1
|
King LB, Walum H, Xiao Y, Caslin AY, Haddad FC, Young LJ, Johnson ZV. An oxytocin receptor gene polymorphism is associated with distinct neural responses to mating encounters in male prairie voles. Horm Behav 2025; 173:105761. [PMID: 40414117 DOI: 10.1016/j.yhbeh.2025.105761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/22/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
Oxytocin is a conserved neuropeptide that regulates social and reproductive behaviors in diverse species. Genetic variation in Oxtr, the gene encoding the oxytocin receptor (OXTR), is associated with variation in social attachment behaviors in rodents and humans; however, it is unclear how genetic variation in Oxtr shapes the function of specific neural systems during social contexts. Here we address this question using the socially monogamous prairie vole (Microtus ochrogaster), a species that expresses an array of OXTR-dependent social behaviors and possesses Oxtr gene polymorphisms that predict individual variation in brain region-specific OXTR expression. We test the neural and behavioral effects of an Oxtr gene polymorphism that has previously been associated with brain region-specific OXTR expression and social attachment behaviors in male prairie voles. Our results suggest that, during brief mating encounters, Oxtr genotype is not associated with differences in mating behavior or in expression levels of the activity-dependent immediate early gene product FOS within brain regions, but it is associated with differences in correlated FOS expression patterns across brain regions.
Collapse
Affiliation(s)
- Lanikea B King
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Hasse Walum
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Yao Xiao
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Asha Y Caslin
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Fuad C Haddad
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Larry J Young
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America
| | - Zachary V Johnson
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329, United States of America; Emory National Primate Research Center, Emory University, Atlanta, GA 30329, United States of America.
| |
Collapse
|
2
|
Chang HT, Cheng KH, Hung YC, Hsu KS. Oxytocin signaling in the ventral tegmental area mediates social isolation-induced craving for social interaction. J Biomed Sci 2025; 32:37. [PMID: 40098181 PMCID: PMC11912778 DOI: 10.1186/s12929-025-01130-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Social interaction is crucial for mental health across animal species. Social experiences, especially in early-life stages, strongly influence brain function and social behavior later in life. Acute social isolation (SI) increases motivation to seek social interaction, but little is known about its underlying neuronal and circuitry mechanisms. Here, we focus on oxytocin signaling in the ventral tegmental area (VTA), a vital node of the brain's reward network, as a potential mechanism for SI-induced craving for social interaction. METHODS Adolescent (4-week-old) or adult (14-week-old) male C57BL/6J mice underwent a 1-week SI. Free interaction, object exploration, three-chamber social approach, and habituation tests were used to assess social and non-social behavior changes. Viral vectors were used to decipher the underlying neural circuitry, and chemogenetic techniques were applied to modify neuronal activity. RESULTS We found that in male C57BL/6J mice, SI during adolescence, but not adulthood, leads to increased craving for social interaction and object exploration, accompanied by impaired social habituation, social novelty preference, and social recognition memory (SRM). SI-induced craving for social interaction and SRM deficit is still observed upon regrouping. Through cell-type-specific manipulations with designer receptors exclusively activated by designer drugs (DREADD), we show that oxytocin neurons in the paraventricular nucleus of the hypothalamus (PVN) are crucial for SI-induced social behavior changes. Chemogenetic activation of PVN oxytocin neurons recapitulates social behavior changes observed in SI mice, whereas chemogenetic inhibition of oxytocin neurons prevents social behavior changes caused by SI. Moreover, we found that dopaminergic neurons in the VTA mediate SI-induced craving for social interaction through their projections to the medial prefrontal cortex (mPFC), but not to the nucleus accumbens. Injection of a specific oxytocin receptor antagonist L368,899 into the VTA or chemical lesions of dopaminergic axon terminals in the mPFC with local application of 6-hydroxydopamine ameliorates SI-induced social behavior changes. CONCLUSIONS These findings suggest that adolescent SI has enduring effects on social behaviors in male mice through an oxytocinergic modulation of the VTA-to-mPFC dopaminergic circuit activity.
Collapse
Affiliation(s)
- Hsin-Tzu Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan City, 70101, Taiwan
| | - Kuan-Hsiang Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan City, 70101, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|
3
|
Câmara AB, Brandão IA. The neuroinflammatory effects of Nociceptin/Orphanin FQ receptor activation can be related to depressive-like behavior. J Psychiatr Res 2025; 183:174-188. [PMID: 39978292 DOI: 10.1016/j.jpsychires.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/08/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
There is limited information on the role of the Nociceptin/Orphanin FQ receptor (NOPR) in neuroinflammation, and there is growing interest in the participation of the NOPR in depression etiology. This study aims to evaluate the neuroinflammatory effects of the NOPR activation in mice submitted to social defeat protocol (SDP). Firstly, male Swiss mice were submitted to the social defeat protocol during 10 or 20 days and treated with the NOPR agonist Ro 65-6570 (1.5 or 2 mg/kg; ip). Subsequently, behavioral tests were applied to evaluate depressive-like behaviors. Finally, inflammatory cytokines were measured in the animals' brains and blood. A meta-analysis, including 11 experiments, was also conducted to evaluate if the NOPR activation contributes to inflammation. The studies' weights, odds ratios, and confidence intervals were used to calculate the average effect size as the main outcome measure. The software SPSS v.29 and R programming language were used to analyze the data. The SDP and/or NOP agonist reduced distance traveled and exploration rate in the open field test. The SDP and/or the NOP agonist also increased immobility time in the tail suspension test, as well as reduced social interaction. Additionally, the NOP agonist increased the concentration of IL-6 and TNF alpha in the hippocampus, as well as reduced the IL-10 concentration in the hippocampus, but not in prefrontal cortex and serum. The SDP increased the concentration of IL-6 and TNF alpha in animals' serum and prefrontal cortex, but not in the hippocampus. The role of NOPR in neuroinflammation was regardless of the social defeat stress in the hippocampus. Meta-analysis also demonstrated the participation of NOPR activation in inducing inflammation in mice models. We suggest that upregulation of NOPR can activate signaling pathways involved in neuroinflammation, contributing to depression etiology.
Collapse
Affiliation(s)
| | - Igor Augusto Brandão
- Bioinformatics Multidisciplinary Environment, Federal University of Rio Grande do Norte, Brazil
| |
Collapse
|
4
|
Yu S, Yin Z, Ling M, Chen Z, Zhang Y, Pan Y, Zhang Y, Cai X, Chen Z, Hao H, Zheng X. Ginsenoside Rg1 enriches gut microbial indole-3-acetic acid to alleviate depression-like behavior in mice via oxytocin signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156186. [PMID: 39515104 DOI: 10.1016/j.phymed.2024.156186] [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: 07/16/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND PURPOSE Although a large collection of data has shown that ginsenosides, the major active ingredients from Ginseng, have neuroprotective and anti-depressant effect, the mechanism of action is incompletely understood. This study aims to elucidate the antidepressant mechanism of ginsenoside Rg1 (Rg1), a poorly absorbed ginsenoside, from the perspective of gut microbe to brain signaling. METHODS A mouse model of depression was induced by unpredictable mild stress (UMS). Behavioral and neurochemical tests were conducted to evaluate the effect and mechanism of Rg1 on depressive behavior. Non-target and target metabolomics were performed to identify the signaling metabolites underlying the antidepressant efficacy of Rg1. Gut microbial structure was analyzed by 16S rRNA sequencing and the potential functional strains associated with Rg1 action were investigated by in vitro bacterial culture. Chemical intervention was used to explore the mechanism of Rg1 and signaling metabolite. RESULTS Rg1 improved UMS-induced despair, anxiety-like and social avoidance behaviors in mice, which were accompanied by increased hypothalamic oxytocin secretion and restored neural proliferation in the hippocampus. Metabolomic analysis of the gut-brain axis revealed that Rg1 increased the concentration of serum and brain indole-3-acetic acid (IAA), a bacterial metabolite that was partially attributed to the enrichment of Lactobacillus murinus in the gut microbiome. Oral supplementation of IAA mimicked the anti-depressant action of Rg1, while oxytocin receptor antagonist abrogated the anti-depressant effects of both Rg1 and IAA. CONCLUSION Our work provides a new gut-to-brain signaling mechanism for the antidepressant effects of Rg1. In particular, Rg1 enriches the abundance of Lactobacillus murinus, which in turn increases the level of brain IAA and potentiates hypothalamic oxytocin signal. These findings suggest a promising pathway for producing antidepressant effects through gut-brain crosstalk.
Collapse
Affiliation(s)
- Siqi Yu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhe Yin
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ming Ling
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhuo Chen
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yangfan Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yarui Pan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Youying Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoying Cai
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zeyu Chen
- School of Medicine, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiao Zheng
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China; School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
5
|
Zhang J, Xie C, Xu P, Tong Q, Xiao L, Zhong J. Projections from subfornical organ to bed nucleus of the stria terminalis modulate inflammation-induced anxiety-like behaviors in mice. SCIENCE ADVANCES 2024; 10:eadp9413. [PMID: 39602546 PMCID: PMC11601211 DOI: 10.1126/sciadv.adp9413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Peripheral inflammation is closely related to the pathogenesis of sickness behaviors and psychiatric disorders such as anxiety and depression. The circumventricular organs (CVOs) are important brain sites to perceive peripheral inflammatory signals, but few studies have reported their role in inflammation-induced anxiety or depression. Using a mouse model of lipopolysaccharide (LPS)-induced inflammation, we identified a previously unreported role of the subfornical organ (SFO), one of the CVOs, in combating inflammation-induced anxiety. LPS treatment induced anxiety-like and sickness behaviors in mice. Although both the SFO and the organum vasculosum of the lamina terminalis (a CVO) neurons were activated after LPS treatment, only manipulating SFO neurons modulated LPS-induced anxiety-like behaviors. Activating or inhibiting SFO neurons alleviated or aggravated LPS-induced anxiety-like behaviors. In addition, SFO exerted this effect through glutamatergic projections to the bed nucleus of the stria terminalis. Manipulating SFO neurons did not affect LPS-induced sickness behaviors. Thus, we uncovered an active role of SFO neurons in counteracting peripheral inflammation-induced anxiety.
