1
|
Wang J, Chen W, Zhu Q, Liu Y, Kang Z, Liu D, Zeng G. Effects of Qingyangshen glycosides on neuroplasticity in a mouse model of social defeat. Animal Model Exp Med 2025; 8:581-594. [PMID: 39921215 PMCID: PMC12008452 DOI: 10.1002/ame2.12499] [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: 12/10/2023] [Accepted: 08/16/2024] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Qingyangshen (Cynanchum otophyllum C.K. Schneid) is a folk drug for treating depression and other mental disorders induced by social defeat stress. Neuroplasticity in the hippocampus is essential for the modulation of cognition and emotion, and its impairment may contribute to the development and progression of depression. Our previous studies have found that Qingyangshen glycosides (QYS) can improve depression-like behavior in social failure mouse models, mainly through PGC-1α/FNDC5/BDNF signaling pathways activation, but its effects and mechanisms on hippocampal neuroplasticity remain unknown. METHODS Chronic social defeat stress (CSDS) was used to induce social defeat in mice. Morphological changes in the hippocampus were observed by H&E staining and Golgi staining. Immunofluorescence double staining was used to detect the expression of synaptophysin (SYN) and postsynaptic density protein-95 (PSD-95), while western blot was employed to evaluate PSD-95, SYN, and doublecortin (DCX) proteins. The pathological processing of social defeat and the therapeutic effects of QYS on it was confirmed through behavioral assessment associated with morphologic observation. RESULTS During the whole study, the sucrose preference indices and OFT activity time of CSDS mice were significantly decreased (p ≤ 0.05), and the tail suspension immobility time was significantly increased (p ≤ 0.05), suggesting that the mice had significant depressive symptoms. Treatment with QYS (25, 50, and 100 mg/kg) significantly alleviated depressive symptoms in CSDS mice, which was demonstrated by significantly (p ≤ 0.05 or p ≤ 0.01) reducing the duration of tail-hanging immobility and increasing the tendency of sucrose preference indices and OFT activity time. QYS treatment also significantly increased the expression of DCX, PSD-95, and SYN proteins, which play a crucial role in depression. CONCLUSIONS QYS alleviated these symptoms by enhancing hippocampal neuroplasticity through upregulating the expression of synapse-associated proteins (SAPs). The therapeutic mechanism of QYS may involve modulating the neuroplasticity of hippocampus neurons by altering the expression of SAPs.
Collapse
Affiliation(s)
- Jingru Wang
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Weishi Chen
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qiang Zhu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yao Liu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zheng Kang
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Dingding Liu
- School of Pharmacy and Research Center for Pharmacodynamic Material Basis and Mechanism of Action, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Guirong Zeng
- Pharmacological research department, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| |
Collapse
|
2
|
Mongi-Bragato B, Sánchez MA, Avalos MP, Boezio MJ, Guzman AS, Rigoni D, Perassi EM, Mas CR, Bisbal M, Bollati FA, Cancela LM. Activation of Nuclear Factor-kappa B in the nucleus accumbens core is necessary for chronic stress-induced glutamate and neuro-immune alterations that facilitate cocaine self-administration. Brain Behav Immun 2025; 128:1-15. [PMID: 40139275 DOI: 10.1016/j.bbi.2025.03.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 03/18/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Stressful events are associated with impaired glutamate signaling and neuroimmune adaptations that may increase the vulnerability of individuals to cocaine addiction. We previously demonstrated that chronic stress induced reactive microglia and increased TNF-α expression in the nucleus accumbens core (NAcore), both alterations strongly linked with impaired glutamate homeostasis and the facilitation of cocaine self-administration. The nuclear factor kappa-B (NF-κB) is a critical regulator of many immune- and addiction-related genes, such as the gene coding for glutamate transporter (GLT-1), and it is considered a master regulator of inflammation, reported to be a key driver of microglia activation in psychiatric diseases. However, no studies have examined the role of NF-κB signaling within the NAcore in the neuroimmune and glutamate mechanism, underpinning stress-induced vulnerability to cocaine self-administration. Here we investigate whether viral dominant negative inhibition of I kappa B kinase (IKKdn), a signaling molecule responsible for NF-κB activation, would prevent stress-induced facilitation to cocaine self-administration and associated changes in accumbal GLT-1 and TNF-α expression. We also explore N-myc proto-oncogene protein (N-myc) levels as a link between NF-κB and stress-induced GLT-1 downregulation. For seven days (days 1-7), adult male rats were restrained for 2 h/day. Animals were administered an intra-NAcore with IKKdn or empty lentiviruses on day 14 after the first restraint stress session. Marked activation of NF-κB was detected in the NAcore of stressed subjects, along with increased NF-κB expression in astrocytes. Consistently, viral NF-κB inhibition prevented stress-induced facilitation of cocaine self-administration. Moreover, NF-κB blockade results in the restoration of stress-induced reduction in GLT-1 levels and was effective in suppressing stress-induced TNF-α within the NAcore. These findings suggest that accumbal NF-κB signaling exerts a central control over stress-altered downstream neuroimmune and glutamate function underlying vulnerability to cocaine use disorders.
Collapse
Affiliation(s)
- Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Marianela Adela Sánchez
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - María Julieta Boezio
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Diana Rigoni
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Físico-Química de Córdoba, INFIQC-CONICET, Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Carlos Ruben Mas
- Centro de Investigaciones en Química Biológica de Córdoba, CIQUIBIC-CONICET, Departamento de Química Bilógica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Friuli 2434, Colinas de Vélez Sarsfield (5016) Córdoba, Argentina, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba, IFEC-CONICET, Departamento de Farmacología Otto Orsingher, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba X5000HUA Córdoba, Argentina.
| |
Collapse
|
3
|
Parise LF, Iñiguez SD, Warren BL, Parise EM, Bachtell RK, Dietz DM, Nestler EJ, Bolaños-Guzmán CA. ERK2 Signaling in the Nucleus Accumbens Facilitates Stress Susceptibility and Cocaine Reinstatement. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100416. [PMID: 39896237 PMCID: PMC11786747 DOI: 10.1016/j.bpsgos.2024.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/09/2024] [Accepted: 10/19/2024] [Indexed: 02/04/2025] Open
Abstract
Background Second-messenger signaling within the mesolimbic reward circuit plays a key role in the negative effects of stress and the underlying mechanisms that promote drug abuse. Because the nucleus accumbens (NAc) integrates reward valence, we investigated how ERK2 (extracellular signal-regulated protein kinase-2) signaling affects the development of stress-related comorbidities, including negative affect and drug sensitivity. Methods We assessed how chronic unpredictable stress influenced the phosphorylation of ERK2-signaling proteins within the NAc of male Sprague Dawley rats. Using a herpes simplex virus, we either upregulated or downregulated NAc ERK2 activation and evaluated behavioral responses to stress-eliciting stimuli (elevated plus maze, open field, forced swim test) and cocaine-seeking behavior (conditioned place preference, self-administration). We also examined ERK2-mediated modifications in spine morphology of medium spiny neurons within the NAc. Results Chronic unpredictable stress increased the phosphorylation of ERK2-signaling proteins within the NAc. Viral-mediated activation of NAc ERK2 enhanced susceptibility to both depression- and anxiety-related stimuli and increased cocaine-seeking behavior (conditioned place preference and reinstatement). These behavioral changes were associated with an increase in stubby and mushroom spines of NAc medium spiny neurons. Conversely, downregulation of ERK2 activation attenuated affect-related behavioral responses in the forced swim test and blunted cocaine's rewarding effects without influencing NAc spine morphology. Conclusions NAc ERK2 contributes to stress-induced behavioral deficits, including anxiety- and depression-like phenotypes, while promoting cocaine-seeking behavior. These findings suggest that ERK2 signaling in the NAc plays a role in the comorbidity of these related syndromes.
Collapse
Affiliation(s)
- Lyonna F. Parise
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas
| | - Sergio D. Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas
| | - Brandon L. Warren
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Eric M. Parise
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ryan K. Bachtell
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado
| | - David M. Dietz
- Department of Pharmacology and Toxicology, Jacobs School of Medicine & Biomedical Sciences, University of Buffalo, Buffalo, New York
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
4
|
Wang X, Wu S, Zuo J, Li K, Chen Y, Fan Z, Wu Z, Yang JX, Song W, Cao JL, Cui M. Selective activation of SIGMAR1 in anterior cingulate cortex glutamatergic neurons facilitates comorbid pain in depression in male mice. Commun Biol 2025; 8:150. [PMID: 39890921 PMCID: PMC11785782 DOI: 10.1038/s42003-025-07590-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Depression and comorbid pain are frequently encountered clinically, and the comorbidity complicates the overall medical management. However, the mechanism whereby depression triggers development of pain needs to be further elucidated. Here, by using the chronic restraint stress (CRS) mouse model of depression and comorbid pain, we showed that CRS hyperactivated the glutamatergic neurons in the anterior cingulate cortex (ACC), as well as increasing the dendrite complexity and number. Chemogenetic activation of these neurons can induce depression and pain, while chemogenetic blockade can reverse such depression-induced pain. Moreover, we utilized translating ribosome affinity purification (TRAP) in combination with c-Fos-tTA strategy and pharmacological approaches and identified SIGMAR1 as a potential therapeutic molecular target. These results revealed a previously unknown neural mechanism for depression and pain comorbidity and provided new mechanistic insights into the antidepressive and analgesic effects of the disease.
Collapse
Affiliation(s)
- Xianlei Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shulin Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Junsheng Zuo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Keying Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yutong Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Zhijie Fan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Zhou Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Weiyi Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Mengqiao Cui
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
5
|
Lee J, Aubry A, Hanif S, Grunfeld IS, Likhtik E, Burghardt NS. Chronic social defeat stress gives rise to social avoidance through fear learning. Behav Brain Res 2025; 476:115245. [PMID: 39241834 PMCID: PMC11513230 DOI: 10.1016/j.bbr.2024.115245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Chronic social defeat stress (CSDS), a widely used rodent model of stress, reliably leads to decreased social interaction in stress susceptible animals. Here, we investigate a role for fear learning in this response using male 129 Sv/Ev mice, a strain that is more vulnerable to CSDS than the commonly used C57BL/6 strain. We first demonstrate that defeated 129 Sv/Ev mice avoid a CD-1 mouse, but not a conspecific, indicating that motivation to socialize is intact in this strain. CD-1 avoidance is characterized by approach behavior that results in running in the opposite direction, activity that is consistent with a threat response. We next test whether CD-1 avoidance is subject to the same behavioral changes found in traditional models of Pavlovian fear conditioning. We find that associative learning occurs across 10 days CSDS, with defeated mice learning to associate the color of the CD-1 coat with threat. This leads to the gradual acquisition of avoidance behavior, a conditioned response that can be extinguished with 7 days of repeated social interaction testing (5 tests/day). Pairing a CD-1 with a tone leads to second-order conditioning, resulting in avoidance of an enclosure without a social target. Finally, we show that social interaction with a conspecific is a highly variable response in defeated mice that may reflect individual differences in generalization of fear to other social targets. Our data indicate that fear conditioning to a social target is a key component of CSDS, implicating the involvement of fear circuits in social avoidance.
