1
|
Chen W, Liu Y, Pu J, Gui S, Wang D, Zhong X, Tao W, Chen X, Chen X, Chen Y, Zhao L, Wu Q, Chen X, Zhang Y, Xie A, Xie P. Comparative transcriptional analyses of the striatum in the chronic social defeat stress model in C57BL/6J male mice and the gut microbiota-dysbiosis model in Kumming mice. Neuroscience 2024; 562:217-226. [PMID: 39489477 DOI: 10.1016/j.neuroscience.2024.10.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/11/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Depression is a complex disorder with multiple contributing factors, and chronic stress has previously been recognized as a major causative factor, while gut microbes have also been found to be involved in depression recently. However, gene expression in depression models with different etiologies is unclear. Here, we compared the transcriptomes of the striatum in chronic social defeat stress (CSDS) model of C57BL/6J male mice and fecal microbiota transplant (FMT) model of Kumming male mice. We found that the proportion of shared differentially expressed genes (DEGs) between the two models was only 24 %. The specific DEGs of FMT model were enriched in immune and inflammatory, and are associated with changes in vascular and ciliated ependymal cells. The specific DEGs of CSDS model were enriched in neuron and synapse. The results of network analysis suggested the expression patterns and biological function of depressive-like behaviors-related modules in the two models are different. Further, the alternative splicing events of CSDS are more than FMT. Our results suggested models of depression induced by different etiologies differ significantly in gene expression and biological function. Our study also suggested us to pay attention to the characteristics of models of depression of different etiologies and provided a more comprehensive understanding of the heterogeneity of depression.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Juncai Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Dongfang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiaogang Zhong
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Wei Tao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiaopeng Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yue Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Libo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University 402160 Chongqing, China
| | - Qingyuan Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing 404000, China
| | - Xiangyu Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China
| | - Yingying Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shan-dong, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shan-dong, China.
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Jin Feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
2
|
Kupferberg A, Hasler G. From antidepressants and psychotherapy to oxytocin, vagus nerve stimulation, ketamine and psychedelics: how established and novel treatments can improve social functioning in major depression. Front Psychiatry 2024; 15:1372650. [PMID: 39469469 PMCID: PMC11513289 DOI: 10.3389/fpsyt.2024.1372650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/05/2024] [Indexed: 10/30/2024] Open
Abstract
Social cognitive deficits and social behavior impairments are common in major depressive disorder (MDD) and affect the quality of life and recovery of patients. This review summarizes the impact of standard and novel treatments on social functioning in MDD and highlights the potential of combining different approaches to enhance their effectiveness. Standard treatments, such as antidepressants, psychotherapies, and brain stimulation, have shown mixed results in improving social functioning, with some limitations and side effects. Newer treatments, such as intranasal oxytocin, mindfulness-based cognitive therapy, and psychedelic-assisted psychotherapy, have demonstrated positive effects on social cognition and behavior by modulating self-referential processing, empathy, and emotion regulation and through enhancement of neuroplasticity. Animal models have provided insights into the neurobiological mechanisms underlying these treatments, such as the role of neuroplasticity. Future research should explore the synergistic effects of combining different treatments and investigate the long-term outcomes and individual differences in response to these promising interventions.