Collapse
Affiliation(s)
- Jinlin Zhang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Chuantong Xie
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Peiyao Xu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qiuping Tong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Xiao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Department of Anesthesiology, Wusong Hospital Branch, Zhongshan Hospital Affiliated to Fudan University, Shanghai 201999, China
| |
Collapse
|
6
|
Chaipunko S, Sookkua T, Nopparat C, Chutabhakdikul N. Oxytocin Protects Against Corticosterone-Induced DA Dysfunction: An Involvement of the PKA/CREB Pathway. Neurochem Res 2024; 50:38. [PMID: 39607552 PMCID: PMC11604774 DOI: 10.1007/s11064-024-04294-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Chronic stress disrupts dopamine (DA) transmission, adversely affecting mood and contribution to neuropsychiatric disorders like ADHD, autism, schizophrenia, anxiety, depression, and drug addiction. The neuropeptide oxytocin (OXT) plays a key role in social cognition, bonding, attachment, and parenting behaviors. In addition, OXT can modulate the activity of the HPA axis, counteracting the effects of stress, and alleviating fear and anxiety. However, whether OXT can mitigate stress-induced DA dysfunction and the underlying mechanisms remains unclear. This study investigated the neuroprotective effects of OXT on corticosterone (CORT) induced DA dysfunction in the neuroblastoma cell line SH-SY5Y. The results revealed that CORT decreases the levels of intracellular signaling molecules associated with DA function, including phosphorylated tyrosine hydroxylase (pTH), phosphorylated cAMP response element-binding protein (pCREB), and protein kinase A (PKA). Interestingly, pretreatment with OXT mitigated CORT-induced DA dysfunction through its potent PKA activator properties. In addition, the neuroprotective effect of OXT was abolished by atosiban (an OXT receptor antagonist) or H89 (a PKA inhibitor). Our results suggest that OXT protects dopaminergic neuroblastoma cells from CORT-induced DA dysfunction, potentially through the involvement of oxytocin receptors and the PKA/CREB signaling pathway. These findings contribute to the understanding of the neurobiological mechanisms underlying stress resilience and highlight potential pathways for developing targeted treatments that leverage the neuroprotective properties of OXT to address disorders characterized by DA dysregulation and impaired stress responses.
Collapse
Affiliation(s)
- Sirinun Chaipunko
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
- Faculty of Physical Therapy, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Tichaporn Sookkua
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
- Faculty of Physical Therapy, Saint Louis College, Bangkok, 10120, Thailand
| | - Chutikorn Nopparat
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
- Innovative Learning Center, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
7
|
Wang Y, Xu H, Chen S, Chen J, Zheng Q, Ma Y, Zhao X, Shi Y, Xiao L. Oxytocin Protects Nigrostriatal Dopamine Signal via Activating GABAergic Circuit in the MPTP-Induced Parkinson's Disease Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310244. [PMID: 39099429 PMCID: PMC11423065 DOI: 10.1002/advs.202310244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 07/23/2024] [Indexed: 08/06/2024]
Abstract
The most pronounced neuropathological feature of Parkinson's disease (PD) is the loss of dopamine (DA) neurons in the substantia nigra compacta (SNc), which depletes striatal DA. Hypothalamic oxytocin is found to be reduced in PD patients and closely interacts with the DA system, but the role of oxytocin in PD remains unclear. Here, the disturbances of endogenous oxytocin level and the substantia nigra (SN) oxytocin receptor expression in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model is observed, correlated with the striatal tyrosine hydroxylase (TH) expression reduction. Killing/silencing hypothalamic oxytocin neurons aggravates the vulnerability of nigrostriatal DA signal to MPTP, whereas elevating oxytocin level by intranasal delivery or microinjecting into the SN promotes the resistance. In addition, knocking out SN oxytocin receptors induces the time-dependent reductions of SNc DA neurons, striatal TH expression, and striatal DA level by increasing neuronal excitotoxicity. These results further uncover that oxytocin dampens the excitatory synaptic inputs onto DA neurons via activating oxytocin receptor-expressed SN GABA neurons, which target GABA(B) receptors expressed in SNc DA neuron-projecting glutamatergic axons, to reduce excitotoxicity. Thus, besides the well-known prosocial effect, oxytocin acts as a key endogenous factor in protecting the nigrostriatal DA system.
Collapse
Affiliation(s)
- Yurong Wang
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Hao Xu
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Saiyong Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Junhao Chen
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Qimeng Zheng
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xinru Zhao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ying Shi
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| |
Collapse
|
8
|
Maejima Y, Yokota S, Yamachi M, Misaka S, Ono T, Oizumi H, Mizuno K, Hidema S, Nishimori K, Aoyama M, de Wet H, Shimomura K. Traditional Japanese medicine Kamikihito ameliorates sucrose preference, chronic inflammation and obesity induced by a high fat diet in middle-aged mice. Front Endocrinol (Lausanne) 2024; 15:1387964. [PMID: 38742193 PMCID: PMC11089234 DOI: 10.3389/fendo.2024.1387964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
The high prevalence of obesity has become a pressing global public health problem and there exists a strong association between increased BMI and mortality at a BMI of 25 kg/m2 or higher. The prevalence of obesity is higher among middle-aged adults than among younger groups and the combination of aging and obesity exacerbate systemic inflammation. Increased inflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha (TNFα) are hallmarks of obesity, and promote the secretion of hepatic C-reactive protein (CRP) which further induces systematic inflammation. The neuropeptide oxytocin has been shown to have anti-obesity and anti-inflammation effects, and also suppress sweet-tasting carbohydrate consumption in mammals. Previously, we have shown that the Japanese herbal medicine Kamikihito (KKT), which is used to treat neuropsychological stress disorders in Japan, functions as an oxytocin receptors agonist. In the present study, we further investigated the effect of KKT on body weight (BW), food intake, inflammation, and sweet preferences in middle-aged obese mice. KKT oral administration for 12 days decreased the expression of pro-inflammatory cytokines in the liver, and the plasma CRP and TNFα levels in obese mice. The effect of KKT administration was found to be different between male and female mice. In the absence of sucrose, KKT administration decreased food intake only in male mice. However, while having access to a 30% sucrose solution, both BW and food intake was decreased by KKT administration in male and female mice; but sucrose intake was decreased in female mice alone. In addition, KKT administration decreased sucrose intake in oxytocin deficient lean mice, but not in the WT lean mice. The present study demonstrates that KKT ameliorates chronic inflammation, which is strongly associated with aging and obesity, and decreases food intake in male mice as well as sucrose intake in female mice; in an oxytocin receptor dependent manner.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Obesity and Inflammation research, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shoko Yokota
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Megumi Yamachi
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shingen Misaka
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tomoyuki Ono
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroaki Oizumi
- Tsumura Kampo Research Laboratories, Kampo Research and Development Division, Tsumura & Co., Ibaraki, Japan
| | - Keita Mizuno
- Tsumura Kampo Research Laboratories, Kampo Research and Development Division, Tsumura & Co., Ibaraki, Japan
| | - Shizu Hidema
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masato Aoyama
- Department of Animal Science, Faculty of Agriculture, Utsunomiya University, Utsunomiya, Japan
| | - Heidi de Wet
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Obesity and Inflammation research, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
9
|
Ren L. The mechanistic basis for the rapid antidepressant-like effects of ketamine: From neural circuits to molecular pathways. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110910. [PMID: 38061484 DOI: 10.1016/j.pnpbp.2023.110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Conventional antidepressants that target monoaminergic receptors require several weeks to be efficacious. This lag represents a significant problem in the currently available treatments for serious depression. Ketamine, acting as an N-methyl-d-aspartate receptor antagonist, was shown to have rapid antidepressant-like effects, marking a significant advancement in the study of mood disorders. However, serious side effects and adverse reactions limit its clinical use. Considering the limitations of ketamine, it is crucial to further define the network targets of ketamine. The rapid action of ketamine an as antidepressant is thought to be mediated by the glutamate system. It is believed that synaptic plasticity is essential for the rapid effects of ketamine as an antidepressant. Other mechanisms include the involvement of the γ-aminobutyric acidergic (GABAergic), 5-HTergic systems, and recent studies have linked astrocytes to ketamine's rapid antidepressant-like effects. The interactions between these systems exert a synergistic rapid antidepressant effect through neural circuits and molecular mechanisms. Here, we discuss the neural circuits and molecular mechanisms underlying the action of ketamine. This work will help explain how molecular and neural targets are responsible for the effects of rapidly acting antidepressants and will aid in the discovery of new therapeutic approaches for major depressive disorder.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu 611137, China.
| |
Collapse
|
10
|
Wu M, Zhang X, Feng S, Freda SN, Kumari P, Dumrongprechachan V, Kozorovitskiy Y. Dopamine pathways mediating affective state transitions after sleep loss. Neuron 2024; 112:141-154.e8. [PMID: 37922904 PMCID: PMC10841919 DOI: 10.1016/j.neuron.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023]
Abstract
The pathophysiology of affective disorders-particularly circuit-level mechanisms underlying bidirectional, periodic affective state transitions-remains poorly understood. In patients, disruptions of sleep and circadian rhythm can trigger transitions to manic episodes, whereas depressive states are reversed. Here, we introduce a hybrid automated sleep deprivation platform to induce transitions of affective states in mice. Acute sleep loss causes mixed behavioral states, featuring hyperactivity, elevated social and sexual behaviors, and diminished depressive-like behaviors, where transitions depend on dopamine (DA). Using DA sensor photometry and projection-targeted chemogenetics, we reveal that elevated DA release in specific brain regions mediates distinct behavioral changes in affective state transitions. Acute sleep loss induces DA-dependent enhancement in dendritic spine density and uncaging-evoked dendritic spinogenesis in the medial prefrontal cortex, whereas optically mediated disassembly of enhanced plasticity reverses the antidepressant effects of sleep deprivation on learned helplessness. These findings demonstrate that brain-wide dopaminergic pathways control sleep-loss-induced polymodal affective state transitions.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL 60208, USA
| | - Xin Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sihan Feng
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Pushpa Kumari
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
11
|
Menon R, Neumann ID. Detection, processing and reinforcement of social cues: regulation by the oxytocin system. Nat Rev Neurosci 2023; 24:761-777. [PMID: 37891399 DOI: 10.1038/s41583-023-00759-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/29/2023]
Abstract
Many social behaviours are evolutionarily conserved and are essential for the healthy development of an individual. The neuropeptide oxytocin (OXT) is crucial for the fine-tuned regulation of social interactions in mammals. The advent and application of state-of-the-art methodological approaches that allow the activity of neuronal circuits involving OXT to be monitored and functionally manipulated in laboratory mammals have deepened our understanding of the roles of OXT in these behaviours. In this Review, we discuss how OXT promotes the sensory detection and evaluation of social cues, the subsequent approach and display of social behaviour, and the rewarding consequences of social interactions in selected reproductive and non-reproductive social behaviours. Social stressors - such as social isolation, exposure to social defeat or social trauma, and partner loss - are often paralleled by maladaptations of the OXT system, and restoring OXT system functioning can reinstate socio-emotional allostasis. Thus, the OXT system acts as a dynamic mediator of appropriate behavioural adaptations to environmental challenges by enhancing and reinforcing social salience and buffering social stress.
Collapse
Affiliation(s)
- Rohit Menon
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
12
|
Bode A. Romantic love evolved by co-opting mother-infant bonding. Front Psychol 2023; 14:1176067. [PMID: 37915523 PMCID: PMC10616966 DOI: 10.3389/fpsyg.2023.1176067] [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: 02/28/2023] [Accepted: 09/18/2023] [Indexed: 11/03/2023] Open
Abstract
For 25 years, the predominant evolutionary theory of romantic love has been Fisher's theory of independent emotion systems. That theory suggests that sex drive, romantic attraction (romantic love), and attachment are associated with distinct neurobiological and endocrinological systems which evolved independently of each other. Psychological and neurobiological evidence, however, suggest that a competing theory requires attention. A theory of co-opting mother-infant bonding sometime in the recent evolutionary history of humans may partially account for the evolution of romantic love. I present a case for this theory and a new approach to the science of romantic love drawing on human psychological, neurobiological, and (neuro)endocrinological studies as well as animal studies. The hope is that this theoretical review, along with other publications, will generate debate in the literature about the merits of the theory of co-opting mother-infant bonding and a new evolutionary approach to the science of romantic love.