Collapse
Affiliation(s)
- Jinah Lee
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA; Psychology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Antonio Aubry
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA; Psychology Program, The Graduate Center, City University of New York, New York, NY, USA; Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sadiyah Hanif
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA
| | - Itamar S Grunfeld
- Psychology Program, The Graduate Center, City University of New York, New York, NY, USA; Department of Biology, Hunter College, City University of New York, New York, NY, USA; Fralin Biomedical Research Institute at Virginia Tech, Roanoke, VA, USA
| | - Ekaterina Likhtik
- Department of Biology, Hunter College, City University of New York, New York, NY, USA; Biology Program, The Graduate Center, City University of New York, New York, NY, USA
| | - Nesha S Burghardt
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA; Psychology Program, The Graduate Center, City University of New York, New York, NY, USA.
| |
Collapse
|
6
|
Solomon MG, Nennig SE, Cotton MR, Whiting KE, Fulenwider HD, Schank JR. Neurokinin-1 receptors in the nucleus accumbens shell influence sensitivity to social defeat stress and stress-induced alcohol consumption in male mice. ADDICTION NEUROSCIENCE 2024; 13:100174. [PMID: 39801674 PMCID: PMC11720327 DOI: 10.1016/j.addicn.2024.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Chronic social defeat stress (SDS) is a widely employed preclinical model of depression involving repeated exposure to physical defeats using a resident-intruder model in male mice. Exposure to SDS induces depressive-like phenotypes including anhedonia, social withdrawal, and increased drug and alcohol consumption. Previously, we found that expression of the neurokinin-1 receptor (NK1R) is increased in the nucleus accumbens (NAC) of mice that are sensitive to this stressor and increase their alcohol intake. The NK1R is the endogenous receptor for the neuropeptide substance P (SP) and plays a prominent role in stress, anxiety, and addiction. In the present study, we assessed changes in NK1R protein levels in the NAC shell and implemented viral vector strategies to demonstrate a functional role of the NK1R in sensitivity to SDS. Specifically, we found that NK1R protein levels were increased in the NAC shell following SDS exposure. Next, we found that NK1R overexpression in the NAC shell increased the sensitivity to SDS and stress-induced alcohol consumption. Together, these experiments provide evidence for a role of the NK1R in the NAC shell in the sensitivity to SDS and the subsequent escalation in alcohol intake.
Collapse
Affiliation(s)
- Matthew G Solomon
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Sadie E Nennig
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Mallory R Cotton
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Kimberly E Whiting
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Hannah D Fulenwider
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| | - Jesse R Schank
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA
| |
Collapse
|
7
|
Wani SN, Grewal AK, Khan H, Singh TG. Elucidating the molecular symphony: unweaving the transcriptional & epigenetic pathways underlying neuroplasticity in opioid dependence and withdrawal. Psychopharmacology (Berl) 2024; 241:1955-1981. [PMID: 39254835 DOI: 10.1007/s00213-024-06684-9] [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: 07/11/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
The persistent use of opioids leads to profound changes in neuroplasticity of the brain, contributing to the emergence and persistence of addiction. However, chronic opioid use disrupts the delicate balance of the reward system in the brain, leading to neuroadaptations that underlie addiction. Chronic cocaine usage leads to synchronized alterations in gene expression, causing modifications in the Nucleus Accumbens (NAc), a vital part of the reward system of the brain. These modifications assist in the development of maladaptive behaviors that resemble addiction. Neuroplasticity in the context of addiction involves changes in synaptic connectivity, neuronal morphology, and molecular signaling pathways. Drug-evoked neuroplasticity in opioid addiction and withdrawal represents a complicated interaction between environmental, genetic, and epigenetic factors. Identifying specific transcriptional and epigenetic targets that can be modulated to restore normal neuroplasticity without disrupting essential physiological processes is a critical consideration. The discussion in this article focuses on the transcriptional aspects of drug-evoked neuroplasticity, emphasizing the role of key transcription factors, including cAMP response element-binding protein (CREB), ΔFosB, NF-kB, Myocyte-enhancing factor 2 (MEF2), Methyl-CpG binding protein 2 (MeCP2), E2F3a, and FOXO3a. These factors regulate gene expression and lead to the neuroadaptive changes observed in addiction and withdrawal. Epigenetic regulation, which involves modifying gene accessibility by controlling these structures, has been identified as a critical component of addiction development. By unraveling these complex molecular processes, this study provides valuable insights that may pave the way for future therapeutic interventions targeting the mechanisms underlying addiction and withdrawal.
Collapse
Affiliation(s)
- Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Aman Pharmacy College, Dholakhera, Udaipurwati, Jhunjhunu, Rajasthan, 333307, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
8
|
Hernandez Silva JC, Pausic N, Marroquin Rivera A, Labonté B, Proulx CD. Chronic Social Defeat Stress Induces Pathway-Specific Adaptations at Lateral Habenula Neuronal Outputs. J Neurosci 2024; 44:e2082232024. [PMID: 39164106 PMCID: PMC11426382 DOI: 10.1523/jneurosci.2082-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 07/15/2024] [Accepted: 08/10/2024] [Indexed: 08/22/2024] Open
Abstract
The lateral habenula (LHb) has emerged as a pivotal brain region implicated in depression, displaying hyperactivity in human and animal models of depression. While the role of LHb efferents in depressive disorders has been acknowledged, the specific synaptic alterations remain elusive. Here, employing optogenetics, retrograde tracing, and ex vivo whole-cell patch-clamp techniques, we investigated synaptic transmission in male mice subjected to chronic social defeat stress (CSDS) at three major LHb neuronal outputs: the dorsal raphe nucleus (DRN), the ventral tegmental area (VTA), and the rostromedial tegmental nucleus (RMTg). Our findings uncovered distinct synaptic adaptations in LHb efferent circuits in response to CSDS. Specifically, CSDS induced in susceptible mice postsynaptic potentiation and postsynaptic depression at the DRN and VTA neurons, respectively, receiving excitatory inputs from the LHb, while CSDS altered presynaptic transmission at the LHb terminals in RMTg in both susceptible and resilient mice. Moreover, whole-cell recordings at projection-defined LHb neurons indicate decreased spontaneous activity in VTA-projecting LHb neurons, accompanied by an imbalance in excitatory-inhibitory inputs at the RMTg-projecting LHb neurons. Collectively, these novel findings underscore the circuit-specific alterations in LHb efferents following chronic social stress, shedding light on potential synaptic adaptations underlying stress-induced depressive-like states.
Collapse
Affiliation(s)
- Jose Cesar Hernandez Silva
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Nikola Pausic
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Arturo Marroquin Rivera
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Benoît Labonté
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| | - Christophe D Proulx
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC G1J 2G3, Canada
| |
Collapse
|
9
|
Zhang JR, Shen SY, Shen ZQ, Yin SY, Ye K, Li W, Li HY, Liang LF, Wang YQ, Guo XY, Yu J. Role of mitochondria-associated membranes in the hippocampus in the pathogenesis of depression. J Affect Disord 2024; 361:637-650. [PMID: 38914161 DOI: 10.1016/j.jad.2024.06.076] [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: 01/25/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Pathological changes, such as microglia activation in the hippocampus frequently occur in individuals with animal models of depression; however, they may share a common cellular mechanism, such as endoplasmic reticulum (ER) stress and mitochondrial dysfunction. Mitochondria associated membranes (MAMs) are communication platforms between ER and mitochondria. This study aimed to investigate the role of intracellular stress responses, especially structural and functional changes of MAMs in depression. METHODS We used chronic social defeat stress (CSDS) to mimic depression in C57 mice to investigate the pathophysiological changes in the hippocampus associated with depression and assess the antidepressant effect of electroacupuncture (EA). Molecular, histological, and electron microscopic techniques were utilized to study intracellular stress responses, including the ER stress pathway reaction, mitochondrial damage, and structural and functional changes in MAMs in the hippocampus after CSDS. Proteomics technology was employed to explore protein-level changes in MAMs caused by CSDS. RESULTS CSDS caused mitochondrial dysfunction, ER stress, closer contact between ER and mitochondria, and enrichment of functional protein clusters at MAMs in hippocampus along with depressive-like behaviors. Also, EA showed beneficial effects on intracellular stress responses and depressive-like behaviors in CSDS mice. LIMITATION The cellular specificity of MAMs related protein changes in CSDS mice was not explored. CONCLUSIONS In the hippocampus, ER stress and mitochondrial damage occur, along with enriched mitochondria-ER interactions and MAM-related protein enrichment, which may contribute to depression's pathophysiology. EA may improve depression by regulating intracellular stress responses.
Collapse
Affiliation(s)
- Jia-Rui Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shi-Yu Shen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zu-Qi Shen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shu-Yuan Yin
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ke Ye
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wei Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hao-Yuan Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ling-Feng Liang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200433, China
| | - Xiao-Yun Guo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| | - Jin Yu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai 200433, China.
| |
Collapse
|
10
|
Pagliusi M, Amorim-Marques AP, Lobo MK, Guimarães FS, Lisboa SF, Gomes FV. The rostral ventromedial medulla modulates pain and depression-related behaviors caused by social stress. Pain 2024; 165:1814-1823. [PMID: 38661577 DOI: 10.1097/j.pain.0000000000003257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 04/26/2024]
Abstract
ABSTRACT The rostral ventromedial medulla (RVM) is a crucial structure in the descending pain modulatory system, playing a key role as a relay for both the facilitation and inhibition of pain. The chronic social defeat stress (CSDS) model has been widely used to study stress-induced behavioral impairments associated with depression in rodents. Several studies suggest that CSDS also causes changes related to chronic pain. In this study, we aimed to investigate the involvement of the RVM in CSDS-induced behavioral impairments, including those associated with chronic pain. We used chemogenetics to activate or inhibit the RVM during stress. The results indicated that the RVM is a vital hub influencing stress outcomes. Rostral ventromedial medulla activation during CSDS ameliorates all the stress outcomes, including social avoidance, allodynia, hyperalgesia, anhedonia, and behavioral despair. In addition, RVM inhibition in animals exposed to a subthreshold social defeat stress protocol induces a susceptible phenotype, facilitating all stress outcomes. Finally, chronic RVM inhibition-without any social stress stimulus-induces chronic pain but not depressive-like behaviors. Our findings provide insights into the comorbidity between chronic pain and depression by indicating the involvement of the RVM in establishing social stress-induced behavioral responses associated with both chronic pain and depression.
Collapse
Affiliation(s)
- Marco Pagliusi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Anna P Amorim-Marques
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sabrina F Lisboa
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Nestler EJ, Russo SJ. Neurobiological basis of stress resilience. Neuron 2024; 112:1911-1929. [PMID: 38795707 PMCID: PMC11189737 DOI: 10.1016/j.neuron.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024]
Abstract
A majority of humans faced with severe stress maintain normal physiological and behavioral function, a process referred to as resilience. Such stress resilience has been modeled in laboratory animals and, over the past 15 years, has transformed our understanding of stress responses and how to approach the treatment of human stress disorders such as depression, post-traumatic stress disorder (PTSD), and anxiety disorders. Work in rodents has demonstrated that resilience to chronic stress is an active process that involves much more than simply avoiding the deleterious effects of the stress. Rather, resilience is mediated largely by the induction of adaptations that are associated uniquely with resilience. Such mechanisms of natural resilience in rodents are being characterized at the molecular, cellular, and circuit levels, with an increasing number being validated in human investigations. Such discoveries raise the novel possibility that treatments for human stress disorders, in addition to being geared toward reversing the damaging effects of stress, can also be based on inducing mechanisms of natural resilience in individuals who are inherently more susceptible. This review provides a progress report on this evolving field.
Collapse
Affiliation(s)
- Eric J Nestler
- Nash Family Department of Neuroscience and Department of Psychiatry, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Scott J Russo
- Nash Family Department of Neuroscience and Department of Psychiatry, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
12
|
Lee J, Aubry A, Hanif S, Grunfeld I, Likhtik E, Burghardt NS. Chronic Social Defeat Stress Gives Rise to Social Avoidance Through Fear Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599597. [PMID: 38948731 PMCID: PMC11213017 DOI: 10.1101/2024.06.18.599597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Chronic social defeat stress (CSDS), a widely used rodent model of stress, reliably leads to decreased social interaction in stress susceptible animals. Here, we investigate a role for fear learning in this response using 129Sv/Ev mice, a strain that is more vulnerable to CSDS than the commonly used C57BL/6 strain. We first demonstrate that defeated 129Sv/Ev mice avoid a CD-1 mouse, but not a conspecific, indicating that motivation to socialize is intact in this strain. CD-1 avoidance is characterized by approach behavior that results in running in the opposite direction, activity that is consistent with a threat response. We next test whether CD-1 avoidance is subject to the same behavioral changes found in traditional models of Pavlovian fear conditioning. We find that associative learning occurs across 10 days CSDS, with defeated mice learning to associate the color of the CD-1 coat with threat. This leads to the gradual acquisition of avoidance behavior, a conditioned response that can be extinguished with 7 days of repeated social interaction testing (5 tests/day). Pairing a CD-1 with a tone leads to second-order conditioning, resulting in avoidance of an enclosure without a social target. Finally, we show that social interaction with a conspecific is a highly variable response in defeated mice that may reflect individual differences in generalization of fear to other social targets. Our data indicate that fear conditioning to a social target is a key component of CSDS, implicating the involvement of fear circuits in social avoidance.
