectivity of brain areas relevant to the pathophysiology of schizophrenia, based on transcript levels of Homer1a, an immediate early gene encoding a key molecule of the dendritic spine. Sprague-Dawley rats (n = 20) were assigned to KET (30 mg/kg) or vehicle (VEH). Each pre-treatment group (n = 10) was randomly split into two arms, receiving ASE (0.3 mg/kg), or VEH. Homer1a mRNA levels were evaluated by in situ hybridization in 33 regions of interest (ROIs). We computed all possible pairwise Pearson correlations and generated a network for each treatment group. Acute KET challenge was associated with negative correlations between the medial portion of cingulate cortex/indusium griseum and other ROIs, not detectable in other treatment groups. KET/ASE group showed significantly higher inter-correlations between medial cingulate cortex/indusium griseum and lateral putamen, the upper lip of the primary somatosensory cortex, septal area nuclei, and claustrum, in comparison to the KET/VEH network. ASE exposure was associated with changes in subcortical-cortical connectivity and an increase in centrality measures of the cingulate cortex and lateral septal nuclei. In conclusion, ASE was found to finely regulate brain connectivity by modelling the synaptic architecture and restoring a functional pattern of interregional co-activation.
Collapse
Affiliation(s)
- Annarita Barone
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Giuseppe De Simone
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Elisabetta Filomena Buonaguro
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | | | - Licia Vellucci
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Laboratory of Translational and Molecular Psychiatry, Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-7463673; Fax: +39-081-7462644
| |
Collapse
|
33
|
Bouras NN, Mack NR, Gao WJ. Prefrontal modulation of anxiety through a lens of noradrenergic signaling. Front Syst Neurosci 2023; 17:1173326. [PMID: 37139472 PMCID: PMC10149815 DOI: 10.3389/fnsys.2023.1173326] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Anxiety disorders are the most common class of mental illness in the U.S., affecting 40 million individuals annually. Anxiety is an adaptive response to a stressful or unpredictable life event. Though evolutionarily thought to aid in survival, excess intensity or duration of anxiogenic response can lead to a plethora of adverse symptoms and cognitive dysfunction. A wealth of data has implicated the medial prefrontal cortex (mPFC) in the regulation of anxiety. Norepinephrine (NE) is a crucial neuromodulator of arousal and vigilance believed to be responsible for many of the symptoms of anxiety disorders. NE is synthesized in the locus coeruleus (LC), which sends major noradrenergic inputs to the mPFC. Given the unique properties of LC-mPFC connections and the heterogeneous subpopulation of prefrontal neurons known to be involved in regulating anxiety-like behaviors, NE likely modulates PFC function in a cell-type and circuit-specific manner. In working memory and stress response, NE follows an inverted-U model, where an overly high or low release of NE is associated with sub-optimal neural functioning. In contrast, based on current literature review of the individual contributions of NE and the PFC in anxiety disorders, we propose a model of NE level- and adrenergic receptor-dependent, circuit-specific NE-PFC modulation of anxiety disorders. Further, the advent of new techniques to measure NE in the PFC with unprecedented spatial and temporal resolution will significantly help us understand how NE modulates PFC function in anxiety disorders.
Collapse
|
34
|
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: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
35
|
Zhang K, Guo YC, Wang XD, Zhu YJ, Pan BX, Deng C, Yuan TF. Lateral septum inputs to nucleus accumbens mediates stress induced suppression of natural reward seeking. Pharmacol Res 2022; 184:106463. [PMID: 36162602 DOI: 10.1016/j.phrs.2022.106463] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022]
Abstract
Stress alters the level of reward evaluation and seeking. However, the neural circuitry mechanisms underlying stress induced effects on natural reward seeking remain unclear. Here we report a septal-accumbens pathway that mediates the effects of acute stress on reward seeking suppression. We first established the sucrose oral self-administration paradigm and measured the effects of acute stress on reward seeking behavior after 21 days of abstinence. Both forced swimming stress and foot shock stress significantly suppressed the natural reward seeking. Among a variety of brain regions, intermediolateral septum (LSi) appear as a strong stress-responsive area containing abundant c-Fos positive cells; chemogenetic inactivation of LSi reinstated the reward seeking behavior. To elucidate the downstream targets receiving LSi projections, we combined pathway-specific retro-labeling and chemogenetic manipulation to confirm the involvement of LSi-nucleus accumbens (NAc) rather than the Ventral tegmental area (VTA) in mediating the observed behavioral responses. In conclusion, the septal-accumbal projection constitute a discrete circuit dictating the stress evoked alterations on reward seeking and may implicate in treatment of stress induced anhedonia.
Collapse
Affiliation(s)
- Ke Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Chen Guo
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Dong Wang
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying-Jie Zhu
- Shenzhen Key Lab of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bing-Xing Pan
- Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Cheng Deng
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China; Institutes for Systems Genetics, Frontiers Science Centre for Disease-Related Molecular Network, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610212, Sichuan, China.
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China; Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Wang M, Li Z, Song Y, Sun Q, Deng L, Lin Z, Zeng Y, Qiu C, Lin J, Guo H, Chen J, Guo W. Genetic tagging of the adenosine A2A receptor reveals its heterogeneous expression in brain regions. Front Neuroanat 2022; 16:978641. [PMID: 36059431 PMCID: PMC9434489 DOI: 10.3389/fnana.2022.978641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/29/2022] [Indexed: 11/22/2022] Open
Abstract
The adenosine A2A receptor (A2AR), a G protein-coupled receptor, is involved in numerous and varied physiological and pathological processes, including inflammation, immune responses, blood flow, and neurotransmission. Accordingly, it has become an important drug target for the treatment of neuropsychiatric disorders. However, the exact brain distribution of A2AR in regions outside the striatum that display relatively low levels of endogenous A2AR expression has hampered the exploration of A2AR functions under both physiological and pathological conditions. To further study the detailed distribution of the A2AR in low-expression regions, we have generated A2AR knock-in mice in which the 3xHA-2xMyc epitope tag sequence was fused to the C-terminus of A2AR (A2AR-tag mice) via CRISPR/Cas9 technology. Here, using CRISPR/Cas9 technology, we have generated A2AR knock-in mice in which the 3xHA-2xMyc epitope tag sequence was fused to the C-terminus of A2AR (A2AR-tag mice). The A2AR-tag mice exhibited normal locomotor activity and emotional state. Consistent with previous studies, A2AR fluorescence was widely detected in the striatum, nucleus accumbens, and olfactory tubercles, with numerous labeled cells being evident in these regions in the A2AR-tag mouse. Importantly, we also identified the presence of a few but clearly labeled cells in heterogeneous brain regions where A2AR expression has not previously been unambiguously detected, including the lateral septum, hippocampus, amygdala, cerebral cortex, and gigantocellular reticular nucleus. The A2AR-tag mouse represents a novel useful genetic tool for monitoring the expression of A2AR and dissecting its functions in brain regions other than the striatum.
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
| | - 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
| | - Yue Song
- 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
| | - Qiuqin Sun
- 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
| | - 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, 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
| | - Yang Zeng
- Shanghai Pregen Biotechnology Co., Ltd., Shanghai, China
| | - Chunhong Qiu
- Shanghai Pregen Biotechnology Co., Ltd., Shanghai, China
| | - Jingjing Lin
- 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, China
| | - Hui 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
| | - 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
- Jiangfan Chen,
| | - 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
- *Correspondence: Wei Guo,
| |
Collapse
|