Collapse
|
13
|
Cui X, Tong Q, Xu H, Xie C, Xiao L. A putative loop connection between VTA dopamine neurons and nucleus accumbens encodes positive valence to compensate for hunger. Prog Neurobiol 2023; 229:102503. [PMID: 37451329 DOI: 10.1016/j.pneurobio.2023.102503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Dopamine (DA) signal play pivotal roles in regulating motivated behaviors, including feeding behavior, but the role of midbrain DA neurons in modulating food intake and neural circuitry mechanisms remain largely unknown. Here, we found that activating but not inhibiting ventral tegmental area (VTA) DA neurons reduces mouse food intake. Furthermore, DA neurons in ventral VTA, especially neurons projecting to the medial nucleus accumbens (NAc), are activated by refeeding in the 24 h fasted mice. Combing neural circuitry tracing, optogenetic, chemogenetic, and pharmacological manipulations, we established that the VTA→medial NAc→VTA loop circuit is critical for the VTA DA neurons activation-induced food intake reduction. Moreover, activating either VTA DA neurons or dopaminergic axons in medial NAc elevates positive valence, which will compensate for the hungry-induced food intake. Thus, our study identifies a subset of positive valence-encoded VTA DA neurons forming possible loop connections with medial NAc that are anorexigenic.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qiuping Tong
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hao Xu
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Chuantong Xie
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Xiao
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
14
|
Schimmer J, Patwell R, Küppers S, Grinevich V. The Relationship Between Oxytocin and Alcohol Dependence. Curr Top Behav Neurosci 2023. [PMID: 37697074 DOI: 10.1007/7854_2023_444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
The hypothalamic neuropeptide oxytocin (OT) is well known for its prosocial, anxiolytic, and ameliorating effects on various psychiatric conditions, including alcohol use disorder (AUD). In this chapter, we will first introduce the basic neurophysiology of the OT system and its interaction with other neuromodulatory and neurotransmitter systems in the brain. Next, we provide an overview over the current state of research examining the effects of acute and chronic alcohol exposure on the OT system as well as the effects of OT system manipulation on alcohol-related behaviors in rodents and humans. In rodent models of AUD, OT has been repeatedly shown to reduce ethanol consumption, particularly in models of acute alcohol exposure. In humans however, the results of OT administration on alcohol-related behaviors are promising but not yet conclusive. Therefore, we further discuss several physiological and methodological limitations to the effective application of OT in the clinic and how they may be mitigated by the application of synthetic OT receptor (OTR) agonists. Finally, we discuss the potential efficacy of cutting-edge pharmacology and gene therapies designed to specifically enhance endogenous OT release and thereby rescue deficient expression of OT in the brains of patients with severe forms of AUD and other incurable mental disorders.
Collapse
Affiliation(s)
- Jonas Schimmer
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Ryan Patwell
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Stephanie Küppers
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
15
|
Priest MF, Freda SN, Rieth IJ, Badong D, Dumrongprechachan V, Kozorovitskiy Y. Peptidergic and functional delineation of the Edinger-Westphal nucleus. Cell Rep 2023; 42:112992. [PMID: 37594894 PMCID: PMC10512657 DOI: 10.1016/j.celrep.2023.112992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 06/15/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
Many neuronal populations that release fast-acting excitatory and inhibitory neurotransmitters in the brain also contain slower-acting neuropeptides. These facultative peptidergic cell types are common, but it remains uncertain whether neurons that solely release peptides exist. Our fluorescence in situ hybridization, genetically targeted electron microscopy, and electrophysiological characterization suggest that most neurons of the non-cholinergic, centrally projecting Edinger-Westphal nucleus in mice are obligately peptidergic. We further show, using anterograde projection mapping, monosynaptic retrograde tracing, angled-tip fiber photometry, and chemogenetic modulation and genetically targeted ablation in conjunction with canonical assays for anxiety, that this peptidergic population activates in response to loss of motor control and promotes anxiety responses. Together, these findings elucidate an integrative, ethologically relevant role for the Edinger-Westphal nucleus and functionally align the nucleus with the periaqueductal gray, where it resides. This work advances our understanding of peptidergic modulation of anxiety and provides a framework for future investigations of peptidergic systems.
Collapse
Affiliation(s)
- Michael F Priest
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Isabelle J Rieth
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanna Badong
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
16
|
Oubraim S, Shen RY, Haj-Dahmane S. Oxytocin excites dorsal raphe serotonin neurons and bidirectionally gates their glutamate synapses. iScience 2023; 26:106707. [PMID: 37250336 PMCID: PMC10214716 DOI: 10.1016/j.isci.2023.106707] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Oxytocin (OXT) modulates wide spectrum of social and emotional behaviors via modulation of numerous neurotransmitter systems, including serotonin (5-HT). However, how OXT controls the function of dorsal raphe nucleus (DRN) 5-HT neurons remains unknown. Here, we reveal that OXT excites and alters the firing pattern of 5-HT neurons via activation of postsynaptic OXT receptors (OXTRs). In addition, OXT induces cell-type-specific depression and potentiation of DRN glutamate synapses by two retrograde lipid messengers, 2-arachidonoylglycerol (2-AG) and arachidonic acid (AA), respectively. Neuronal mapping demonstrates that OXT preferentially potentiates glutamate synapses of 5-HT neurons projecting to medial prefrontal cortex (mPFC) and depresses glutamatergic inputs to 5-HT neurons projecting to lateral habenula (LHb) and central amygdala (CeA). Thus, by engaging distinct retrograde lipid messengers, OXT exerts a target-specific gating of glutamate synapses on the DRN. As such, our data uncovers the neuronal mechanisms by which OXT modulates the function of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
17
|
Ahmed IA, Liu JJ, Gieniec KA, Bair-Marshall CJ, Adewakun AB, Hetzler BE, Arp CJ, Khatri L, Vanwalleghem GC, Seidenberg AT, Cowin P, Trauner D, Chao MV, Davis FM, Tsien RW, Froemke RC. Optopharmacological tools for precise spatiotemporal control of oxytocin signaling in the central nervous system and periphery. RESEARCH SQUARE 2023:rs.3.rs-2715993. [PMID: 37034806 PMCID: PMC10081362 DOI: 10.21203/rs.3.rs-2715993/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Oxytocin is a neuropeptide critical for maternal physiology and social behavior, and is thought to be dysregulated in several neuropsychiatric disorders. Despite the biological and neurocognitive importance of oxytocin signaling, methods are lacking to activate oxytocin receptors with high spatiotemporal precision in the brain and peripheral mammalian tissues. Here we developed and validated caged analogs of oxytocin which are functionally inert until cage release is triggered by ultraviolet light. We examined how focal versus global oxytocin application affected oxytocin-driven Ca2+ wave propagation in mouse mammary tissue. We also validated the application of caged oxytocin in the hippocampus and auditory cortex with electrophysiological recordings in vitro, and demonstrated that oxytocin uncaging can accelerate the onset of mouse maternal behavior in vivo. Together, these results demonstrate that optopharmacological control of caged peptides is a robust tool with spatiotemporal precision for modulating neuropeptide signaling throughout the brain and body.
Collapse
Affiliation(s)
- Ismail A. Ahmed
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jing-Jing Liu
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Krystyna A. Gieniec
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Chloe J. Bair-Marshall
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ayomiposi B. Adewakun
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Belinda E. Hetzler
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Christopher J. Arp
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Latika Khatri
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Gilles C. Vanwalleghem
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine. Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Alec T. Seidenberg
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Pamela Cowin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Dermatology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Dirk Trauner
- Department of Chemistry, New York University, 100 Washington Square East, New York, NY 10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses V. Chao
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10003, USA
| | - Felicity M. Davis
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Richard W. Tsien
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Robert C. Froemke
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Otolaryngology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Center for Neural Science, New York University, New York, NY, 10003, USA
| |
Collapse
|
18
|
D1 receptor-expressing neurons in ventral tegmental area alleviate mouse anxiety-like behaviors via glutamatergic projection to lateral septum. Mol Psychiatry 2023; 28:625-638. [PMID: 36195641 PMCID: PMC9531220 DOI: 10.1038/s41380-022-01809-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022]
Abstract
Dopamine (DA) acts as a key regulator in controlling emotion, and dysfunction of DA signal has been implicated in the pathophysiology of some psychiatric disorders, including anxiety. Ventral tegmental area (VTA) is one of main regions with DA-producing neurons. VTA DAergic projections in mesolimbic brain regions play a crucial role in regulating anxiety-like behaviors, however, the function of DA signal within VTA in regulating emotion remains unclear. Here, we observe that pharmacological activation/inhibition of VTA D1 receptors will alleviate/aggravate mouse anxiety-like behaviors, and knockdown of VTA D1 receptor expression also exerts anxiogenic effect. With fluorescence in situ hybridization and electrophysiological recording, we find that D1 receptors are functionally expressed in VTA neurons. Silencing/activating VTA D1 neurons bidirectionally modulate mouse anxiety-like behaviors. Furthermore, knocking down D1 receptors in VTA DA and glutamate neurons elevates anxiety-like state, but in GABA neurons has the opposite effect. In addition, we identify the glutamatergic projection from VTA D1 neurons to lateral septum is mainly responsible for the anxiolytic effect induced by activating VTA D1 neurons. Thus, our study not only characterizes the functional expression of D1 receptors in VTA neurons, but also uncovers the pivotal role of DA signal within VTA in mediating anxiety-like behaviors.
Collapse
|
19
|
A fluorescent sensor for real-time measurement of extracellular oxytocin dynamics in the brain. Nat Methods 2022; 19:1286-1294. [PMID: 36138174 PMCID: PMC9550624 DOI: 10.1038/s41592-022-01597-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/01/2022] [Indexed: 12/21/2022]
Abstract
Oxytocin (OT), a hypothalamic neuropeptide that acts as a neuromodulator in the brain, orchestrates a variety of animal behaviors. However, the relationship between brain OT dynamics and complex animal behaviors remains largely elusive, partly because of the lack of a suitable technique for its real-time recording in vivo. Here, we describe MTRIAOT, a G-protein-coupled receptor-based green fluorescent OT sensor that has a large dynamic range, suitable affinity, ligand specificity for OT orthologs, minimal effects on downstream signaling and long-term fluorescence stability. By combining viral gene delivery and fiber photometry-mediated fluorescence measurements, we demonstrate the utility of MTRIAOT for real-time detection of brain OT dynamics in living mice. MTRIAOT-mediated measurements indicate variability of OT dynamics depending on the behavioral context and physical condition of an animal. MTRIAOT will likely enable the analysis of OT dynamics in a variety of physiological and pathological processes.