Collapse
|
13
|
Cathomas F, Lin HY, Chan KL, Li L, Parise LF, Alvarez J, Durand-de Cuttoli R, Aubry AV, Muhareb S, Desland F, Shimo Y, Ramakrishnan A, Estill M, Ferrer-Pérez C, Parise EM, Wilk CM, Kaster MP, Wang J, Sowa A, Janssen WG, Costi S, Rahman A, Fernandez N, Campbell M, Swirski FK, Nestler EJ, Shen L, Merad M, Murrough JW, Russo SJ. Circulating myeloid-derived MMP8 in stress susceptibility and depression. Nature 2024; 626:1108-1115. [PMID: 38326622 PMCID: PMC10901735 DOI: 10.1038/s41586-023-07015-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Psychosocial stress has profound effects on the body, including the immune system and the brain1,2. Although a large number of pre-clinical and clinical studies have linked peripheral immune system alterations to stress-related disorders such as major depressive disorder (MDD)3, the underlying mechanisms are not well understood. Here we show that expression of a circulating myeloid cell-specific proteinase, matrix metalloproteinase 8 (MMP8), is increased in the serum of humans with MDD as well as in stress-susceptible mice following chronic social defeat stress (CSDS). In mice, we show that this increase leads to alterations in extracellular space and neurophysiological changes in the nucleus accumbens (NAc), as well as altered social behaviour. Using a combination of mass cytometry and single-cell RNA sequencing, we performed high-dimensional phenotyping of immune cells in circulation and in the brain and demonstrate that peripheral monocytes are strongly affected by stress. In stress-susceptible mice, both circulating monocytes and monocytes that traffic to the brain showed increased Mmp8 expression following chronic social defeat stress. We further demonstrate that circulating MMP8 directly infiltrates the NAc parenchyma and controls the ultrastructure of the extracellular space. Depleting MMP8 prevented stress-induced social avoidance behaviour and alterations in NAc neurophysiology and extracellular space. Collectively, these data establish a mechanism by which peripheral immune factors can affect central nervous system function and behaviour in the context of stress. Targeting specific peripheral immune cell-derived matrix metalloproteinases could constitute novel therapeutic targets for stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hsiao-Yun Lin
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L Chan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johana Alvarez
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio V Aubry
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samer Muhareb
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fiona Desland
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Ferrer-Pérez
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Matthias Wilk
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuella P Kaster
- Department of Biochemistry, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Jun Wang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison Sowa
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Costi
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Adeeb Rahman
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas Fernandez
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Hodes GE, Bangasser D, Sotiropoulos I, Kokras N, Dalla C. Sex Differences in Stress Response: Classical Mechanisms and Beyond. Curr Neuropharmacol 2024; 22:475-494. [PMID: 37855285 PMCID: PMC10845083 DOI: 10.2174/1570159x22666231005090134] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 10/20/2023] Open
Abstract
Neuropsychiatric disorders, which are associated with stress hormone dysregulation, occur at different rates in men and women. Moreover, nowadays, preclinical and clinical evidence demonstrates that sex and gender can lead to differences in stress responses that predispose males and females to different expressions of similar pathologies. In this curated review, we focus on what is known about sex differences in classic mechanisms of stress response, such as glucocorticoid hormones and corticotrophin-releasing factor (CRF), which are components of the hypothalamicpituitary- adrenal (HPA) axis. Then, we present sex differences in neurotransmitter levels, such as serotonin, dopamine, glutamate and GABA, as well as indices of neurodegeneration, such as amyloid β and Tau. Gonadal hormone effects, such as estrogens and testosterone, are also discussed throughout the review. We also review in detail preclinical data investigating sex differences caused by recentlyrecognized regulators of stress and disease, such as the immune system, genetic and epigenetic mechanisms, as well neurosteroids. Finally, we discuss how understanding sex differences in stress responses, as well as in pharmacology, can be leveraged into novel, more efficacious therapeutics for all. Based on the supporting evidence, it is obvious that incorporating sex as a biological variable into preclinical research is imperative for the understanding and treatment of stress-related neuropsychiatric disorders, such as depression, anxiety and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Debra Bangasser
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| | - Ioannis Sotiropoulos
- Institute of Biosciences & Applications NCSR “Demokritos”, Athens, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
15
|
Parise LF, Iñiguez SD, Warren BL, Parise EM, Bachtell RK, Dietz D, Nestler EJ, Bolaños-Guzmán CA. Viral-mediated expression of Erk2 in the nucleus accumbens regulates responses to rewarding and aversive stimuli. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560689. [PMID: 37873069 PMCID: PMC10592906 DOI: 10.1101/2023.10.03.560689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Second-messenger signaling within the mesolimbic reward circuit is involved in both the long-lived effects of stress and in the underlying mechanisms that promote drug abuse liability. To determine the direct role of kinase signaling within the nucleus accumbens, specifically mitogen-activated protein kinase 1 (ERK2), in mood- and drug-related behavior, we used a herpes-simplex virus to up- or down-regulate ERK2 in adult male rats. We then exposed rats to a battery of behavioral tasks including the elevated plus-maze, open field test, forced-swim test, conditioned place preference, and finally cocaine self-administration. Herein, we show that viral overexpression or knockdown of ERK2 in the nucleus accumbens induces distinct behavioral phenotypes. Specifically, over expression of ERK2 facilitated depression- and anxiety-like behavior while also increasing sensitivity to cocaine. Conversely, down-regulation of ERK2 attenuated behavioral deficits, while blunting sensitivity to cocaine. Taken together, these data implicate ERK2 signaling, within the nucleus accumbens, in the regulation of affective behaviors and modulating sensitivity to the rewarding properties of cocaine.
Collapse
|
16
|
Deng Q, Zhang SQ, Yang PF, Dong WT, Wang F, Long LH, Chen JG. α-MSH-catabolic enzyme prolylcarboxypeptidase in nucleus accumbens shell ameliorates stress susceptibility in mice through regulating synaptic plasticity. Acta Pharmacol Sin 2023; 44:1576-1588. [PMID: 37012493 PMCID: PMC10374542 DOI: 10.1038/s41401-023-01074-x] [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: 12/22/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023]
Abstract
Emerging evidence demonstrates the vital role of synaptic transmission and structural remodeling in major depressive disorder. Activation of melanocortin receptors facilitates stress-induced emotional behavior. Prolylcarboxypeptidase (PRCP) is a serine protease, which splits the C-terminal amino acid of α-MSH and inactivates it. In this study, we asked whether PRCP, the endogenous enzyme of melanocortin system, might play a role in stress susceptibility via regulating synaptic adaptations. Mice were subjected to chronic social defeat stress (CSDS) or subthreshold social defeat stress (SSDS). Depressive-like behavior was assessed in SIT, SPT, TST and FST. Based on to behavioral assessments, mice were divided into the susceptible (SUS) and resilient (RES) groups. After social defeat stress, drug infusion or viral expression and behavioral tests, morphological and electrophysiological analysis were conducted in PFX-fixed and fresh brain slices containing the nucleus accumbens shell (NAcsh). We showed that PRCP was downregulated in NAcsh of susceptible mice. Administration of fluoxetine (20 mg·kg-1·d-1, i.p., for 2 weeks) ameliorated the depressive-like behavior, and restored the expression levels of PRCP in NAcsh of susceptible mice. Pharmacological or genetic inhibition of PRCP in NAcsh by microinjection of N-benzyloxycarbonyl-L-prolyl-L-prolinal (ZPP) or LV-shPRCP enhanced the excitatory synaptic transmission in NAcsh, facilitating stress susceptibility via central melanocortin receptors. On the contrary, overexpression of PRCP in NAcsh by microinjection of AAV-PRCP alleviated the depressive-like behavior and reversed the enhanced excitatory synaptic transmission, abnormal dendritogenesis and spinogenesis in NAcsh induced by chronic stress. Furthermore, chronic stress increased the level of CaMKIIα, a kinase closely related to synaptic plasticity, in NAcsh. The elevated level of CaMKIIα was reversed by overexpression of PRCP in NAcsh. Pharmacological inhibition of CaMKIIα in NAcsh alleviated stress susceptibility induced by PRCP knockdown. This study has revealed the essential role of PRCP in relieving stress susceptibility through melanocortin signaling-mediated synaptic plasticity in NAcsh.
Collapse
Affiliation(s)
- Qiao Deng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shao-Qi Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ping-Fen Yang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wan-Ting Dong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, 430030, China
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Hong Long
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China.
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, 430030, China.
- The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, 430030, China.
- Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Favoretto CA, Pagliusi M, Morais-Silva G. Involvement of brain cell phenotypes in stress-vulnerability and resilience. Front Neurosci 2023; 17:1175514. [PMID: 37476833 PMCID: PMC10354562 DOI: 10.3389/fnins.2023.1175514] [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: 02/27/2023] [Accepted: 06/19/2023] [Indexed: 07/22/2023] Open
Abstract
Stress-related disorders' prevalence is epidemically increasing in modern society, leading to a severe impact on individuals' well-being and a great economic burden on public resources. Based on this, it is critical to understand the mechanisms by which stress induces these disorders. The study of stress made great progress in the past decades, from deeper into the hypothalamic-pituitary-adrenal axis to the understanding of the involvement of a single cell subtype on stress outcomes. In fact, many studies have used state-of-the-art tools such as chemogenetic, optogenetic, genetic manipulation, electrophysiology, pharmacology, and immunohistochemistry to investigate the role of specific cell subtypes in the stress response. In this review, we aim to gather studies addressing the involvement of specific brain cell subtypes in stress-related responses, exploring possible mechanisms associated with stress vulnerability versus resilience in preclinical models. We particularly focus on the involvement of the astrocytes, microglia, medium spiny neurons, parvalbumin neurons, pyramidal neurons, serotonergic neurons, and interneurons of different brain areas in stress-induced outcomes, resilience, and vulnerability to stress. We believe that this review can shed light on how diverse molecular mechanisms, involving specific receptors, neurotrophic factors, epigenetic enzymes, and miRNAs, among others, within these brain cell subtypes, are associated with the expression of a stress-susceptible or resilient phenotype, advancing the understanding/knowledge on the specific machinery implicate in those events.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Marco Pagliusi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Gessynger Morais-Silva
- Laboratory of Pharmacology, Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
18
|
Decker Ramirez EB, Arnold ME, McConnell KT, Solomon MG, Amico KN, Schank JR. The effects of lipopolysaccharide exposure on social interaction, cytokine expression, and alcohol consumption in male and female mice. Physiol Behav 2023; 265:114159. [PMID: 36931488 PMCID: PMC10121933 DOI: 10.1016/j.physbeh.2023.114159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Much recent research has demonstrated a role of inflammatory pathways in depressive-like behavior and excess alcohol consumption. Lipopolysaccharide (LPS) is a cell wall component of gram-negative bacteria that can be used to trigger a strong inflammatory response in rodents in a preclinical research setting to study the mechanisms behind this relationship. In our study, we exposed male and female mice to LPS and assessed depressive-like behavior using the social interaction (SI) test, alcohol consumption in the two-bottle choice procedure, and expression of inflammatory mediators using quantitative PCR. We found that LPS administration decreased SI in female mice but had no significant impact on male mice when assessed 24 h after injection. LPS resulted in increased proinflammatory cytokine expression in both male and female mice; however, some aspects of the cytokine upregulation observed was greater in female mice as compared to males. A separate cohort of male and female mice underwent drinking for 12 days before receiving a saline or LPS injection, which we found to increase alcohol intake in both males and females. We have previously observed a role of the neurokinin-1 receptor (NK1R) in escalated alcohol intake, and in the inflammatory and behavioral response to LPS. The NK1R is the endogenous target of the neuropeptide SP, and this system has wide ranging roles in depression, anxiety, drug/alcohol seeking, pain, and inflammation. Thus, we administered a NK1R antagonist prior to alcohol access. This treatment reduced escalated alcohol consumption in female mice treated with LPS but did not affect drinking in males. Taken together, these results indicate that females are more sensitive to some physiological and behavioral effects of LPS administration, but that LPS escalates alcohol consumption in both sexes. Furthermore, NK1R antagonism can reduce alcohol consumption that is escalated by LPS treatment, in line with our previous findings.