Collapse
Affiliation(s)
- Aleksandra Kupferberg
- Molecular Psychiatry Lab, Faculty of Science and Medicine, University of Freiburg, Villars-sur-Glâne, Switzerland
| | - Gregor Hasler
- Molecular Psychiatry Lab, Faculty of Science and Medicine, University of Freiburg, Villars-sur-Glâne, Switzerland
- University Psychiatry Research Unit, Freiburg Mental Health Network, Villars-sur-Glâne, Switzerland
- Department of Neuropsychology, Lake Lucerne Institute, Vitznau, Switzerland
| |
Collapse
|
3
|
Burek DJ, Ibrahim KM, Hall AG, Sharma A, Musiek ES, Morón JA, Carlezon WA. Inflammatory pain in mice induces light cycle-dependent effects on sleep architecture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610124. [PMID: 39257818 PMCID: PMC11383991 DOI: 10.1101/2024.08.28.610124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
As a syndrome, chronic pain comprises physical, emotional, and cognitive symptoms such as disability, negative affect, feelings of stress, and fatigue. A rodent model of long-term inflammatory pain, induced by complete Freund's adjuvant (CFA) injection, has previously been shown to cause anhedonia and dysregulated naturalistic behaviors, in a manner similar to animal models of stress. We examined whether this extended to alterations in circadian rhythms and sleep, such as those induced by chronic social defeat stress, using actigraphy and wireless EEG. CFA-induced inflammatory pain profoundly altered sleep architecture in male and female mice. Injection of the hind paw, whether with CFA or saline, reduced some measures of circadian rhythmicity such as variance, period, and amplitude. CFA increased sleep duration primarily in the dark phase, while sleep bout length was decreased in the light and increased in the dark phase. Additionally, CFA reduced wake bout length, especially during the dark phase. Increases in REM and SWS duration and bouts were most significant in the dark phase, regardless of whether CFA had been injected at its onset or 12 hours prior. Taken together, these results indicate that inflammatory pain acutely promotes but also fragments sleep.
Collapse
|
4
|
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
|
5
|
Warren AL, Lankri D, Cunningham MJ, Serrano IC, Parise LF, Kruegel AC, Duggan P, Zilberg G, Capper MJ, Havel V, Russo SJ, Sames D, Wacker D. Structural pharmacology and therapeutic potential of 5-methoxytryptamines. Nature 2024; 630:237-246. [PMID: 38720072 PMCID: PMC11152992 DOI: 10.1038/s41586-024-07403-2] [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: 02/14/2023] [Accepted: 04/09/2024] [Indexed: 06/07/2024]
Abstract
Psychedelic substances such as lysergic acid diethylamide (LSD) and psilocybin show potential for the treatment of various neuropsychiatric disorders1-3. These compounds are thought to mediate their hallucinogenic and therapeutic effects through the serotonin (5-hydroxytryptamine (5-HT)) receptor 5-HT2A (ref. 4). However, 5-HT1A also plays a part in the behavioural effects of tryptamine hallucinogens5, particularly 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT), a psychedelic found in the toxin of Colorado River toads6. Although 5-HT1A is a validated therapeutic target7,8, little is known about how psychedelics engage 5-HT1A and which effects are mediated by this receptor. Here we map the molecular underpinnings of 5-MeO-DMT pharmacology through five cryogenic electron microscopy (cryo-EM) structures of 5-HT1A, systematic medicinal chemistry, receptor mutagenesis and mouse behaviour. Structure-activity relationship analyses of 5-methoxytryptamines at both 5-HT1A and 5-HT2A enable the characterization of molecular determinants of 5-HT1A signalling potency, efficacy and selectivity. Moreover, we contrast the structural interactions and in vitro pharmacology of 5-MeO-DMT and analogues to the pan-serotonergic agonist LSD and clinically used 5-HT1A agonists. We show that a 5-HT1A-selective 5-MeO-DMT analogue is devoid of hallucinogenic-like effects while retaining anxiolytic-like and antidepressant-like activity in socially defeated animals. Our studies uncover molecular aspects of 5-HT1A-targeted psychedelics and therapeutics, which may facilitate the future development of new medications for neuropsychiatric disorders.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- 5-Methoxytryptamine/analogs & derivatives
- 5-Methoxytryptamine/chemistry
- 5-Methoxytryptamine/pharmacology
- 5-Methoxytryptamine/therapeutic use
- Anti-Anxiety Agents/chemistry
- Anti-Anxiety Agents/pharmacology
- Anti-Anxiety Agents/therapeutic use
- Antidepressive Agents/chemistry
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Cryoelectron Microscopy
- Hallucinogens
- Lysergic Acid Diethylamide/chemistry
- Lysergic Acid Diethylamide/pharmacology
- Methoxydimethyltryptamines/chemistry
- Methoxydimethyltryptamines/pharmacology
- Methoxydimethyltryptamines/therapeutic use
- Models, Molecular
- Receptor, Serotonin, 5-HT1A/chemistry
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT1A/ultrastructure
- Receptor, Serotonin, 5-HT2A/chemistry
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptor, Serotonin, 5-HT2A/ultrastructure
- Serotonin Receptor Agonists/chemistry
- Serotonin Receptor Agonists/pharmacology
- Serotonin Receptor Agonists/therapeutic use
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Audrey L Warren
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Lankri
- Department of Chemistry, Columbia University, New York, NY, USA
| | | | - Inis C Serrano
- Department of Chemistry, Columbia University, New York, NY, USA
| | - Lyonna F Parise
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Gregory Zilberg
- Zuckerman Institute of Mind, Brain, Behavior, Columbia University, New York, NY, USA
| | - Michael J Capper
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vaclav Havel
- Department of Chemistry, Columbia University, New York, NY, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY, USA.