Collapse
|
20
|
López-Gutiérrez MF, Mejía-Chávez S, Alcauter S, Portillo W. The neural circuits of monogamous behavior. Front Neural Circuits 2022; 16:978344. [PMID: 36247729 PMCID: PMC9559370 DOI: 10.3389/fncir.2022.978344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
The interest in studying the neural circuits related to mating behavior and mate choice in monogamous species lies in the parallels found between human social structure and sexual behavior and that of other mammals that exhibit social monogamy, potentially expanding our understanding of human neurobiology and its underlying mechanisms. Extensive research has suggested that social monogamy, as opposed to non-monogamy in mammals, is a consequence of the neural encoding of sociosensory information from the sexual partner with an increased reward value. Thus, the reinforced value of the mate outweighs the reward value of mating with any other potential sexual partners. This mechanism reinforces the social relationship of a breeding pair, commonly defined as a pair bond. In addition to accentuated prosocial behaviors toward the partner, other characteristic behaviors may appear, such as territorial and partner guarding, selective aggression toward unfamiliar conspecifics, and biparental care. Concomitantly, social buffering and distress upon partner separation are also observed. The following work intends to overview and compare known neural and functional circuits that are related to mating and sexual behavior in monogamous mammals. We will particularly discuss reports on Cricetid rodents of the Microtus and Peromyscus genus, and New World primates (NWP), such as the Callicebinae subfamily of the titi monkey and the marmoset (Callithrix spp.). In addition, we will mention the main factors that modulate the neural circuits related to social monogamy and how that modulation may reflect phenotypic differences, ultimately creating the widely observed diversity in social behavior.
Collapse
Affiliation(s)
| | | | | | - Wendy Portillo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| |
Collapse
|
21
|
Hu S, Wang Y, Han X, Dai M, Zhang Y, Ma Y, Weng S, Xiao L. Activation of oxytocin receptors in mouse GABAergic amacrine cells modulates retinal dopaminergic signaling. BMC Biol 2022; 20:205. [PMID: 36127701 PMCID: PMC9490981 DOI: 10.1186/s12915-022-01405-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
Background Oxytocin, secreted by oxytocin neurons in the hypothalamus, is an endogenous neuropeptide involved in modulating multiple sensory information processing pathways, and its roles in the brain have been associated with prosocial, maternal, and feeding-related behaviors. Visual information is necessary for initiating these behaviors, with the retina consisting of the first stage in the visual system mediating external stimulus perception. Oxytocin has been detected in the mammalian retina; however, the expression and possible function of oxytocin receptors (OxtR) in the retina remain unknown. Here, we explore the role of oxytocin in regulating visual information processing in the retina. Results We observed that OxtR mRNA and protein are expressed in the mouse retina. With Oxtr-Cre transgenic mice, immunostaining, and fluorescence in situ hybridization, we found that OxtRs are mainly expressed in GABAergic amacrine cells (ACs) in both the inner nuclear layer (INL) and ganglion cell layer (GCL). Further immunoreactivity studies showed that GABAergic OxtR+ neurons are mainly cholinergic and dopaminergic neurons in the INL and are cholinergic and corticotrophin-releasing hormone neurons in the GCL. Surprisingly, a high level of Oxtr mRNAs was detected in retinal dopaminergic neurons, and exogenous oxytocin application activated dopaminergic neurons to elevate the retinal dopamine level. Relying on in vivo electroretinographic recording, we found that activating retinal OxtRs reduced the activity of bipolar cells via OxtRs and dopamine receptors. Conclusions These data indicate the functional expression of OxtRs in retinal GABAergic ACs, especially dopaminergic ACs, and expand the interactions between oxytocinergic and dopaminergic systems. This study suggests that visual perception, from the first stage of information processing in the retina, is modulated by hypothalamic oxytocin signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01405-0.
Collapse
Affiliation(s)
- Songhui Hu
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yurong Wang
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xu Han
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Min Dai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yongxing Zhang
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shijun Weng
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
22
|
Rigney N, de Vries GJ, Petrulis A, Young LJ. Oxytocin, Vasopressin, and Social Behavior: From Neural Circuits to Clinical Opportunities. Endocrinology 2022; 163:bqac111. [PMID: 35863332 PMCID: PMC9337272 DOI: 10.1210/endocr/bqac111] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Indexed: 11/19/2022]
Abstract
Oxytocin and vasopressin are peptide hormones secreted from the pituitary that are well known for their peripheral endocrine effects on childbirth/nursing and blood pressure/urine concentration, respectively. However, both peptides are also released in the brain, where they modulate several aspects of social behaviors. Oxytocin promotes maternal nurturing and bonding, enhances social reward, and increases the salience of social stimuli. Vasopressin modulates social communication, social investigation, territorial behavior, and aggression, predominantly in males. Both peptides facilitate social memory and pair bonding behaviors in monogamous species. Here we review the latest research delineating the neural circuitry of the brain oxytocin and vasopressin systems and summarize recent investigations into the circuit-based mechanisms modulating social behaviors. We highlight research using modern molecular genetic technologies to map, monitor activity of, or manipulate neuropeptide circuits. Species diversity in oxytocin and vasopressin effects on social behaviors are also discussed. We conclude with a discussion of the translational implications of oxytocin and vasopressin for improving social functioning in disorders with social impairments, such as autism spectrum disorder.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Geert J de Vries
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
- Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA
| | - Aras Petrulis
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, Georgia 30303, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Emory University, Atlanta, Georgia 30329, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
23
|
Borie AM, Young LJ, Liu RC. Sex-specific and social experience-dependent oxytocin-endocannabinoid interactions in the nucleus accumbens: implications for social behaviour. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210057. [PMID: 35858094 PMCID: PMC9272148 DOI: 10.1098/rstb.2021.0057] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/13/2022] [Indexed: 08/31/2023] Open
Abstract
Oxytocin modulates social behaviour across diverse vertebrate taxa, but the precise nature of its effects varies across species, individuals and lifetimes. Contributing to this variation is the fact that oxytocin's physiological effects are mediated through interaction with diverse neuromodulatory systems and can depend on the specifics of the local circuits it acts on. Furthermore, those effects can be influenced by both genetics and experience. Here we discuss this complexity through the lens of a specific neuromodulatory system, endocannabinoids, interacting with oxytocin in the nucleus accumbens to modulate prosocial behaviours in prairie voles. We provide a survey of current knowledge of oxytocin-endocannabinoid interactions in relation to social behaviour. We review in detail recent research in monogamous female prairie voles demonstrating that social experience, such as mating and pair bonding, can change how oxytocin modulates nucleus accumbens glutamatergic signalling through the recruitment of endocannabinoids to modulate prosocial behaviour toward the partner. We then discuss potential sex differences in experience-dependent modulation of the nucleus accumbens by oxytocin in voles based on new data in males. Finally, we propose that future oxytocin-based precision medicine therapies should consider how prior social experience interacts with sex and genetics to influence oxytocin actions. This article is part of the theme issue 'Interplays between oxytocin and other neuromodulators in shaping complex social behaviours'.
Collapse
Affiliation(s)
- Amélie M. Borie
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Larry J. Young
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Robert C. Liu
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA
- Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
24
|
Dunigan AI, Roseberry AG. Actions of feeding-related peptides on the mesolimbic dopamine system in regulation of natural and drug rewards. ADDICTION NEUROSCIENCE 2022; 2:100011. [PMID: 37220637 PMCID: PMC10201992 DOI: 10.1016/j.addicn.2022.100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The mesolimbic dopamine system is the primary neural circuit mediating motivation, reinforcement, and reward-related behavior. The activity of this system and multiple behaviors controlled by it are affected by changes in feeding and body weight, such as fasting, food restriction, or the development of obesity. Multiple different peptides and hormones that have been implicated in the control of feeding and body weight interact with the mesolimbic dopamine system to regulate many different dopamine-dependent, reward-related behaviors. In this review, we summarize the effects of a selected set of feeding-related peptides and hormones acting within the ventral tegmental area and nucleus accumbens to alter feeding, as well as food, drug, and social reward.
Collapse
Affiliation(s)
- Anna I. Dunigan
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron G. Roseberry
- Department of Biology and Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
25
|
Canton-Josh JE, Qin J, Salvo J, Kozorovitskiy Y. Dopaminergic regulation of vestibulo-cerebellar circuits through unipolar brush cells. eLife 2022; 11:e76912. [PMID: 35476632 PMCID: PMC9106328 DOI: 10.7554/elife.76912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
While multiple monoamines modulate cerebellar output, the mechanistic details of dopaminergic signaling in the cerebellum remain poorly understood. We show that dopamine type 1 receptors (Drd1) are expressed in unipolar brush cells (UBCs) of the mouse cerebellar vermis. Drd1 activation increases UBC firing rate and post-synaptic NMDAR -mediated currents. Using anatomical tracing and in situ hybridization, we test three hypotheses about the source of cerebellar dopamine. We exclude midbrain dopaminergic nuclei and tyrosine hydroxylase-positive Purkinje (Pkj) cells as potential sources, supporting the possibility of dopaminergic co-release from locus coeruleus (LC) axons. Using an optical dopamine sensor GRABDA2h, electrical stimulation, and optogenetic activation of LC fibers in the acute slice, we find evidence for monoamine release onto Drd1-expressing UBCs. Altogether, we propose that the LC regulates cerebellar cortex activity by co-releasing dopamine onto UBCs to modulate their response to cerebellar inputs. Pkj cells directly inhibit these Drd1-positive UBCs, forming a dopamine-sensitive recurrent vestibulo-cerebellar circuit.
Collapse
Affiliation(s)
| | - Joanna Qin
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Joseph Salvo
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | | |
Collapse
|
26
|
Borie AM, Agezo S, Lunsford P, Boender AJ, Guo JD, Zhu H, Berman GJ, Young LJ, Liu RC. Social experience alters oxytocinergic modulation in the nucleus accumbens of female prairie voles. Curr Biol 2022; 32:1026-1037.e4. [PMID: 35108521 PMCID: PMC8930613 DOI: 10.1016/j.cub.2022.01.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/11/2021] [Accepted: 01/06/2022] [Indexed: 12/17/2022]
Abstract
Social relationships are dynamic and evolve with shared and personal experiences. Whether the functional role of social neuromodulators also evolves with experience to shape the trajectory of relationships is unknown. We utilized pair bonding in the socially monogamous prairie vole as an example of socio-sexual experience that dramatically alters behaviors displayed toward other individuals. We investigated oxytocin-dependent modulation of excitatory synaptic transmission in the nucleus accumbens as a function of pair-bonding status. We found that an oxytocin receptor agonist decreases the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) in sexually naive virgin, but not pair-bonded, female voles, while it increases the amplitude of electrically evoked EPSCs in paired voles, but not in virgins. This oxytocin-induced potentiation of synaptic transmission relies on the de novo coupling between oxytocin receptor signaling and endocannabinoid receptor type 1 (CB1) receptor signaling in pair-bonded voles. Blocking CB1 receptors after pair-bond formation increases the occurrence of a specific form of social rejection-defensive upright response-that is displayed toward the partner, but not toward a novel individual. Altogether, our results demonstrate that oxytocin's action in the nucleus accumbens is changed through social experience in a way that regulates the trajectory of social interactions as the relationship with the partner unfolds, potentially promoting the maintenance of a pair bond by inhibiting aggressive responses. These results provide a mechanism by which social experience and context shift oxytocinergic signaling to impact neural and behavioral responses to social cues.