Collapse
Affiliation(s)
- E B Decker Ramirez
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - M E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - K T McConnell
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - M G Solomon
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - K N Amico
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA
| | - J R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA 30602 USA.
| |
Collapse
|
19
|
Yao J, Chen C, Guo Y, Yang Y, Liu X, Chu S, Ai Q, Zhang Z, Lin M, Yang S, Chen N. A Review of Research on the Association between Neuron-Astrocyte Signaling Processes and Depressive Symptoms. Int J Mol Sci 2023; 24:ijms24086985. [PMID: 37108148 PMCID: PMC10139177 DOI: 10.3390/ijms24086985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Depression is a mental illness that has a serious negative impact on physical and mental health. The pathophysiology of depression is still unknown, and therapeutic medications have drawbacks, such as poor effectiveness, strong dependence, adverse drug withdrawal symptoms, and harmful side effects. Therefore, the primary purpose of contemporary research is to understand the exact pathophysiology of depression. The connection between astrocytes, neurons, and their interactions with depression has recently become the focus of great research interest. This review summarizes the pathological changes of neurons and astrocytes, and their interactions in depression, including the alterations of mid-spiny neurons and pyramidal neurons, the alterations of astrocyte-related biomarkers, and the alterations of gliotransmitters between astrocytes and neurons. In addition to providing the subjects of this research and suggestions for the pathogenesis and treatment techniques of depression, the intention of this article is to more clearly identify links between neuronal-astrocyte signaling processes and depressive symptoms.
Collapse
Affiliation(s)
- Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Cong Chen
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yi Guo
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xinya Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha 410208, China
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
20
|
Jiang Y, Zou M, Wang Y, Wang Y. Nucleus accumbens in the pathogenesis of major depressive disorder: A brief review. Brain Res Bull 2023; 196:68-75. [PMID: 36889362 DOI: 10.1016/j.brainresbull.2023.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Major depressive disorder (MDD) is the most prevalent mental disorder characterized by anhedonia, loss of motivation, avolition, behavioral despair and cognitive abnormalities. Despite substantial advancements in the pathophysiology of MDD in recent years, the pathogenesis of this disorder is not fully understood. Meanwhile,the treatment of MDD with currently available antidepressants is inadequate, highlighting the urgent need for clarifying the pathophysiology of MDD and developing novel therapeutics. Extensive studies have demonstrated the involvement of nuclei such as the prefrontal cortex (PFC), hippocampus (HIP), nucleus accumbens (NAc), hypothalamus, etc., in MDD. NAc,a region critical for reward and motivation,dysregulation of its activity seems to be a hallmark of this mood disorder. In this paper, we present a review of NAc related circuits, cellular and molecular mechanisms underlying MDD and share an analysis of the gaps in current research and possible future research directions.
Collapse
Affiliation(s)
- Yajie Jiang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Manshu Zou
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Yeqing Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China.
| |
Collapse
|
21
|
Foster WB, Beach KF, Carson PF, Harris KC, Alonso BL, Costa LT, Simamora RC, Corbin JE, Hoag KF, Mercado SI, Bernhard AG, Leung CH, Nestler EJ, Been LE. Estradiol withdrawal following a hormone simulated pregnancy induces deficits in affective behaviors and increases ∆FosB in D1 and D2 neurons in the nucleus accumbens core in mice. Horm Behav 2023; 149:105312. [PMID: 36645923 PMCID: PMC9974842 DOI: 10.1016/j.yhbeh.2023.105312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/02/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
In placental mammals, estradiol levels are chronically elevated during pregnancy, but quickly drop to prepartum levels following birth. This may produce an "estrogen withdrawal" state that has been linked to changes in affective states in humans and rodents during the postpartum period. The neural mechanisms underlying these affective changes, however, are understudied. We used a hormone-simulated pseudopregnancy (HSP), a model of postpartum estrogen withdrawal, in adult female C57BL/6 mice to test the impact of postpartum estradiol withdrawal on several behavioral measures of anxiety and motivation. We found that estradiol withdrawal following HSP increased anxiety-like behavior in the elevated plus maze, but not in the open field or marble burying tests. Although hormone treatment during HSP consistently increased sucrose consumption, sucrose preference was generally not impacted by hormone treatment or subsequent estradiol withdrawal. In the social motivation test, estradiol withdrawal decreased the amount of time spent in proximity to a social stimulus animal. These behavioral changes were accompanied by changes in the expression of ∆FosB, a transcription factor correlated with stable long-term plasticity, in the nucleus accumbens (NAc). Specifically, estrogen-withdrawn females had higher ∆FosB expression in the nucleus accumbens core, but ∆FosB expression did not vary across hormone conditions in the nucleus accumbens shell. Using transgenic reporter mice, we found that this increase in ∆FosB occurred in both D1- and D2-expressing cells in the NAc core. Together, these results suggest that postpartum estrogen withdrawal impacts anxiety and motivation and increases ∆FosB in the NAc core.
Collapse
Affiliation(s)
| | | | - Paige F Carson
- Haverford College, Department of Psychology, Haverford, PA, USA
| | - Kagan C Harris
- Haverford College, Department of Psychology, Haverford, PA, USA
| | | | - Leo T Costa
- Haverford College, Department of Psychology, Haverford, PA, USA
| | - Roy C Simamora
- Haverford College, Department of Psychology, Haverford, PA, USA
| | - Jaclyn E Corbin
- Haverford College, Department of Psychology, Haverford, PA, USA
| | - Keegan F Hoag
- Haverford College, Department of Psychology, Haverford, PA, USA
| | | | - Anya G Bernhard
- Haverford College, Department of Psychology, Haverford, PA, USA
| | - Cary H Leung
- Widener College, Department of Biology, Chester, PA, USA
| | - Eric J Nestler
- Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, USA
| | - Laura E Been
- Haverford College, Department of Psychology, Haverford, PA, USA.
| |
Collapse
|
22
|
Kropp DR, Hodes GE. Sex differences in depression: An immunological perspective. Brain Res Bull 2023; 196:34-45. [PMID: 36863664 DOI: 10.1016/j.brainresbull.2023.02.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/05/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
Depression is a heterogenous disorder with symptoms that present differently across individuals. In a subset of people depression is associated with alterations of the immune system that may contribute to disorder onset and symptomology. Women are twice as likely to develop depression and on average have a more sensitive adaptive and innate immune system when compared to men. Sex differences in pattern recognition receptors (PRRs), release of damage-associated molecular patterns (DAMPs), cell populations, and circulating cytokines play a critical role in inflammation onset. Sex differences in innate and adaptive immunity change the response of and repair to damage caused by dangerous pathogens or molecules in the body. This article reviews the evidence for sex specific immune responses that contribute to the sex differences in symptoms of depression that may account for the higher rate of depression in women.
Collapse
Affiliation(s)
- Dawson R Kropp
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Georgia E Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
23
|
Ecological validity of social defeat stressors in mouse models of vulnerability and resilience. Neurosci Biobehav Rev 2023; 145:105032. [PMID: 36608919 DOI: 10.1016/j.neubiorev.2023.105032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Laboratory mouse models offer opportunities to bridge the gap between basic neuroscience and applied stress research. Here we consider the ecological validity of social defeat stressors in mouse models of emotional vulnerability and resilience. Reports identified in PubMed from 1980 to 2020 are reviewed for the ecological validity of social defeat stressors, sex of subjects, and whether results are discussed in terms of vulnerability alone, resilience alone, or both vulnerability and resilience. Most of the 318 reviewed reports (95%) focus on males, and many reports (71%) discuss vulnerability and resilience. Limited ecological validity is associated with increased vulnerability and decreased resilience. Elements of limited ecological validity include frequent and repeated exposure to defeat stressors without opportunities to avoid or escape from unfamiliar conspecifics that are pre-screened and selected for aggressive behavior. These elements ensure defeat and may be required to induce vulnerability, but they are not representative of naturalistic conditions. Research aimed at establishing causality is needed to determine whether ecologically valid stressors build resilience in both sexes of mice.
Collapse
|
24
|
Cathomas F, Lin HY, Chan KL, Li L, Durand-de Cuttoli R, Parise LF, Aubry AV, Muhareb S, Desland F, Shimo Y, Ramakrishnan A, Estill M, Ferrer-Pérez C, Parise EM, Wang J, Sowa A, Janssen WG, Costi S, Rahman A, Fernandez N, Swirski FK, Nestler EJ, Shen L, Merad M, Murrough JW, Russo SJ. Peripheral immune-derived matrix metalloproteinase promotes stress susceptibility. RESEARCH SQUARE 2023:rs.3.rs-1647827. [PMID: 36778505 PMCID: PMC9915787 DOI: 10.21203/rs.3.rs-1647827/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Psychosocial stress has profound effects on the body, including the peripheral immune system and the brain1,2. Although a large number of pre-clinical and clinical studies have linked peripheral immune system alterations to stress-related disorders such as major depressive disorder (MDD)3,4,5, the underlying mechanisms are not well understood. Here we show that a peripheral myeloid cell-specific proteinase, matrix metalloproteinase 8 (MMP8), is elevated in serum of subjects with MDD as well as in stress-susceptible (SUS) mice following chronic social defeat stress (CSDS). In mice, we show that this increase leads to alterations in extracellular space and neurophysiological changes in the nucleus accumbens (NAc), thereby altering social behaviour. Using a combination of mass cytometry and single-cell RNA-sequencing, we performed high-dimensional phenotyping of immune cells in circulation and brain and demonstrate that peripheral monocytes are strongly affected by stress. Both peripheral and brain-infiltrating monocytes of SUS mice showed increased Mmp8 expression following CSDS. We further demonstrate that peripheral MMP8 directly infiltrates the NAc parenchyma to control the ultrastructure of the extracellular space. Depleting MMP8 prevented stress-induced social avoidance behaviour and alterations in NAc neurophysiology and extracellular space. Collectively, these data establish a novel mechanism by which peripheral immune factors can affect central nervous system function and behaviour in the context of stress. Targeting specific peripheral immune cell-derived matrix metalloproteinases could constitute novel therapeutic targets for stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsiao-Yun Lin
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L. Chan
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Li
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F. Parise
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio V. Aubry
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samer Muhareb
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fiona Desland
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Ferrer-Pérez
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M. Parise
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Wang
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison Sowa
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G. Janssen
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Costi
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Adeeb Rahman
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicolas Fernandez
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Filip K. Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - James W. Murrough
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Scott J. Russo
- Nash Family Department of Neuroscience, Brain & Body Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
25
|
Anderson KR, Rogu PJ, Palumbo TB, Miwa JM. Abnormal response to chronic social defeat stress and fear extinction in a mouse model of cholinergic dysregulation. RESEARCH SQUARE 2023:rs.3.rs-2492514. [PMID: 36778356 PMCID: PMC9915767 DOI: 10.21203/rs.3.rs-2492514/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cholinergic signaling is critical for an individual to react appropriately and adaptably to salient stimuli while navigating a complex environment. The cholinergic neurotransmitter system drives attention to salient stimuli, such as stressors, and aids in orchestrating the proper neural and behavioral response. Fine-tuned regulation of the cholinergic system has been linked to appropriate stress responses and subsequent mood regulation while dysregulation has been implicated in mood disorders. Among the multiple layers of regulation are cholinergic protein modulators. Here, we use validated models of experiential-based affective disorders to investigate differences in responses to stress in a genetic mouse model of cholinergic dysregulation based on the loss of protein modulator. The lynx2 nicotinic receptor modulatory protein provides negative cholinergic regulation within the amygdala, medial prefrontal cortex, and other brain regions. We discovered here that lynx2 knockout (KO) mice demonstrate an inability to update behavior with an inability to extinguish learned fear during a fear extinction test. We also observed, under an increased stress load following exposure to chronic social defeat stress (CSDS) paradigm, there was a unified resilience phenotype in lynx2KO mice, as opposed to the wild-type cohort which was split between resilience and susceptible phenotypes. Furthermore, we provide evidence for the functional role of α7 nicotinic receptor subtypes by phenotypic rescue with MLA or crossing with an α7 null mutant mouse (e.g. lynx2/α7 double KO mice). We demonstrate a direct physical interaction between lynx2 and α7 nAChR by co-immunoprecipitation of complexes from mouse BLA extracts. The genetic predisposition to heightened basal anxiety-like behavior and altered cholinergic signaling impairs individual behavior responses stressors. Together, these data indicate that the effects of social stress can be influenced by baseline genetic factors involved in anxiety regulation.