Collapse
Affiliation(s)
- Amélie M Borie
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Sena Agezo
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Parker Lunsford
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Arjen J Boender
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
| | - Ji-Dong Guo
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA
| | - Hong Zhu
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Gordon J Berman
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA 30322, USA; Center for Social Neural Networks, University of Tsukuba, Tsukuba 305-8555, Japan.
| | - Robert C Liu
- Center for Translational Social Neuroscience, Emory University, Atlanta, GA 30322, USA; Silvio O. Conte Center for Oxytocin and Social Cognition, Emory University, Atlanta, GA 30322, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Neural circuit control of innate behaviors. SCIENCE CHINA. LIFE SCIENCES 2022; 65:466-499. [PMID: 34985643 DOI: 10.1007/s11427-021-2043-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022]
Abstract
All animals possess a plethora of innate behaviors that do not require extensive learning and are fundamental for their survival and propagation. With the advent of newly-developed techniques such as viral tracing and optogenetic and chemogenetic tools, recent studies are gradually unraveling neural circuits underlying different innate behaviors. Here, we summarize current development in our understanding of the neural circuits controlling predation, feeding, male-typical mating, and urination, highlighting the role of genetically defined neurons and their connections in sensory triggering, sensory to motor/motivation transformation, motor/motivation encoding during these different behaviors. Along the way, we discuss possible mechanisms underlying binge-eating disorder and the pro-social effects of the neuropeptide oxytocin, elucidating the clinical relevance of studying neural circuits underlying essential innate functions. Finally, we discuss some exciting brain structures recurrently appearing in the regulation of different behaviors, which suggests both divergence and convergence in the neural encoding of specific innate behaviors. Going forward, we emphasize the importance of multi-angle and cross-species dissections in delineating neural circuits that control innate behaviors.
Collapse
|
28
|
Martins D, Lockwood P, Cutler J, Moran R, Paloyelis Y. Oxytocin modulates neurocomputational mechanisms underlying prosocial reinforcement learning. Prog Neurobiol 2022; 213:102253. [DOI: 10.1016/j.pneurobio.2022.102253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 12/18/2022]
|
29
|
Oxytocin and Food Intake Control: Neural, Behavioral, and Signaling Mechanisms. Int J Mol Sci 2021; 22:ijms221910859. [PMID: 34639199 PMCID: PMC8509519 DOI: 10.3390/ijms221910859] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/03/2021] [Indexed: 11/16/2022] Open
Abstract
The neuropeptide oxytocin is produced in the paraventricular hypothalamic nucleus and the supraoptic nucleus of the hypothalamus. In addition to its extensively studied influence on social behavior and reproductive function, central oxytocin signaling potently reduces food intake in both humans and animal models and has potential therapeutic use for obesity treatment. In this review, we highlight rodent model research that illuminates various neural, behavioral, and signaling mechanisms through which oxytocin’s anorexigenic effects occur. The research supports a framework through which oxytocin reduces food intake via amplification of within-meal physiological satiation signals rather than by altering between-meal interoceptive hunger and satiety states. We also emphasize the distributed neural sites of action for oxytocin’s effects on food intake and review evidence supporting the notion that central oxytocin is communicated throughout the brain, at least in part, through humoral-like volume transmission. Finally, we highlight mechanisms through which oxytocin interacts with various energy balance-associated neuropeptide and endocrine systems (e.g., agouti-related peptide, melanin-concentrating hormone, leptin), as well as the behavioral mechanisms through which oxytocin inhibits food intake, including effects on nutrient-specific ingestion, meal size control, food reward-motivated responses, and competing motivations.
Collapse
|
30
|
Dumrongprechachan V, Salisbury RB, Soto G, Kumar M, MacDonald ML, Kozorovitskiy Y. Cell-type and subcellular compartment-specific APEX2 proximity labeling reveals activity-dependent nuclear proteome dynamics in the striatum. Nat Commun 2021; 12:4855. [PMID: 34381044 PMCID: PMC8357913 DOI: 10.1038/s41467-021-25144-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
The vertebrate brain consists of diverse neuronal types, classified by distinct anatomy and function, along with divergent transcriptomes and proteomes. Defining the cell-type specific neuroproteomes is important for understanding the development and functional organization of neural circuits. This task remains challenging in complex tissue, due to suboptimal protein isolation techniques that often result in loss of cell-type specific information and incomplete capture of subcellular compartments. Here, we develop a genetically targeted proximity labeling approach to identify cell-type specific subcellular proteomes in the mouse brain, confirmed by imaging, electron microscopy, and mass spectrometry. We virally express subcellular-localized APEX2 to map the proteome of direct and indirect pathway spiny projection neurons in the striatum. The workflow provides sufficient depth to uncover changes in the proteome of striatal neurons following chemogenetic activation of Gαq-coupled signaling cascades. This method enables flexible, cell-type specific quantitative profiling of subcellular proteome snapshots in the mouse brain. Mapping neuronal proteomes with genetic, subcellular, and temporal specificity is a challenging task. This study uncovers proteome dynamics in two classes of striatal spiny projection neurons in the mouse brain using a genetically targeted APEX2-based proximity labeling approach.
Collapse
Affiliation(s)
- V Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.,The Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - R B Salisbury
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.,Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - G Soto
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - M Kumar
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - M L MacDonald
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA. .,Biomedical Mass Spectrometry Center, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Y Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA. .,The Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
31
|
Wireless, battery-free, subdermally implantable platforms for transcranial and long-range optogenetics in freely moving animals. Proc Natl Acad Sci U S A 2021; 118:2025775118. [PMID: 34301889 DOI: 10.1073/pnas.2025775118] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Wireless, battery-free, and fully subdermally implantable optogenetic tools are poised to transform neurobiological research in freely moving animals. Current-generation wireless devices are sufficiently small, thin, and light for subdermal implantation, offering some advantages over tethered methods for naturalistic behavior. Yet current devices using wireless power delivery require invasive stimulus delivery, penetrating the skull and disrupting the blood-brain barrier. This can cause tissue displacement, neuronal damage, and scarring. Power delivery constraints also sharply curtail operational arena size. Here, we implement highly miniaturized, capacitive power storage on the platform of wireless subdermal implants. With approaches to digitally manage power delivery to optoelectronic components, we enable two classes of applications: transcranial optogenetic activation millimeters into the brain (validated using motor cortex stimulation to induce turning behaviors) and wireless optogenetics in arenas of more than 1 m2 in size. This methodology allows for previously impossible behavioral experiments leveraging the modern optogenetic toolkit.
Collapse
|
32
|
Yang Y, Wu M, Vázquez-Guardado A, Wegener AJ, Grajales-Reyes JG, Deng Y, Wang T, Avila R, Moreno JA, Minkowicz S, Dumrongprechachan V, Lee J, Zhang S, Legaria AA, Ma Y, Mehta S, Franklin D, Hartman L, Bai W, Han M, Zhao H, Lu W, Yu Y, Sheng X, Banks A, Yu X, Donaldson ZR, Gereau RW, Good CH, Xie Z, Huang Y, Kozorovitskiy Y, Rogers JA. Wireless multilateral devices for optogenetic studies of individual and social behaviors. Nat Neurosci 2021; 24:1035-1045. [PMID: 33972800 PMCID: PMC8694284 DOI: 10.1038/s41593-021-00849-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/26/2021] [Indexed: 12/31/2022]
Abstract
Advanced technologies for controlled delivery of light to targeted locations in biological tissues are essential to neuroscience research that applies optogenetics in animal models. Fully implantable, miniaturized devices with wireless control and power-harvesting strategies offer an appealing set of attributes in this context, particularly for studies that are incompatible with conventional fiber-optic approaches or battery-powered head stages. Limited programmable control and narrow options in illumination profiles constrain the use of existing devices. The results reported here overcome these drawbacks via two platforms, both with real-time user programmability over multiple independent light sources, in head-mounted and back-mounted designs. Engineering studies of the optoelectronic and thermal properties of these systems define their capabilities and key design considerations. Neuroscience applications demonstrate that induction of interbrain neuronal synchrony in the medial prefrontal cortex shapes social interaction within groups of mice, highlighting the power of real-time subject-specific programmability of the wireless optogenetic platforms introduced here.
Collapse
Affiliation(s)
- Yiyuan Yang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | | | - Amy J Wegener
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Jose G Grajales-Reyes
- Washington University Pain Center and Department of Anesthesiology, Washington University, St. Louis, MO, USA
| | - Yujun Deng
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai, China
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Taoyi Wang
- Department of Physics, Tsinghua University, Beijing, China
| | - Raudel Avila
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Justin A Moreno
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
- SURVICE Engineering, Belcamp, MD, USA
| | - Samuel Minkowicz
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA
| | | | - Shuangyang Zhang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- School of Civil Engineering, Southwest JiaoTong University, Chengdu, China
| | - Alex A Legaria
- Washington University Pain Center and Department of Anesthesiology, Washington University, St. Louis, MO, USA
| | - Yuhang Ma
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, People's Republic of China
| | - Sunita Mehta
- CSIR-Central Scientific Instruments Organization, Ministry of Science & Technology, Sector 30-C, Chandigarh, India
| | - Daniel Franklin
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Layne Hartman
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Wubin Bai
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Mengdi Han
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Hangbo Zhao
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Wei Lu
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Yongjoon Yu
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA
| | - Xing Sheng
- Department of Electronic Engineering, Tsinghua University, Beijing, China
| | - Anthony Banks
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Neurolux Inc, Evanston, IL, USA
- Simpson Querrey Institute & Feinberg Medical School, Northwestern University, Evanston, IL, USA
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloong Tong, Hong Kong
| | - Zoe R Donaldson
- Psychology and Neuroscience, Molecular Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University, St. Louis, MO, USA
| | - Cameron H Good
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Zhaoqian Xie
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, P.R. China.
| | - Yonggang Huang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA.
| | - John A Rogers
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Neurolux Inc, Evanston, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute & Feinberg Medical School, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Northwestern University, Evanston, IL, USA.
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA.
- Department of Computer Science, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
33
|
Xie S, Hu Y, Fang L, Chen S, Botchway BOA, Tan X, Fang M, Hu Z. The association of oxytocin with major depressive disorder: role of confounding effects of antidepressants. Rev Neurosci 2021; 33:59-77. [PMID: 33989469 DOI: 10.1515/revneuro-2020-0128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 04/18/2021] [Indexed: 01/15/2023]
Abstract
Major depressive disorder is a genetic susceptible disease, and a psychiatric syndrome with a high rate of incidence and recurrence. Because of its complexity concerning etiology and pathogenesis, the cure rate of first-line antidepressants is low. In recent years, accumulative evidences revealed that oxytocin act as a physiological or pathological participant in a variety of complex neuropsychological activities, including major depressive disorder. Six electronic databases (Web of Science, PubMed, Scopus, Google Scholar, CNKI, and Wanfang) were employed for researching relevant publications. At last, 226 articles were extracted. The current review addresses the correlation of the oxytocin system and major depressive disorder. Besides, we summarize the mechanisms by which the oxytocin system exerts potential antidepressant effects, including regulating neuronal activity, influencing neuroplasticity and regeneration, altering neurotransmitter release, down regulating hypothalamic-pituitary-adrenal axis, anti-inflammatory, antioxidation, and genetic effects. Increasing evidence shows that oxytocin and its receptor gene may play a potential role in major depressive disorder. Future research should focus on the predictive ability of the oxytocin system as a biomarker, as well as its role in targeted prevention and early intervention of major depressive disorder.
Collapse
Affiliation(s)
- Shiyi Xie
- Obstetrics & Gynecology Department, Integrated Chinese and West Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, 208 Huanchendong Road, 310003Hangzhou, China.,Clinical Medical College, Zhejiang Chinese Medical University, 310053Hangzhou, China
| | - Yan Hu
- Clinical Medical College, Zhejiang Chinese Medical University, 310053Hangzhou, China
| | - Li Fang
- Obstetrics & Gynecology Department, Integrated Chinese and West Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, 208 Huanchendong Road, 310003Hangzhou, China
| | - Shijia Chen
- Institute of Neuroscience, Zhejiang University School of Medicine, 310058Hangzhou, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, 310058Hangzhou, China
| | - Xiaoning Tan
- Institute of Neuroscience, Zhejiang University School of Medicine, 310058Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, 310058Hangzhou, China
| | - Zhiying Hu
- Obstetrics & Gynecology Department, Integrated Chinese and West Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, 208 Huanchendong Road, 310003Hangzhou, China
| |
Collapse
|
34
|
Hoffman AF, Hwang EK, Lupica CR. Impairment of Synaptic Plasticity by Cannabis, Δ 9-THC, and Synthetic Cannabinoids. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a039743. [PMID: 32341064 PMCID: PMC8091957 DOI: 10.1101/cshperspect.a039743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability of neurons to dynamically and flexibly encode synaptic inputs via short- and long-term plasticity is critical to an organism's ability to learn and adapt to the environment. Whereas synaptic plasticity may be encoded by pre- or postsynaptic mechanisms, current evidence suggests that optimization of learning requires both forms of plasticity. Endogenous cannabinoids (eCBs) play critical roles in modulating synaptic transmission via activation of cannabinoid CB1 receptors (CB1Rs) in many central nervous system (CNS) regions, and the eCB system has been implicated, either directly or indirectly, in several forms of synaptic plasticity. Because of this, perturbations within the eCB signaling system can lead to impairments in a variety of learned behaviors. One agent of altered eCB signaling is exposure to "exogenous cannabinoids" such as the primary psychoactive constituent of cannabis, Δ9-THC, or illicit synthetic cannabinoids that in many cases have higher potency and efficacy than Δ9-THC. Thus, by targeting the eCB system, these agonists can produce widespread impairment of synaptic plasticity by disrupting ongoing eCB function. Here, we review studies in which Δ9-THC and synthetic cannabinoids impair synaptic plasticity in a variety of neuronal circuits and examine evidence that this contributes to their well-documented ability to disrupt cognition and behavior.
Collapse
Affiliation(s)
- Alexander F Hoffman
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Eun-Kyung Hwang
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Carl R Lupica
- Electrophysiology Research Section, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
35
|
Wu M, Minkowicz S, Dumrongprechachan V, Hamilton P, Xiao L, Kozorovitskiy Y. Attenuated dopamine signaling after aversive learning is restored by ketamine to rescue escape actions. eLife 2021; 10:64041. [PMID: 33904412 PMCID: PMC8211450 DOI: 10.7554/elife.64041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Escaping aversive stimuli is essential for complex organisms, but prolonged exposure to stress leads to maladaptive learning. Stress alters neuronal activity and neuromodulatory signaling in distributed networks, modifying behavior. Here, we describe changes in dopaminergic neuron activity and signaling following aversive learning in a learned helplessness paradigm in mice. A single dose of ketamine suffices to restore escape behavior after aversive learning. Dopaminergic neuron activity in the ventral tegmental area (VTA) systematically varies across learning, correlating with future sensitivity to ketamine treatment. Ketamine’s effects are blocked by chemogenetic inhibition of dopamine signaling. Rather than directly altering the activity of dopaminergic neurons, ketamine appears to rescue dopamine dynamics through actions in the medial prefrontal cortex (mPFC). Chemogenetic activation of Drd1 receptor positive mPFC neurons mimics ketamine’s effects on behavior. Together, our data link neuromodulatory dynamics in mPFC-VTA circuits, aversive learning, and the effects of ketamine. Over 264 million people around the world suffer from depression, according to the World Health Organization (WHO). Depression can be debilitating, and while anti-depressant drugs are available, they do not always work. A small molecule drug mainly used for anesthesia called ketamine has recently been shown to ameliorate depressive symptoms within hours, much faster than most anti-depressants. However, the molecular mechanisms behind this effect are still largely unknown. Most anti-depressant drugs work by restoring the normal balance of dopamine and other chemical messengers in the brain. Dopamine is released by a specialized group of cells called dopaminergic neurons, and helps us make decisions by influencing a wide range of other cells in the brain. In a healthy brain, dopamine directs us to rewarding choices, while avoiding actions with negative outcomes. During depression, these dopamine signals are perturbed, resulting in reduced motivation and pleasure. But it remained unclear whether ketamine’s anti-depressant activity also relied on dopamine. To investigate this, Wu et al. used a behavioral study called “learned helplessness” which simulates depression by putting mice in unavoidable stressful situations. Over time the mice learn that their actions do not change the outcome and eventually stop trying to escape from unpleasant situations, even if they are avoidable. The experiment showed that dopaminergic neurons in an area of the brain that is an important part of the “reward and aversion” system became less sensitive to unpleasant stimuli following learned helplessness. When the mice received ketamine, these neurons recovered after a few hours. Individual mice also responded differently to ketamine. The most ‘resilient’, stress-resistant mice, which had distinct patterns of dopamine signaling, also responded most strongly to the drug. Genetic and chemical manipulation of dopaminergic neurons confirmed that ketamine needed intact dopamine signals to work, and revealed that it acted indirectly on dopamine dynamics via another brain region called the medial prefrontal cortex. These results shed new light on how a promising new anti-depressant works. In the future, they may also explain why drugs like ketamine work better for some people than others, ultimately helping clinicians select the most effective treatment for individual patients.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Samuel Minkowicz
- Department of Neurobiology, Northwestern University, Evanston, United States
| | | | - Pauline Hamilton
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Lei Xiao
- Department of Neurobiology, Northwestern University, Evanston, United States
| | | |
Collapse
|
36
|
Abstract
Oxytocin regulates parturition, lactation, parental nurturing, and many other social behaviors in both sexes. The circuit mechanisms by which oxytocin modulates social behavior are receiving increasing attention. Here, we review recent studies on oxytocin modulation of neural circuit function and social behavior, largely enabled by new methods of monitoring and manipulating oxytocin or oxytocin receptor neurons in vivo. These studies indicate that oxytocin can enhance the salience of social stimuli and increase signal-to-noise ratios by modulating spiking and synaptic plasticity in the context of circuits and networks. We highlight oxytocin effects on social behavior in nontraditional organisms such as prairie voles and discuss opportunities to enhance the utility of these organisms for studying circuit-level modulation of social behaviors. We then discuss recent insights into oxytocin neuron activity during social interactions. We conclude by discussing some of the major questions and opportunities in the field ahead.
Collapse
Affiliation(s)
- Robert C Froemke
- Skirball Institute, Neuroscience Institute, and Departments of Otolaryngology and Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, USA; .,Center for Neural Science, New York University, New York, NY 10003, USA
| | - Larry J Young
- Silvio O. Conte Center for Oxytocin and Social Cognition, Center for Translational Social Neuroscience, and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA.,Center for Social Neural Networks, Faculty of Human Sciences, University of Tsukuba, Tsukuba 305-8577, Japan.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
37
|
Sundar M, Patel D, Young Z, Leong KC. Oxytocin and Addiction: Potential Glutamatergic Mechanisms. Int J Mol Sci 2021; 22:ijms22052405. [PMID: 33673694 PMCID: PMC7957657 DOI: 10.3390/ijms22052405] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022] Open
Abstract
Recently, oxytocin (OXT) has been investigated for its potential therapeutic role in addiction. OXT has been found to diminish various drug-seeking and drug-induced behaviors. Although its behavioral effects are well-established, there is not much consensus on how this neuropeptide exerts its effects. Previous research has given thought to how dopamine (DA) may be involved in oxytocinergic mechanisms, but there has not been as strong of a focus on the role that glutamate (Glu) has. The glutamatergic system is critical for the processing of rewards and the disruption of glutamatergic projections produces the behaviors seen in drug addicts. We introduce the idea that OXT has direct effects on Glu transmission within the reward processing pathway. Thus, OXT may reduce addictive behaviors by restoring abnormal drug-induced changes in the glutamatergic system and in its interactions with other neurotransmitters. This review offers insight into the mechanisms through which a potentially viable therapeutic target, OXT, could be used to reduce addiction-related behaviors.
Collapse
|
38
|
Salles J, Lacassagne E, Eddiry S, Franchitto N, Salles JP, Tauber M. What can we learn from PWS and SNORD116 genes about the pathophysiology of addictive disorders? Mol Psychiatry 2021; 26:51-59. [PMID: 33082508 DOI: 10.1038/s41380-020-00917-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
Addictive disorders have been much investigated and many studies have underlined the role of environmental factors such as social interaction in the vulnerability to and maintenance of addictive behaviors. Research on addiction pathophysiology now suggests that certain behavioral disorders are addictive, one example being food addiction. Yet, despite the growing body of knowledge on addiction, it is still unknown why only some of the individuals exposed to a drug become addicted to it. This observation has prompted the consideration of genetic heritage, neurodevelopmental trajectories, and gene-environment interactions in addiction vulnerability. Prader-Willi syndrome (PWS) is a rare neurodevelopmental disorder in which children become addicted to food and show early social impairment. PWS is caused by the deficiency of imprinted genes located on the 15q11-q13 chromosome. Among them, the SNORD116 gene was identified as the minimal gene responsible for the PWS phenotype. Several studies have also indicated the role of the Snord116 gene in animal and cellular models to explain PWS pathophysiology and phenotype (including social impairment and food addiction). We thus present here the evidence suggesting the potential involvement of the SNORD116 gene in addictive disorders.