Collapse
|
26
|
Matcha Tea Powder's Antidepressant-like Effect through the Activation of the Dopaminergic System in Mice Is Dependent on Social Isolation Stress. Nutrients 2023; 15:nu15030581. [PMID: 36771286 PMCID: PMC9921318 DOI: 10.3390/nu15030581] [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: 12/09/2022] [Revised: 01/02/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Matcha tea powder is believed to have various physiological benefits; however, its detailed mechanism of action has been poorly understood. Here, we investigated whether the mental state of mice, due to social isolation stress, affects the antidepressant-like effect of Matcha tea powder by using the tail suspension test. Oral administration of Matcha tea powder reduced the duration of immobility in the stress-susceptible C57BL/6J strain, but not in BALB/c strain. In C57BL/6J mice, SCH23390, a dopamine D1 receptor blocker, prevented Matcha tea powder from exerting its antidepressant-like effect. Matcha tea powder also increased the number of c-Fos-positive cells in the prefrontal cortex (PFC) region and the nucleus accumbens (NAc) region in C57BL/6J mice, but not in BALB/c mice. In contrast, Matcha tea powder did not change the number of c-Fos-positive cells in the ventral tegmental area (VTA) region. Notably, C57BL/6J mice with a shorter immobility time had a higher number of c-Fos-positive cells in the PFC, NAc, and VTA regions. However, no such correlation was observed in the stress-tolerant BALB/c mice. These results suggest that Matcha tea powder exerts an antidepressant-like effect through the activation of the dopaminergic system including the PFC-NAc-VTA circuit and that mental states are important factors affecting the physiological benefits of Matcha tea powder.
Collapse
|
27
|
Deng Q, Zhang S, Yang P, Dong W, Wang J, Chen J, Wang F, Long L. A thalamic circuit facilitates stress susceptibility via melanocortin 4 receptor-mediated activation of nucleus accumbens shell. CNS Neurosci Ther 2022; 29:646-658. [PMID: 36510669 PMCID: PMC9873525 DOI: 10.1111/cns.14046] [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: 01/05/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
AIMS Central melanocortin 4 receptor (MC4R) has been reported to induce anhedonia via eliciting dysfunction of excitatory synapses. It is evident that metabolic signals are closely related to chronic stress-induced depression. Here, we investigated that a neural circuit is involved in melanocortin signaling contributing to susceptibility to stress. METHODS Chronic social defeat stress (CSDS) was used to develop depressive-like behavior. Electrophysiologic and chemogenetic approaches were performed to evaluate the role of paraventricular thalamus (PVT) glutamatergic to nucleus accumbens shell (NAcsh) circuit in stress susceptibility. Pharmacological and genetic manipulations were applied to investigate the molecular mechanisms of melanocortin signaling in the circuit. RESULTS CSDS increases the excitatory neurotransmission in NAcsh through MC4R signaling. The enhanced excitatory synaptic input in NAcsh is projected from PVT glutamatergic neurons. Moreover, chemogenetic manipulation of PVTGlu -NAcsh projection mediates the susceptibility to stress, which is dependent on MC4R signaling. Overall, these results reveal that the strengthened excitatory neurotransmission in NAcsh originates from PVT glutamatergic neurons, facilitating the susceptibility to stress through melanocortin signaling. CONCLUSIONS Our results make a strong case for harnessing a thalamic circuit to reorganize excitatory synaptic transmission in relieving stress susceptibility and provide insights gained on metabolic underpinnings of protection against stress-induced depressive-like behavior.
Collapse
Affiliation(s)
- Qiao Deng
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina
| | - Shao‐Qi Zhang
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina
| | - Ping‐Fen Yang
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina
| | - Wan‐Ting Dong
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina
| | - Jia‐Lin Wang
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina
| | - Jian‐Guo Chen
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina,The Research Center for DepressionTongji Medical College, Huazhong University of Science and TechnologyWuhanChina,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhanChina,Key Laboratory of Neurological Diseases (HUST)Ministry of Education of ChinaWuhan CityHubeiChina,Laboratory of Neuropsychiatric DiseasesThe Institute of Brain Research, Huazhong University of Science and TechnologyWuhanChina
| | - Fang Wang
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina,The Research Center for DepressionTongji Medical College, Huazhong University of Science and TechnologyWuhanChina,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhanChina,Key Laboratory of Neurological Diseases (HUST)Ministry of Education of ChinaWuhan CityHubeiChina,Laboratory of Neuropsychiatric DiseasesThe Institute of Brain Research, Huazhong University of Science and TechnologyWuhanChina
| | - Li‐Hong Long
- Department of PharmacologySchool of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan CityHubeiChina,The Research Center for DepressionTongji Medical College, Huazhong University of Science and TechnologyWuhanChina,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei ProvinceWuhanChina
| |
Collapse
|
28
|
Biltz RG, Sawicki CM, Sheridan JF, Godbout JP. The neuroimmunology of social-stress-induced sensitization. Nat Immunol 2022; 23:1527-1535. [PMID: 36369271 PMCID: PMC10000282 DOI: 10.1038/s41590-022-01321-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
Myriad clinical findings provide links between chronic stressors, inflammation, and mood disorders. Furthermore, traumatic or chronic exposure to psychological stressors may promote stress sensitization, in which individuals have long-term complications, including increased vulnerability to subsequent stressors. Post-traumatic stress disorder (PTSD) is a clinically relevant example of stress sensitization. PTSD alters neuronal circuitry and mood; however, the mechanisms underlying long-term stress sensitization within this disorder are unclear. Rodent models of chronic social defeat recapitulate several key physiological, immunological, and behavioral responses associated with psychological stress in humans. Repeated social defeat (RSD) uniquely promotes the convergence of neuronal, central inflammatory (microglial), and peripheral immune (monocyte) pathways, leading to prolonged anxiety, social withdrawal, and cognitive impairment. Moreover, RSD promotes stress sensitization, in which mice are highly sensitive to subthreshold stress exposure and recurrence of anxiety weeks after the cessation of stress. Therefore, the purpose of this Review is to discuss the influence of social-defeat stress on the immune system that may underlie stress sensitization within three key cellular compartments: neurons, microglia, and monocytes. Delineating the mechanisms of stress sensitization is critical in understanding and treating conditions such as PTSD.
Collapse
Affiliation(s)
- Rebecca G Biltz
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Caroline M Sawicki
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH, USA
| | - John F Sheridan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
29
|
Avalos MP, Guzman AS, Garcia-Keller C, Mongi-Bragato B, Esparza MA, Rigoni D, Sanchez MA, Calfa GD, Bollati FA, Cancela LM. Impairment of glutamate homeostasis in the nucleus accumbens core underpins cross-sensitization to cocaine following chronic restraint stress. Front Physiol 2022; 13:896268. [PMID: 36091376 PMCID: PMC9462460 DOI: 10.3389/fphys.2022.896268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Though the facilitating influence of stress on drug abuse is well documented, the mechanisms underlying this interaction have yet to be fully elucidated. The present study explores the neurobiological mechanisms underpinning the sensitized response to the psychomotor-stimulating effects of cocaine following chronic restraint stress (CRS), emphasizing the differential contribution of both subcompartments of the nucleus accumbens (NA), the core (NAcore) and shell (NAshell), to this phenomenon. Adult male Wistar rats were restrained for 2 h/day for 7 days and, 2 weeks after the last stress exposure (day 21), all animals were randomly assigned to behavioral, biochemical or neurochemical tests. Our results demonstrated that the enduring CRS-induced increase in psychostimulant response to cocaine was paralleled by an increase of extracellular dopamine levels in the NAcore, but not the NAshell, greater than that observed in the non-stress group. Furthermore, we found that CRS induced an impairment of glutamate homeostasis in the NAcore, but not the NAshell. Its hallmarks were increased basal extracellular glutamate concentrations driven by a CRS-induced downregulation of GLT-1, blunted glutamate levels in response to cocaine and postsynaptic structural remodeling in pre-stressed animals. In addition, ceftriaxone, a known GLT-1 enhancer, prevented the CRS-induced GLT-1 downregulation, increased basal extracellular glutamate concentrations and changes in structural plasticity in the NAcore as well as behavioral cross-sensitization to cocaine, emphasizing the biological importance of GLT-1 in the comorbidity between chronic stress exposure and drug abuse. A future perspective concerning the paramount relevance of the stress-induced disruption of glutamate homeostasis as a vulnerability factor to the development of stress and substance use disorders during early life or adulthood of descendants is provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Flavia A. Bollati
- Departamento de Farmacología Otto Orsingher, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Liliana M. Cancela
- Departamento de Farmacología Otto Orsingher, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
30
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
31
|
Life extension factor klotho regulates behavioral responses to stress via modulation of GluN2B function in the nucleus accumbens. Neuropsychopharmacology 2022; 47:1710-1720. [PMID: 35449449 PMCID: PMC9283408 DOI: 10.1038/s41386-022-01323-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/23/2022] [Accepted: 04/05/2022] [Indexed: 11/08/2022]
Abstract
Klotho is a life extension factor that has the ability to regulate the function of GluN2B-containing N-methyl-D-aspartate receptors (NMDARs), whose dysfunction in the nucleus accumbens (NAc) underlies critical aspects of the pathophysiology of major depression. Here, we study the functional relevance of klotho in the pathogenesis of depression. A chronic social defeat stress paradigm, in which mice are categorized as either susceptible or unsusceptible based on their performance in a social interaction test, was used in this study. We found that the expression of klotho was largely decreased in the NAc of susceptible mice compared to control or unsusceptible mice. Genetic knockdown of klotho in the NAc induced behavioral alterations relevant to depression in naive mice, while overexpression of klotho produced an antidepressive effect in normal mice and ameliorated the behavioral responses to stress in susceptible mice. Molecularly, knockdown of klotho in the NAc resulted in selective decreases in total and synaptic GluN2B expression that were identical to those in susceptible mice. Elevation of klotho in the NAc reversed the reductions in GluN2B expressions and altered synaptic transmission and spine density in the NAc of susceptible mice. Furthermore, blockade of GluN2B with a specific antagonist abolished the beneficial effects of klotho elevation in susceptible mice. Collectively, we demonstrated that klotho in the NAc modulates behavioral responses to stress by regulating the function of GluN2B-containing NMDARs. These results reveal a novel role for klotho in the pathogenesis of depression, providing new insights into the molecular basis of major depression.