Collapse
Affiliation(s)
- Juliette Salles
- Université de Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Service de psychiatrie et psychologie, psychiatrie Toulouse, F-31000, Toulouse, France.,Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Institut des Handicaps Neurologiques, Psychiatriques et Sensoriels, F-31000, Toulouse, France
| | - Emmanuelle Lacassagne
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Sanaa Eddiry
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Nicolas Franchitto
- Université de Toulouse III, F-31000, Toulouse, France.,CHU de Toulouse, Service d'addictologie clinique, urgences réanimation médecine, F-31000, Toulouse, France
| | - Jean-Pierre Salles
- Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France
| | - Maithé Tauber
- Université de Toulouse III, F-31000, Toulouse, France. .,Inserm Unité 1043, CNRS 5828, Université Paul Sabatier, Toulouse III, F-31000, Toulouse, France. .,CHU de Toulouse, Institut des Handicaps Neurologiques, Psychiatriques et Sensoriels, F-31000, Toulouse, France. .,CHU de Toulouse, Centre de référence du Syndrome de Prader-Willi et autres syndromes avec troubles du comportement alimentaire, Unité d'endocrinologie, obésités, maladies osseuses, génétique et gynécologie médicale, F-31000, Toulouse, France.
| |
Collapse
|
39
|
Arakawa H. Dynamic regulation of oxytocin neuronal circuits in the sequential processes of prosocial behavior in rodent models. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2:100011. [PMID: 36246512 PMCID: PMC9559098 DOI: 10.1016/j.crneur.2021.100011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 12/16/2022] Open
|
40
|
Liu CM, Hsu TM, Suarez AN, Subramanian KS, Fatemi RA, Cortella AM, Noble EE, Roitman MF, Kanoski SE. Central oxytocin signaling inhibits food reward-motivated behaviors and VTA dopamine responses to food-predictive cues in male rats. Horm Behav 2020; 126:104855. [PMID: 32991888 PMCID: PMC7757852 DOI: 10.1016/j.yhbeh.2020.104855] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/02/2020] [Accepted: 08/23/2020] [Indexed: 01/08/2023]
Abstract
Oxytocin potently reduces food intake and is a potential target system for obesity treatment. A better understanding of the behavioral and neurobiological mechanisms mediating oxytocin's anorexigenic effects may guide more effective obesity pharmacotherapy development. The present study examined the effects of central (lateral intracerebroventricular [ICV]) administration of oxytocin in rats on motivated responding for palatable food. Various conditioning procedures were employed to measure distinct appetitive behavioral domains, including food seeking in the absence of consumption (conditioned place preference expression), impulsive responding for food (differential reinforcement of low rates of responding), effort-based appetitive decision making (high-effort palatable vs. low-effort bland food), and sucrose reward value encoding following a motivational shift (incentive learning). Results reveal that ICV oxytocin potently reduces food-seeking behavior, impulsivity, and effort-based palatable food choice, yet does not influence encoding of sucrose reward value in the incentive learning task. To investigate a potential neurobiological mechanism mediating these behavioral outcomes, we utilized in vivo fiber photometry in ventral tegmental area (VTA) dopamine neurons to examine oxytocin's effect on phasic dopamine neuron responses to sucrose-predictive Pavlovian cues. Results reveal that ICV oxytocin significantly reduced food cue-evoked dopamine neuron activity. Collectively, these data reveal that central oxytocin signaling inhibits various obesity-relevant conditioned appetitive behaviors, potentially via reductions in food cue-driven phasic dopamine neural responses in the VTA.
Collapse
Affiliation(s)
- Clarissa M Liu
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States
| | - Ted M Hsu
- Department of Psychology, University of Illinois at Chicago, 1007 W. Harrison St., Chicago, IL 60607-7137, United States
| | - Andrea N Suarez
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States
| | - Keshav S Subramanian
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States
| | - Ryan A Fatemi
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States
| | - Alyssa M Cortella
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States
| | - Emily E Noble
- Department of Foods and Nutrition, University of Georgia, 129 Barrow Hall, Athens, GA 30602, United States
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois at Chicago, 1007 W. Harrison St., Chicago, IL 60607-7137, United States
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States; Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, 3616 Trousdale Parkway, AHF 252, Los Angeles, CA 90089, United States.
| |
Collapse
|
41
|
Shemetov AA, Monakhov MV, Zhang Q, Canton-Josh JE, Kumar M, Chen M, Matlashov ME, Li X, Yang W, Nie L, Shcherbakova DM, Kozorovitskiy Y, Yao J, Ji N, Verkhusha VV. A near-infrared genetically encoded calcium indicator for in vivo imaging. Nat Biotechnol 2020; 39:368-377. [PMID: 33106681 PMCID: PMC7956128 DOI: 10.1038/s41587-020-0710-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 09/18/2020] [Indexed: 12/18/2022]
Abstract
While calcium imaging has become a mainstay of modern neuroscience, the spectral properties of current fluorescent calcium indicators limit deep tissue imaging as well as simultaneous use with other probes. Using two monomeric near-infrared fluorescent proteins, we engineered a near-infrared FRET-based genetically encoded calcium indicator (iGECI). iGECI exhibits high brightness, high photostability, and up to 600% increase in fluorescence response to calcium. In dissociated neurons, iGECI detects spontaneous neuronal activity, and electrically and optogenetically induced firing. We validated iGECI performance up to a depth of almost 400 μm in acute brain slices using one-photon light-sheet imaging. Applying hybrid photoacoustic and fluorescence microscopy, we simultaneously monitored neuronal and hemodynamic activities in the mouse brain through an intact skull, with ~3 μm lateral and ~25–50 μm axial resolution. Using two-photon imaging, we detected evoked and spontaneous neuronal activity in the mouse visual cortex, with fluorescence changes of up to 25%. iGECI allows biosensors and optogenetic actuators to be multiplexed without spectral crosstalk.
Collapse
Affiliation(s)
- Anton A Shemetov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Autonomous Therapeutics, Inc., New York, NY, USA
| | - Mikhail V Monakhov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qinrong Zhang
- Department of Physics, University of California, Berkeley, Berkeley, CA, USA
| | - Jose Ernesto Canton-Josh
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Manish Kumar
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Maomao Chen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.,Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Mikhail E Matlashov
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xuan Li
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Wei Yang
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Liming Nie
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China.,Department of Radiology and Optical Imaging Laboratory, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Daria M Shcherbakova
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Weinberg School of Arts and Sciences, Northwestern University, Evanston, IL, USA.,Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Junjie Yao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA, USA
| | - Vladislav V Verkhusha
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
42
|
Wald HS, Chandra A, Kalluri A, Ong ZY, Hayes MR, Grill HJ. NTS and VTA oxytocin reduces food motivation and food seeking. Am J Physiol Regul Integr Comp Physiol 2020; 319:R673-R683. [PMID: 33026822 DOI: 10.1152/ajpregu.00201.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oxytocin (OT) is a neuropeptide whose central receptor-mediated actions include reducing food intake. One mechanism of its behavioral action is the amplification of the feeding inhibitory effects of gastrointestinal (GI) satiation signals processed by hindbrain neurons. OT treatment also reduces carbohydrate intake in humans and rodents, and correspondingly, deficits in central OT receptor (OT-R) signaling increase sucrose self-administration. This suggests that additional processes contribute to central OT effects on feeding. This study investigated the hypothesis that central OT reduces food intake by decreasing food seeking and food motivation. As central OT-Rs are expressed widely, a related focus was to assess the role of one or more OT-R-expressing nuclei in food motivation and food-seeking behavior. OT was delivered to the lateral ventricle (LV), nucleus tractus solitarius (NTS), or ventral tegmental area (VTA), and a progressive ratio (PR) schedule of operant reinforcement and an operant reinstatement paradigm were used to measure motivated feeding behavior and food-seeking behavior, respectively. OT delivered to the LV, NTS, or VTA reduced 1) motivation to work for food and 2) reinstatement of food-seeking behavior. Results provide a novel and additional interpretation for central OT-driven food intake inhibition to include the reduction of food motivation and food seeking.
Collapse
Affiliation(s)
- Hallie S Wald
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ananya Chandra
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Kalluri
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhi Yi Ong
- School of Psychology, University of New South Wales, Sydney, Australia
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Harvey J Grill
- Department of Psychology, Institute of Diabetes Obesity and Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
43
|
Chen C, Soto G, Dumrongprechachan V, Bannon N, Kang S, Kozorovitskiy Y, Parisiadou L. Pathway-specific dysregulation of striatal excitatory synapses by LRRK2 mutations. eLife 2020; 9:58997. [PMID: 33006315 PMCID: PMC7609054 DOI: 10.7554/elife.58997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022] Open
Abstract
LRRK2 is a kinase expressed in striatal spiny projection neurons (SPNs), cells which lose dopaminergic input in Parkinson’s disease (PD). R1441C and G2019S are the most common pathogenic mutations of LRRK2. How these mutations alter the structure and function of individual synapses on direct and indirect pathway SPNs is unknown and may reveal pre-clinical changes in dopamine-recipient neurons that predispose toward disease. Here, R1441C and G2019S knock-in mice enabled thorough evaluation of dendritic spines and synapses on pathway-identified SPNs. Biochemical synaptic preparations and super-resolution imaging revealed increased levels and altered organization of glutamatergic AMPA receptors in LRRK2 mutants. Relatedly, decreased frequency of miniature excitatory post-synaptic currents accompanied changes in dendritic spine nano-architecture, and single-synapse currents, evaluated using two-photon glutamate uncaging. Overall, LRRK2 mutations reshaped synaptic structure and function, an effect exaggerated in R1441C dSPNs. These data open the possibility of new neuroprotective therapies aimed at SPN synapse function, prior to disease onset. Parkinson’s disease is caused by progressive damage to regions of the brain that regulate movement. This leads to a loss in nerve cells that produce a signaling molecule called dopamine, and causes patients to experience shakiness, slow movement and stiffness. When dopamine is released, it travels to a part of the brain known as the striatum, where it is received by cells called spiny projection neurons (SPNs), which are rich in a protein called LRRK2. Mutations in this protein have been shown to cause the motor impairments associated with Parkinson’s disease. SPNs send signals to other regions of the brain either via a ‘direct’ route, which promotes movement, or an ‘indirect’ route, which suppresses movement. Previous studies suggest that mutations in the gene for LRRK2 influence the activity of these pathways even before dopamine signaling has been lost. Yet, it remained unclear how different mutations independently affected each pathway. To investigate this further, Chen et al. studied two of the mutations most commonly found in the human gene for LRRK2, known as G2019S and R1441C. This involved introducing one of these mutations in to the genetic code of mice, and using fluorescent proteins to mark single SPNs in either the direct or indirect pathway. The experiments showed that both mutations disrupted the connections between SPNs in the direct and indirect pathway, which altered the activity of nerve cells in the striatum. Chen et al. found that individual connections were more strongly affected by the R1441C mutation. Further experiments showed that this was caused by the re-organization of a receptor protein in the nerve cells of the direct pathway, which increased how SPNs responded to inputs from other nerve cells. These findings suggest that LRRK2 mutations disrupt neural activity in the striatum before dopamine levels become depleted. This discovery could help researchers identify new therapies for treating the early stages of Parkinson’s disease before the symptoms of dopamine loss arise.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Giulia Soto
- Department of Neurobiology, Northwestern University, Chicago, United States
| | | | - Nicholas Bannon
- Department of Neurobiology, Northwestern University, Chicago, United States
| | - Shuo Kang
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | | | - Loukia Parisiadou
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
44
|
Abramova O, Zorkina Y, Ushakova V, Zubkov E, Morozova A, Chekhonin V. The role of oxytocin and vasopressin dysfunction in cognitive impairment and mental disorders. Neuropeptides 2020; 83:102079. [PMID: 32839007 DOI: 10.1016/j.npep.2020.102079] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023]
Abstract
Oxytocin (OXT) and arginine-vasopressin (AVP) are structurally homologous peptide hormones synthesized in the hypothalamus. Nowadays, the role of OXT and AVP in the regulation of social behaviour and emotions is generally known. However, recent researches indicate that peptides also participate in cognitive functioning. This review presents the evidence that the OXT/AVP systems are involved in the formation of social, working, spatial and episodic memory, mediated by such brain structures as the hippocampal CA2 and CA3 regions, amygdala and prefrontal cortex. Some data have demonstrated that the OXT receptor's polymorphisms are associated with impaired memory in humans, and OXT knockout in mice is connected with memory deficit. Additionally, OXT and AVP are involved in mental disorders' progression. Stress-induced imbalance of the OXT/AVP systems leads to an increased risk of various mental disorders, including depression, schizophrenia, and autism. At the same time, cognitive deficits are observed in stress and mental disorders, and perhaps peptide hormones play a part in this. The final part of the review describes possible therapeutic strategies for the use of OXT and AVP for treatment of various mental disorders.