Collapse
|
32
|
Abstract
Depression is an episodic form of mental illness characterized by mood state transitions with poorly understood neurobiological mechanisms. Antidepressants reverse the effects of stress and depression on synapse function, enhancing neurotransmission, increasing plasticity, and generating new synapses in stress-sensitive brain regions. These properties are shared to varying degrees by all known antidepressants, suggesting that synaptic remodeling could play a key role in depression pathophysiology and antidepressant function. Still, it is unclear whether and precisely how synaptogenesis contributes to mood state transitions. Here, we review evidence supporting an emerging model in which depression is defined by a distinct brain state distributed across multiple stress-sensitive circuits, with neurons assuming altered functional properties, synapse configurations, and, importantly, a reduced capacity for plasticity and adaptation. Antidepressants act initially by facilitating plasticity and enabling a functional reconfiguration of this brain state. Subsequently, synaptogenesis plays a specific role in sustaining these changes over time.
Collapse
Affiliation(s)
- Puja K Parekh
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| | - Shane B Johnson
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| | - Conor Liston
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| |
Collapse
|
33
|
Cathomas F, Holt LM, Parise EM, Liu J, Murrough JW, Casaccia P, Nestler EJ, Russo SJ. Beyond the neuron: Role of non-neuronal cells in stress disorders. Neuron 2022; 110:1116-1138. [PMID: 35182484 PMCID: PMC8989648 DOI: 10.1016/j.neuron.2022.01.033] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Stress disorders are leading causes of disease burden in the U.S. and worldwide, yet available therapies are fully effective in less than half of all individuals with these disorders. Although to date, much of the focus has been on neuron-intrinsic mechanisms, emerging evidence suggests that chronic stress can affect a wide range of cell types in the brain and periphery, which are linked to maladaptive behavioral outcomes. Here, we synthesize emerging literature and discuss mechanisms of how non-neuronal cells in limbic regions of brain interface at synapses, the neurovascular unit, and other sites of intercellular communication to mediate the deleterious, or adaptive (i.e., pro-resilient), effects of chronic stress in rodent models and in human stress-related disorders. We believe that such an approach may one day allow us to adopt a holistic "whole body" approach to stress disorder research, which could lead to more precise diagnostic tests and personalized treatment strategies. Stress is a major risk factor for many psychiatric disorders. Cathomas et al. review new insight into how non-neuronal cells mediate the deleterious effects, as well as the adaptive, protective effects, of stress in rodent models and human stress-related disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Abstract
Histone deacetylases (HDACs) have been implicated in learning and memory, and their dysregulation has been linked to cognitive impairment in brain aging and neurodegenerative diseases. In this review, we focus on HDAC1 and HDAC2, highlighting recent progress on their roles in regulating brain function through distinct mechanisms, including gene repression and DNA repair pathways. Moreover, we discuss evidence demonstrating how HDAC1 and HDAC2 could be modulated and their potential as targets to combat memory deficits.
Collapse
Affiliation(s)
- Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
35
|
Avalos MP, Guzman AS, Rigoni D, Gorostiza EA, Sanchez MA, Mongi-Bragato B, Garcia-Keller C, Perassi EM, Virgolini MB, Peralta Ramos JM, Iribarren P, Calfa GD, Bollati FA, Cancela LM. Minocycline prevents chronic restraint stress-induced vulnerability to developing cocaine self-administration and associated glutamatergic mechanisms: a potential role of microglia. Brain Behav Immun 2022; 101:359-376. [PMID: 35065197 DOI: 10.1016/j.bbi.2022.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/24/2021] [Accepted: 01/16/2022] [Indexed: 12/14/2022] Open
Abstract
Stressful experience-induced cocaine-related behaviors are associated with a significant impairment of glutamatergic mechanisms in the Nucleus Accumbens core (NAcore). The hallmarks of disrupted glutamate homeostasis following restraint stress are the enduring imbalance of glutamate efflux after a cocaine stimulus and increased basal concentrations of extracellular glutamate attributed to GLT-1 downregulation in the NAcore. Glutamate transmission is tightly linked to microglia functioning. However, the role of microglia in the biological basis of stress-induced addictive behaviors is still unknown. By using minocycline, a potent inhibitor of microglia activation with anti-inflammatory properties, we determined whether microglia could aid chronic restraint stress (CRS)-induced glutamate homeostasis disruption in the NAcore, underpinning stress-induced cocaine self-administration. In this study, adult male rats were restrained for 2 h/day for seven days (day 1-7). From day 16 until completing the experimental protocol, animals received a vehicle or minocycline treatment (30 mg/Kg/12h i.p.). On day 21, animals were assigned to microscopic, biochemical, neurochemical or behavioral studies. We confirm that the CRS-induced facilitation of cocaine self-administration is associated with enduring GLT-1 downregulation, an increase of basal extracellular glutamate and postsynaptic structural plasticity in the NAcore. These alterations were strongly related to the CRS-induced reactive microglia and increased TNF-α mRNA and protein expression, since by administering minocycline, the impaired glutamate homeostasis and the facilitation of cocaine self-administration were prevented. Our findings are the first to demonstrate that minocycline suppresses the CRS-induced facilitation of cocaine self-administration and glutamate homeostasis disruption in the NAcore. A role of microglia is proposed for the development of glutamatergic mechanisms underpinning stress-induced vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Daiana Rigoni
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Ezequiel Axel Gorostiza
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Marianela Adela Sanchez
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Constanza Garcia-Keller
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC-CONICET), Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Miriam Beatriz Virgolini
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Javier María Peralta Ramos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Pablo Iribarren
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Gastón Diego Calfa
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
36
|
Franco D, Wulff AB, Lobo MK, Fox ME. Chronic Physical and Vicarious Psychosocial Stress Alter Fentanyl Consumption and Nucleus Accumbens Rho GTPases in Male and Female C57BL/6 Mice. Front Behav Neurosci 2022; 16:821080. [PMID: 35221946 PMCID: PMC8867005 DOI: 10.3389/fnbeh.2022.821080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Chronic stress can increase the risk of developing a substance use disorder in vulnerable individuals. Numerous models have been developed to probe the underlying neurobiological mechanisms, however, most prior work has been restricted to male rodents, conducted only in rats, or introduces physical injury that can complicate opioid studies. Here we sought to establish how chronic psychosocial stress influences fentanyl consumption in male and female C57BL/6 mice. We used chronic social defeat stress (CSDS), or the modified vicarious chronic witness defeat stress (CWDS), and used social interaction to stratify mice as stress-susceptible or resilient. We then subjected mice to a 15 days fentanyl drinking paradigm in the home cage that consisted of alternating forced and choice periods with increasing fentanyl concentrations. Male mice susceptible to either CWDS or CSDS consumed more fentanyl relative to unstressed mice. CWDS-susceptible female mice did not differ from unstressed mice during the forced periods, but showed increased preference for fentanyl over time. We also found decreased expression of nucleus accumbens Rho GTPases in male, but not female mice following stress and fentanyl drinking. We also compare fentanyl drinking behavior in mice that had free access to plain water throughout. Our results indicate that stress-sensitized fentanyl consumption is dependent on both sex and behavioral outcomes to stress.
Collapse
Affiliation(s)
- Daniela Franco
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andreas B. Wulff
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Megan E. Fox
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, PA, United States,*Correspondence: Megan E. Fox,
| |
Collapse
|
37
|
Robison AJ, Nestler EJ. ΔFOSB: A Potentially Druggable Master Orchestrator of Activity-Dependent Gene Expression. ACS Chem Neurosci 2022; 13:296-307. [PMID: 35020364 PMCID: PMC8879420 DOI: 10.1021/acschemneuro.1c00723] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
ΔFOSB is a uniquely stable member of the FOS family of immediate early gene AP1 transcription factors. Its accumulation in specific cell types and tissues in response to a range of chronic stimuli is associated with biological phenomena as diverse as memory formation, drug addiction, stress resilience, and immune cell activity. Causal connections between ΔFOSB expression and the physiological and behavioral sequelae of chronic stimuli have been established in rodent and, in some cases, primate models for numerous healthy and pathological states with such preclinical observations often supported by human data demonstrating tissue-specific ΔFOSB expression associated with several specific syndromes. However, the viability of ΔFOSB as a target for therapeutic intervention might be questioned over presumptive concerns of side effects given its expression in such a wide range of cell types and circumstances. Here, we summarize numerous insights from the past three decades of research into ΔFOSB structure, function, mechanisms of induction, and regulation of target genes that support its potential as a druggable target. We pay particular attention to the potential for targeting distinct ΔFOSB isoforms or distinct ΔFOSB-containing multiprotein complexes to achieve cell type or tissue specificity to overcome off-target concerns. We also cover critical gaps in knowledge that currently limit the exploitation of ΔFOSB's therapeutic possibilities and how they may be addressed. Finally, we summarize both current and potential future strategies for generating small molecules or genetic tools for the manipulation of ΔFOSB in the clinic.
Collapse
Affiliation(s)
- Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
38
|
Rigoni D, Avalos MP, Boezio MJ, Guzmán AS, Calfa GD, Perassi EM, Pierotti SM, Bisbal M, Garcia-Keller C, Cancela LM, Bollati F. Stress-induced vulnerability to develop cocaine addiction depends on cofilin modulation. Neurobiol Stress 2021; 15:100349. [PMID: 34169122 PMCID: PMC8209265 DOI: 10.1016/j.ynstr.2021.100349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022] Open
Abstract
Actin dynamics in dendritic spines can be associated with the neurobiological mechanisms supporting the comorbidity between stress exposure and cocaine increase rewards. The actin cytoskeleton remodeling in the nucleus accumbens (NA) has been implicated in the expression of stress-induced cross-sensitization with cocaine. The present study evaluates the involvement of cofilin, a direct regulator of actin dynamics, in the impact of stress on vulnerability to cocaine addiction. We assess whether the neurobiological mechanisms that modulate repeated-cocaine administration also occur in a chronic restraint stress-induced cocaine self-administration model. We also determine if chronic stress induces alterations in dendritic spines through dysregulation of cofilin activity in the NA core. Here, we show that the inhibition of cofilin expression in the NA core using viral short-hairpin RNA is sufficient to prevent the cocaine sensitization induced by chronic stress. The reduced cofilin levels also impede a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor surface expression enhancement and promote the reduction of head diameter in animals pre-exposed to stress after a cocaine challenge in the NA core. Moreover, downregulation of cofilin expression prevents facilitation of the acquisition of cocaine self-administration (SA) in male rats pre-exposed to chronic stress without modifying performance in sucrose SA. These findings reveal a novel, crucial role for cofilin in the neurobiological mechanisms underpinning the comorbidity between stress exposure and addiction-related disorders.