Collapse
Affiliation(s)
- Olga Abramova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia.
| | - Yana Zorkina
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Valeria Ushakova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia; Department of Biology, Lomonosov Moscow State University, Russia
| | - Eugene Zubkov
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Anna Morozova
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V.P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia; Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
45
|
Chen ZY, Xie DF, Liu ZY, Zhong YQ, Zeng JY, Chen Z, Chen XL. Identification of the significant pathways of Banxia Houpu decoction in the treatment of depression based on network pharmacology. PLoS One 2020; 15:e0239843. [PMID: 32997725 PMCID: PMC7527207 DOI: 10.1371/journal.pone.0239843] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Banxia Houpu decoction (BXHPD) has been used to treat depression in clinical practice for centuries. However, the pharmacological mechanisms of BXHPD still remain unclear. Network Pharmacology (NP) approach was used to explore the potential molecular mechanisms of BXHPD in treating depression. Potential active compounds of BXHPD were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform Database. STRING database was used to build a interaction network between the active compounds and target genes associated with depression. The topological features of nodes were visualized and calculated. Significant pathways and biological functions were identified using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. A total of 44 active compounds were obtained from BXHPD, and 121 potential target genes were considered to be therapeutically relevant. Pathway analysis indicated that MAPK signaling pathway, ErbB signaling pathway, HIF-1 signaling pathway and PI3K-Akt pathway were significant pathways in depression. They were mainly involved in promoting nerve growth and nutrition and alleviating neuroinflammatory conditions. The result provided some potential ways for modern medicine in the treatment of depression.
Collapse
Affiliation(s)
- Zi-ying Chen
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan-feng Xie
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi-yuan Liu
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong-qi Zhong
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing-yan Zeng
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zheng Chen
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- * E-mail: (XLC); (ZC)
| | - Xin-lin Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
- * E-mail: (XLC); (ZC)
| |
Collapse
|
46
|
Cheng Z, Cui R, Ge T, Yang W, Li B. Optogenetics: What it has uncovered in potential pathways of depression. Pharmacol Res 2020; 152:104596. [DOI: 10.1016/j.phrs.2019.104596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/29/2019] [Accepted: 12/11/2019] [Indexed: 01/07/2023]
|
47
|
Weaver KR, Melkus GD, Fletcher J, Henderson WA. Relevance of Sex and Subtype in Patients With IBS: An Exploratory Study of Gene Expression. Biol Res Nurs 2020; 22:13-23. [PMID: 31833409 PMCID: PMC7068753 DOI: 10.1177/1099800419889189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Psychological state, stress level, and gastrointestinal function are intricately related and relevant to symptom exacerbation in patients with irritable bowel syndrome (IBS), but genetic contributors to this brain-gut connection are not fully understood. The purpose of this exploratory study was to compare gene expression in participants with IBS to that of healthy controls (HC) and to examine patterns of expression in participants with IBS by sex and IBS subtype. METHOD Participants were recruited to an ongoing protocol at the National Institutes of Health. Differences in demographic and clinical characteristics were assessed using descriptive statistics and Mann-Whitney U tests. Expression levels of 84 genes were evaluated in peripheral whole blood using Custom RT2 Profiler polymerase chain reaction (PCR) Arrays, and data analysis was performed through GeneGlobe Data Analysis Center. RESULTS Participants with IBS (n = 27) reported greater levels of perceived stress (p = .037) and differed in expression values of ±2 for the genes ADIPOR1, ADIPOR2, CNR2, COMT, OXTR, and PPARA compared to HC (n = 43). Further analyses by sex and IBS subtype revealed differential patterns of gene expression related to the endocannabinoid system, cytokines, stress, and sex steroid hormones. CONCLUSIONS Diverse yet interconnected processes such as metabolism, inflammation, immunity, social behavior, and pain are associated with differences in gene expression between participants with IBS and HC. These findings lend support for genomic associations with the brain-gut connection in patients with IBS and highlight the relevance of sex and IBS subtype in performing such analyses.
Collapse
Affiliation(s)
- Kristen R. Weaver
- Department of Pain and Translational Symptom Science, University of Maryland
School of Nursing, Baltimore, MD, USA
- National Institute of Nursing Research, National Institutes of Health,
Bethesda, MD, USA
| | | | - Jason Fletcher
- Rory Meyers College of Nursing, New York University, NY, USA
| | - Wendy A. Henderson
- Digestive Disorders Unit, Division of Intramural Research, National
Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Leng H, Zhang X, Wang Q, Luan X, Sun X, Guo F, Gao S, Liu X, Xu L. Regulation of stress-induced gastric ulcers via central oxytocin and a potential mechanism through the VTA-NAc dopamine pathway. Neurogastroenterol Motil 2019; 31:e13655. [PMID: 31172654 DOI: 10.1111/nmo.13655] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/29/2019] [Accepted: 05/24/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Oxytocin (OT) plays an important role in regulating gastric function. How OT regulates stress-induced gastric ulcers is not understood. We investigated OT's protective role in stress-induced gastric ulcers, with a focus on OT's interaction with the ventral tegmental area (VTA) to nucleus accumbens (NAc) dopamine pathway. METHODS Drugs administration into the rats brain nuclei by brain stereotaxic apparatus, to examine related changes in gastric ulcer index, pH of gastric content, and mucus secretion, and to determine complex interactions between OT and DA systems in the regulation of stress and gastric functions. KEY RESULTS Neurons in the VTA were co-immunoreactive for the OT receptor (OTR) and DA. In a rat model of stress-induced ulcer, water-immersion restricted stress, direct administration of OT into the VTA significantly reduced gastric ulcer index and increased the pH of gastric content and mucus secretion. OT's effects were eliminated by pretreatment with the OTR antagonist atosiban in the VTA and weakened with pretreatment of the DA D2 receptor (DA D2R) antagonist raclopride in the NAc. In OTR gene knockout (Oxtr-/- ) mice, OT's protective effect was lost. OT administered to the VTA of dorsal motor nucleus of the vagus (DMV)-lesioned rats had minimal protective effects on gastric mucosa. CONCLUSIONS AND INFERENCES This study provides important data necessary for a deeper understanding of the complex interactions between OT and DA systems in the regulation of stress and gastric functions. It provides relevant mechanistic clues into OT's role as a protective factor against stress-induced changes to gastric function.
Collapse
Affiliation(s)
- Hui Leng
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Xiaoqian Zhang
- Doctoral School of Biomedical Sciences, Leuven, Belgium.,Family Medicine Department, Qingdao United Family Hospital, Qingdao, China
| | - Qian Wang
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Xiao Luan
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Feifei Guo
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Shengli Gao
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Xuehuan Liu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Luo Xu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Onaka T, Takayanagi Y. Role of oxytocin in the control of stress and food intake. J Neuroendocrinol 2019; 31:e12700. [PMID: 30786104 PMCID: PMC7217012 DOI: 10.1111/jne.12700] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/20/2022]
Abstract
Oxytocin neurones in the hypothalamus are activated by stressful stimuli and food intake. The oxytocin receptor is located in various brain regions, including the sensory information-processing cerebral cortex; the cognitive information-processing prefrontal cortex; reward-related regions such as the ventral tegmental areas, nucleus accumbens and raphe nucleus; stress-related areas such as the amygdala, hippocampus, ventrolateral part of the ventromedial hypothalamus and ventrolateral periaqueductal gray; homeostasis-controlling hypothalamus; and the dorsal motor complex controlling intestinal functions. Oxytocin affects behavioural and neuroendocrine stress responses and terminates food intake by acting on the metabolic or nutritional homeostasis system, modulating emotional processing, reducing reward values of food intake, and facilitating sensory and cognitive processing via multiple brain regions. Oxytocin also plays a role in interactive actions between stress and food intake and contributes to adaptive active coping behaviours.
Collapse
Affiliation(s)
- Tatsushi Onaka
- Division of Brain and NeurophysiologyDepartment of PhysiologyJichi Medical UniversityShimotsuke‐shiJapan
| | - Yuki Takayanagi
- Division of Brain and NeurophysiologyDepartment of PhysiologyJichi Medical UniversityShimotsuke‐shiJapan
| |
Collapse
|
50
|
Mayo LM, Heilig M. In the face of stress: Interpreting individual differences in stress-induced facial expressions. Neurobiol Stress 2019; 10:100166. [PMID: 31193535 PMCID: PMC6535645 DOI: 10.1016/j.ynstr.2019.100166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/17/2019] [Accepted: 04/19/2019] [Indexed: 11/22/2022] Open
Abstract
Stress is an inevitable part of life that can profoundly impact social and emotional functioning, contributing to the development of psychiatric disease. One key component of emotion and social processing is facial expressions, which humans can readily detect and react to even without conscious awareness. Facial expressions have been the focus of philosophic and scientific interest for centuries. Historically, facial expressions have been relegated to peripheral indices of fixed emotion states. More recently, affective neuroscience has undergone a conceptual revolution, resulting in novel interpretations of these muscle movements. Here, we review the role of facial expressions according to the leading affective neuroscience theories, including constructed emotion and social-motivation accounts. We specifically highlight recent data (Mayo et al, 2018) demonstrating the way in which stress shapes facial expressions and how this is influenced by individual factors. In particular, we focus on the consequence of genetic variation within the endocannabinoid system, a neuromodulatory system implicated in stress and emotion, and its impact on stress-induced facial muscle activity. In a re-analysis of this dataset, we highlight how gender may also influence these processes, conceptualized as variation in the "fight-or-flight" or "tend-and-befriend" behavioral responses to stress. We speculate on how these interpretations may contribute to a broader understanding of facial expressions, discuss the potential use of facial expressions as a trans-diagnostic marker of psychiatric disease, and suggest future work necessary to resolve outstanding questions.
Collapse
Affiliation(s)
- Leah M. Mayo
- Center for Social and Affective Neuroscience, Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | | |
Collapse
|