Collapse
Affiliation(s)
- Daiana Rigoni
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Maria P. Avalos
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Maria J. Boezio
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Andrea S. Guzmán
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Gaston D. Calfa
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Eduardo M. Perassi
- Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC), Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Silvia M. Pierotti
- Cátedra de Bioestadística I y II (Departamento de Matemática), Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Avenida Velez Sarfield 161, (5000), Córdoba, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Friuli 2434, Colinas de Vélez Sarsfield (5016) Córdoba, Argentina
| | - Constanza Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Liliana M. Cancela
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| | - Flavia Bollati
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, (5000), Córdoba, Argentina
| |
Collapse
|
39
|
Décarie-Spain L, Hryhorczuk C, Lau D, Jacob-Brassard É, Fisette A, Fulton S. Prolonged saturated, but not monounsaturated, high-fat feeding provokes anxiodepressive-like behaviors in female mice despite similar metabolic consequences. Brain Behav Immun Health 2021; 16:100324. [PMID: 34589811 PMCID: PMC8474568 DOI: 10.1016/j.bbih.2021.100324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 11/18/2022] Open
Abstract
Obesity significantly increases the risk for anxiety and depression. Our group has recently demonstrated a role for nucleus accumbens (NAc) pro-inflammatory nuclear factor kappa-B (NFkB) signaling in the development of anxiodepressive-like behaviors by diet-induced obesity in male mice. The NAc is a brain region involved in goal-oriented behavior and mood regulation whose functions are critical to hedonic feeding and motivation. While the incidence of depression and anxiety disorders is significantly higher in women than in men, the use of female animal models in psychiatric research remains limited. We set out to investigate the impact of chronic intake of saturated and monounsaturated high-fat diets (HFD) on energy metabolism and on anxiety- and despair-like behaviors in female mice and to ascertain the contribution of NAc NFkB-mediated inflammation herein. Adult C57Bl6N female mice were fed either a saturated HFD, an isocaloric monounsaturated HFD or a control low-fat diet for 24 weeks, after which metabolic profiling and behavioral testing for anxiodepressive-like behaviors were conducted. Plasma was collected at time of sacrifice for quantification of leptin, inflammatory markers as well as 17 β-estradiol levels and brains were harvested to analyze NAc expression of pro-inflammatory genes and estrogen-signaling molecules. In another group of female mice placed on the saturated HFD or the control diet for 24 weeks, we performed adenoviral-mediated invalidation of the NFkB signaling pathway in the NAc prior to behavioral testing. While both HFDs provoked obesity and metabolic impairments, only the saturated HFD triggered anxiodepressive-like behaviors and caused marked elevations in plasma estrogen. This saturated HFD-specific behavioral phenotype could not be explained by NAc inflammation alone and was unaffected by NAc invalidation of the NFkB signaling pathway. Instead, we found changes in the expression of estrogen signaling markers. Such results diverge from the inflammatory mechanisms underlying diet- and obesity-induced metabolic dysfunction and anxiodepressive-like behavior onset in male mice and call attention to the role of estrogen signaling in diet-related anxiodepressive-like phenotypes in female mice.
Collapse
Affiliation(s)
- Léa Décarie-Spain
- Centre de recherche du CHUM & Montreal Diabetes Research Centre, Canada.,Department of Neuroscience, Faculty of Medicine, University of Montreal, Canada
| | - Cécile Hryhorczuk
- Centre de recherche du CHUM & Montreal Diabetes Research Centre, Canada
| | - David Lau
- Centre de recherche du CHUM & Montreal Diabetes Research Centre, Canada.,Department of Neuroscience, Faculty of Medicine, University of Montreal, Canada
| | | | - Alexandre Fisette
- Centre de recherche du CHUM & Montreal Diabetes Research Centre, Canada.,Department of Nutrition, Faculty of Medicine, University of Montreal, Canada
| | - Stephanie Fulton
- Centre de recherche du CHUM & Montreal Diabetes Research Centre, Canada.,Department of Nutrition, Faculty of Medicine, University of Montreal, Canada
| |
Collapse
|
40
|
Bravo-Tobar ID, Fernández P, Sáez JC, Dagnino-Subiabre A. Long-term effects of stress resilience: Hippocampal neuroinflammation and behavioral approach in male rats. J Neurosci Res 2021; 99:2493-2510. [PMID: 34184764 DOI: 10.1002/jnr.24902] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 01/28/2023]
Abstract
Resilience to stress is the ability to quickly adapt to adversity. There is evidence that exposure to prolonged stress triggers neuroinflammation what produces individual differences in stress vulnerability. However, the relationship between stress resilience, neuroinflammation, and depressive-like behaviors remains unknown. The aim of this study was to analyze the long-term effects of social defeat stress (SDS) on neuroinflammation in the hippocampus and depressive-like behaviors. Male rats were subjected to the SDS paradigm. Social interaction was analyzed 1 and 2 weeks after ending the SDS to determine which animals were susceptible or resilient to stress. Neuroinflammation markers glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, and elevated membrane permeability in astrocytes and microglia, as well as depressive-like behaviors in the sucrose preference test and forced swim test were evaluated in all rats. One week after SDS, resilient rats increased their sucrose preference, and time spent in the floating behavior decreased in the forced swim test compared to susceptible rats. Surprisingly, resilient rats became susceptible to stress, and presented neuroinflammation 2 weeks after SDS. These findings suggest that SDS-induced hippocampal neuroinflammation persists in post-stress stages, regardless of whether rats were initially resilient or not. Our study opens a new approach to understanding the neurobiology of stress resilience.
Collapse
Affiliation(s)
- Iván D Bravo-Tobar
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Paola Fernández
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Juan C Sáez
- Instituto de Neurociencia, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Centre for Integrative Neurobiology and Pathophysiology, Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
41
|
Kimura LF, Novaes LS, Picolo G, Munhoz CD, Cheung CW, Camarini R. How environmental enrichment balances out neuroinflammation in chronic pain and comorbid depression and anxiety disorders. Br J Pharmacol 2021; 179:1640-1660. [PMID: 34076891 DOI: 10.1111/bph.15584] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/05/2021] [Accepted: 05/17/2021] [Indexed: 11/30/2022] Open
Abstract
Depression and anxiety commonly occur in chronic pain states and the coexistence of these diseases worsens outcomes for both disorders and may reduce treatment adherence and response. Despite the advances in the knowledge of chronic pain mechanisms, pharmacological treatment is still unsatisfactory. Research based on exposure to environmental enrichment is currently under investigation and seems to offer a promising low-cost strategy with no side effects. In this review, we discuss the role of inflammation as a major biological substrate and aetiological factor of chronic pain and depression/anxiety and report a collection of preclinical evidence of the effects and mechanisms of environmental enrichment. As microglia participates in the development of both conditions, we also discuss microglia as a potential target underlying the beneficial actions of environmental enrichment in chronic pain and comorbid depression/anxiety. We also discuss how alternative interventions under clinical guidelines, such as environmental enrichment, may improve treatment compliance and patient outcomes.
Collapse
Affiliation(s)
- Louise F Kimura
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo, Brazil
| | - Leonardo S Novaes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, São Paulo, Brazil
| | - Carolina D Munhoz
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Chi W Cheung
- Department of Anesthesiology, University of Hong Kong, Hong Kong
| | - Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Gebara E, Zanoletti O, Ghosal S, Grosse J, Schneider BL, Knott G, Astori S, Sandi C. Mitofusin-2 in the Nucleus Accumbens Regulates Anxiety and Depression-like Behaviors Through Mitochondrial and Neuronal Actions. Biol Psychiatry 2021; 89:1033-1044. [PMID: 33583561 DOI: 10.1016/j.biopsych.2020.12.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 11/11/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Emerging evidence points to a central role of mitochondria in psychiatric disorders. However, little is known about the molecular players that regulate mitochondria in neural circuits regulating anxiety and depression and about how they impact neuronal structure and function. Here, we investigated the role of molecules involved in mitochondrial dynamics in medium spiny neurons (MSNs) from the nucleus accumbens (NAc), a hub of the brain's motivation system. METHODS We assessed how individual differences in anxiety-like (measured via the elevated plus maze and open field tests) and depression-like (measured via the forced swim and saccharin preference tests) behaviors in outbred rats relate to mitochondrial morphology (electron microscopy and 3-dimensional reconstructions) and function (mitochondrial respirometry). Mitochondrial molecules were measured for protein (Western blot) and messenger RNA (quantitative reverse transcriptase polymerase chain reaction, RNAscope) content. Dendritic arborization (Golgi Sholl analyses), spine morphology, and MSN excitatory inputs (patch-clamp electrophysiology) were characterized. MFN2 overexpression in the NAc was induced through an AAV9-syn1-MFN2. RESULTS Highly anxious animals showed increased depression-like behaviors, as well as reduced expression of the mitochondrial GTPase MFN2 in the NAc. They also showed alterations in mitochondria (i.e., respiration, volume, and interactions with the endoplasmic reticulum) and MSNs (i.e., dendritic complexity, spine density and typology, and excitatory inputs). Viral MFN2 overexpression in the NAc reversed all of these behavioral, mitochondrial, and neuronal phenotypes. CONCLUSIONS Our results implicate a causal role for accumbal MFN2 on the regulation of anxiety and depression-like behaviors through actions on mitochondrial and MSN structure and function. MFN2 is posited as a promising therapeutic target to treat anxiety and associated behavioral disturbances.
Collapse
Affiliation(s)
- Elias Gebara
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sriparna Ghosal
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bernard L Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Graham Knott
- Biological Electron Microscopy Facility, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Simone Astori
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
43
|
Bortolozzi A, Manashirov S, Chen A, Artigas F. Oligonucleotides as therapeutic tools for brain disorders: Focus on major depressive disorder and Parkinson's disease. Pharmacol Ther 2021; 227:107873. [PMID: 33915178 DOI: 10.1016/j.pharmthera.2021.107873] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/05/2021] [Indexed: 12/25/2022]
Abstract
Remarkable advances in understanding the role of RNA in health and disease have expanded considerably in the last decade. RNA is becoming an increasingly important target for therapeutic intervention; therefore, it is critical to develop strategies for therapeutic modulation of RNA function. Oligonucleotides, including antisense oligonucleotide (ASO), small interfering RNA (siRNA), microRNA mimic (miRNA), and anti-microRNA (antagomir) are perhaps the most direct therapeutic strategies for addressing RNA. Among other mechanisms, most oligonucleotide designs involve the formation of a hybrid with RNA that promotes its degradation by activation of endogenous enzymes such as RNase-H (e.g., ASO) or the RISC complex (e.g. RNA interference - RNAi for siRNA and miRNA). However, the use of oligonucleotides for the treatment of brain disorders is seriously compromised by two main limitations: i) how to deliver oligonucleotides to the brain compartment, avoiding the action of peripheral RNAses? and once there, ii) how to target specific neuronal populations? We review the main molecular pathways in major depressive disorder (MDD) and Parkinson's disease (PD), and discuss the challenges associated with the development of novel oligonucleotide therapeutics. We pay special attention to the use of conjugated ligand-oligonucleotide approach in which the oligonucleotide sequence is covalently bound to monoamine transporter inhibitors (e.g. sertraline, reboxetine, indatraline). This strategy allows their selective accumulation in the monoamine neurons of mice and monkeys after their intranasal or intracerebroventricular administration, evoking preclinical changes predictive of a clinical therapeutic action after knocking-down disease-related genes. In addition, recent advances in oligonucleotide therapeutic clinical trials are also reviewed.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain; Institut d'Investigacions August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain.
| | - Sharon Manashirov
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain; miCure Therapeutics LTD., Tel-Aviv, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Francesc Artigas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), 08036 Barcelona, Spain; Institut d'Investigacions August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain
| |
Collapse
|
44
|
Bao H, Li H, Jia Y, Xiao Y, Luo S, Zhang D, Han L, Dai L, Xiao C, Feng L, Feng Y, Yang Y, Wang H, Wang G, Du J. Ganoderic acid A exerted antidepressant-like action through FXR modulated NLRP3 inflammasome and synaptic activity. Biochem Pharmacol 2021; 188:114561. [PMID: 33857491 DOI: 10.1016/j.bcp.2021.114561] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/28/2022]
Abstract
Major depressive disorder (MDD) is a common, chronic, recurrent disease. The existing drugs are ineffective for approximately half of patients, so the development of antidepressant drugs with novel mechanisms is urgent. Cumulative evidence has shown neuro-inflammation plays a key role in the etiology of major depressive disorder. Clinical studies implicated that bile acids, an important component of gut-brain axis, inhibit neuro-inflammation and mediate the pathophysiology of the MDD. Here, we found that ganoderic acid A (GAA) modulated bile acid receptor FXR (farnesoid X receptor), inhibited brain inflammatory activity, and showed antidepressant effects in the chronic social defeat stress depression model, tail suspension, forced swimming, and sucrose preference tests. GAA directly inhibited the activity of the NLRP3 inflammasome, and activated the phosphorylation and expression of the AMPA receptor by modulating FXR in the prefrontal cortex of mice. If we knocked out FXR or injected the FXR-specific inhibitor z-gugglesterone (GS), the antidepressant effects induced by GAA were completely abolished. These results suggest that GAA modulates the bile acid receptor FXR and subsequently regulates neuroimmune and antidepressant behaviors. GAA and its receptor FXR have potential as targets for the treatment of MDD.
Collapse
Affiliation(s)
- Hongkun Bao
- School of Medicine, Yunnan University, Kunming 650091, China.
| | - Haoran Li
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Yue Jia
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming 650118, China
| | - Yuhuan Xiao
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Shaolei Luo
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Dandan Zhang
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Li Han
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Lili Dai
- School of Agronomy and Life Sciences, Kunming University, Kunming 650214, China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming 650091, China
| | - Lei Feng
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Yuan Feng
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Yang Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Han Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China
| | - Jing Du
- School of Medicine, Yunnan University, Kunming 650091, China; The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital & Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100088, China.
| |
Collapse
|
45
|
Potiris A, Manousopoulou A, Zouridis A, Sarli PM, Pervanidou P, Eliades G, Perrea DN, Deligeoroglou E, Garbis SD, Eleftheriades M. The Effect of Prenatal Food Restriction on Brain Proteome in Appropriately Grown and Growth Restricted Male Wistar Rats. Front Neurosci 2021; 15:665354. [PMID: 33935642 PMCID: PMC8079747 DOI: 10.3389/fnins.2021.665354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Background Fetal growth restriction (FGR) has been associated with a higher risk of developing adverse perinatal outcomes and distinct neurodevelopmental and neurobehavioral disorders. The aim of the present study was to investigate the impact of prenatal food restriction on the brain proteome in both FGR and appropriately grown rats and to identify potential pathways connecting maternal malnutrition with altered brain development. Methods Ten time-dated pregnant Wistar rats were housed individually at their 12th day of gestation. On the 15th day of gestation, the rats were randomly divided into two groups, namely the food restricted one (n = 6) and the control group (n = 4). From days 15 to 21 the control group had unlimited access to food and the food restricted group was given half the amount of food that was on average consumed by the control group, based on measurements taken place the day before. On the 21st day of gestation, all rats delivered spontaneously and after birth all newborn pups of the food restricted group were weighed and matched as appropriately grown (non-FGR) or growth restricted (FGR) and brain tissues were immediately collected. A multiplex experiment was performed analyzing brain tissues from 4 FGR, 4 non-FGR, and 3 control male offspring. Differentially expressed proteins (DEPs) were subjected to bioinformatics analysis in order to identify over-represented processes. Results Proteomic analysis resulted in the profiling of 3,964 proteins. Gene ontology analysis of the common DEPs using DAVID (https://david.ncifcrf.gov/) showed significant enrichment for terms related to cellular morphology, learning, memory and positive regulation of NF-kappaB signaling. Ingenuity Pathway Analysis showed significant induction of inflammation in FGR pups, whereas significant induction of cell migration and cell spreading were observed in non-FGR pups. Conclusion This study demonstrated that in both FGR and non-FGR neonates, a range of adaptive neurodevelopmental processes takes place, which may result in altered cellular morphology, chronic stress, poor memory and learning outcomes. Furthermore, this study highlighted that not only FGR, but also appropriately grown pups, which have been exposed to prenatal food deprivation may be at increased risk for impaired cognitive and developmental outcomes.
Collapse
Affiliation(s)
- Anastasios Potiris
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Manousopoulou
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Andreas Zouridis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Polyxeni-Maria Sarli
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Paediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George Eliades
- Biomaterials Laboratory, School of Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina N Perrea
- Laboratory of Experimental Surgery and Surgical Research "N.S. Christeas", Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymios Deligeoroglou
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spiros D Garbis
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States.,Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
46
|
Bekhbat M, Mukhara D, Dozmorov MG, Stansfield JC, Benusa SD, Hyer MM, Rowson SA, Kelly SD, Qin Z, Dupree JL, Tharp GK, Tansey MG, Neigh GN. Adolescent stress sensitizes the adult neuroimmune transcriptome and leads to sex-specific microglial and behavioral phenotypes. Neuropsychopharmacology 2021; 46:949-958. [PMID: 33558677 PMCID: PMC8115118 DOI: 10.1038/s41386-021-00970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 01/30/2023]
Abstract
Adolescent exposure to chronic stress, a risk factor for mood disorders in adulthood, sensitizes the neuroinflammatory response to a subsequent immune challenge. We previously showed that chronic adolescent stress (CAS) in rats led to distinct patterns of neuroimmune priming in adult male and female rats. However, sex differences in the neuroimmune consequences of CAS and their underlying mechanisms are not fully understood. Here we hypothesized that biological sex would dictate differential induction of inflammation-related transcriptomic pathways and immune cell involvement (microglia activation and leukocyte presence) in the hippocampus of male and female rats with a history of CAS. Adolescent rats underwent CAS (six restraint and six social defeat episodes during postnatal days 38-49), and behavioral assessments were conducted in adolescence and adulthood. Neuroimmune measures were obtained following vehicle or a systemic lipopolysaccharide (LPS) challenge in adulthood. CAS led to increased time in the corners of the open field in adolescence. In males, CAS also increased social avoidance. As adults, CAS rats displayed an exaggerated enrichment of the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) pathway and chemokine induction following LPS challenge, and increased number of perivascular CD45+ cells in the hippocampus. However, CAS females, but not males, showed exaggerated glucocorticoid receptor (GR) pathway enrichment and increased microglial complexity. These results provide further insight to the mechanisms by which peripheral immune events may influence neuroimmune responses differentially among males and females and further demonstrate the importance of adolescent stress in shaping adult responses.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Deepika Mukhara
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Mikhail G. Dozmorov
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - John C. Stansfield
- grid.417264.20000 0001 2194 2791Center for Clinical and Translational Research, Virginia Commonwealth University Medical Center, Richmond, VA 23298 USA
| | - Savannah D. Benusa
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Molly M. Hyer
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Sydney A. Rowson
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Sean D. Kelly
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Zhaohui Qin
- grid.189967.80000 0001 0941 6502Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Jeffrey L. Dupree
- grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA
| | - Gregory K. Tharp
- grid.189967.80000 0001 0941 6502Division of Developmental & Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30322 USA
| | - Malú G. Tansey
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA
| | - Gretchen N. Neigh
- grid.189967.80000 0001 0941 6502Department of Physiology, Emory University, Atlanta, GA 30322 USA ,grid.224260.00000 0004 0458 8737Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298 USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322 USA
| |
Collapse
|
47
|
PPM1F in hippocampal dentate gyrus regulates the depression-related behaviors by modulating neuronal excitability. Exp Neurol 2021; 340:113657. [PMID: 33639208 DOI: 10.1016/j.expneurol.2021.113657] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/28/2020] [Accepted: 02/21/2021] [Indexed: 01/21/2023]
Abstract
Major depressive disorder (MDD) is a common, serious, debilitating mental illness. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been reported to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on depressive behaviors remain largely unknown. Here, we showed that PPM1F is widely distributed in the hippocampus, and chronic unpredictable stress (CUS) can induce increased expression of PPM1F in the hippocampus, which was correlated with depression-associated behaviors. Overexpression of PPM1F mediated by adeno-associated virus (AAV) in the dentate gyrus (DG) produced depression-related behaviors and enhanced susceptibility to subthreshold CUS (SCUS) in both male and female mice, while, knockout of PPM1F in DG produced antidepressant phonotypes under stress conditions. Whole-cell patch-clamp recordings demonstrated that overexpression of PPM1F increased the neuronal excitability of the granule cells in the DG. Consistent with neuronal hyperexcitability, overexpression of PPM1F regulated the expression of certain ion channel genes and induced decreased phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in hippocampus. These results suggest that PPM1F in the DG regulates depression-related behaviors by modulating neuronal excitability, which might be an important pathological gene for depression or other mental diseases.
Collapse
|
48
|
Kirouac GJ. The Paraventricular Nucleus of the Thalamus as an Integrating and Relay Node in the Brain Anxiety Network. Front Behav Neurosci 2021; 15:627633. [PMID: 33732118 PMCID: PMC7959748 DOI: 10.3389/fnbeh.2021.627633] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/28/2021] [Indexed: 12/25/2022] Open
Abstract
The brain anxiety network is composed of a number of interconnected cortical regions that detect threats and execute appropriate defensive responses via projections to the shell of the nucleus accumbens (NAcSh), dorsolateral region of the bed nucleus of the stria terminalis (BSTDL) and lateral region of the central nucleus of the amygdala (CeL). The paraventricular nucleus of the thalamus (PVT) is anatomically positioned to integrate threat- and arousal-related signals from cortex and hypothalamus and then relay these signals to neural circuits in the NAcSh, BSTDL, and CeL that mediate defensive responses. This review describes the anatomical connections of the PVT that support the view that the PVT may be a critical node in the brain anxiety network. Experimental findings are reviewed showing that the arousal peptides orexins (hypocretins) act at the PVT to promote avoidance of potential threats especially following exposure of rats to a single episode of footshocks. Recent anatomical and experimental findings are discussed which show that neurons in the PVT provide divergent projections to subcortical regions that mediate defensive behaviors and that the projection to the NAcSh is critical for the enhanced social avoidance displayed in rats exposed to footshocks. A theoretical model is proposed for how the PVT integrates cortical and hypothalamic signals to modulate the behavioral responses associated with anxiety and other challenging situations.
Collapse
Affiliation(s)
- Gilbert J. Kirouac
- Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
49
|
Cooperative synaptic and intrinsic plasticity in a disynaptic limbic circuit drive stress-induced anhedonia and passive coping in mice. Mol Psychiatry 2021; 26:1860-1879. [PMID: 32161361 PMCID: PMC7735389 DOI: 10.1038/s41380-020-0686-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/19/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022]
Abstract
Stress promotes negative affective states, which include anhedonia and passive coping. While these features are in part mediated by neuroadaptations in brain reward circuitry, a comprehensive framework of how stress-induced negative affect may be encoded within key nodes of this circuit is lacking. Here, we show in a mouse model for stress-induced anhedonia and passive coping that these phenomena are associated with increased synaptic strength of ventral hippocampus (VH) excitatory synapses onto D1 medium spiny neurons (D1-MSNs) in the nucleus accumbens medial shell (NAcmSh), and with lateral hypothalamus (LH)-projecting D1-MSN hyperexcitability mediated by decreased inwardly rectifying potassium channel (IRK) function. Stress-induced negative affective states are prevented by depotentiation of VH to NAcmSh synapses, restoring Kir2.1 function in D1R-MSNs, or disrupting co-participation of these synaptic and intrinsic adaptations in D1-MSNs. In conclusion, our data provide strong evidence for a disynaptic pathway controlling maladaptive emotional behavior.
Collapse
|
50
|
Baik JH. Stress and the dopaminergic reward system. Exp Mol Med 2020; 52:1879-1890. [PMID: 33257725 PMCID: PMC8080624 DOI: 10.1038/s12276-020-00532-4] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022] Open
Abstract
Dopamine regulates reward-related behavior through the mesolimbic dopaminergic pathway. Stress affects dopamine levels and dopaminergic neuronal activity in the mesolimbic dopamine system. Changes in mesolimbic dopaminergic neurotransmission are important for coping with stress, as they allow adaption to behavioral responses to various environmental stimuli. Upon stress exposure, modulation of the dopaminergic reward system is necessary for monitoring and selecting the optimal process for coping with stressful situations. Aversive stressful events may negatively regulate the dopaminergic reward system, perturbing reward sensitivity, which is closely associated with chronic stress-induced depression. The mesolimbic dopamine system is excited not only by reward but also by aversive stressful stimuli, which adds further intriguing complexity to the relationship between stress and the reward system. This review focuses on lines of evidence related to how stress, especially chronic stress, affects the mesolimbic dopamine system, and discusses the role of the dopaminergic reward system in chronic stress-induced depression.
Collapse
Affiliation(s)
- Ja-Hyun Baik
- Molecular Neurobiology Laboratory, Department of Life Sciences, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|