- Zuckerman Institute of Mind, Brain, Behavior, Columbia University, New York, NY, USA.
| | - Daniel Wacker
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Rodriguez M, Themann A, Garcia-Carachure I, Lira O, Robison AJ, Cushing BS, Iñiguez SD. Chronic social defeat stress in prairie voles (Microtus ochrogaster): A preclinical model for the study of depression-related phenotypes. J Affect Disord 2024; 351:833-842. [PMID: 38341153 DOI: 10.1016/j.jad.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Stress-induced illnesses, like major depression, are among the leading causes of disability across the world. Consequently, there is a dire need for the validation of translationally-suited animal models incorporating social stress to uncover the etiology of depression. Prairie voles (Microtus ochrogaster) are more translationally relevant than many other rodent models as they display monogamous social and bi-parental behaviors. Therefore, we evaluated whether a novel social defeat stress (SDS) model in male prairie voles induces depression-relevant behavioral outcomes. METHODS Adult sexually-naïve male prairie voles experienced SDS bouts from a conspecific pair-bonded male aggressor, 10 min per day for 10 consecutive days. Non-stressed controls (same-sex siblings) were housed in similar conditions but never experienced physical stress. Twenty-four h later, voles were evaluated in social interaction, sucrose preference, and Morris water maze tests - behavioral endpoints validated to assess social withdrawal, anhedonia-related behavior, and spatial memory performance, respectively. RESULTS SDS-exposed voles displayed lower sociability and body weight, decreased preference for a sucrose solution, and impairment of spatial memory retrieval. Importantly, no differences in general locomotor activity were observed as a function of SDS exposure. LIMITATIONS This study does not include female voles in the experimental design. CONCLUSIONS We found that repeated SDS exposure, in male prairie voles, results in a depression-relevant phenotype resembling an anhedonia-like outcome (per reductions in sucrose preference) along with social withdrawal and spatial memory impairment - highlighting that the prairie vole is a valuable model with potential to study the neurobiology of social stress-induced depression-related outcomes.
Collapse
Affiliation(s)
- Minerva Rodriguez
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Anapaula Themann
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | | | - Omar Lira
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Bruce S Cushing
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, United States
| | - Sergio D Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, TX, United States.
| |
Collapse
|
7
|
Zhang L, Wang Y, Li S, Otani S, Chen F. Post-stress Social Interaction and 3-Cyano-N-(1,3-Diphenyl-1H-Pyrazol-5-yl) Benzamide Treatment Attenuate Depressive-like Behavior Induced by Repeated Social Defeat Stress. Neuroscience 2024; 538:11-21. [PMID: 38103860 DOI: 10.1016/j.neuroscience.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Persistent stress increases the probability for developing depression significantly thereafter. Repeated social defeat stress is a widely used model to investigate depressive-like behavior in preclinical models. Hence, the repeated social defeat stress model provided an ideal animal model, through which the hypotheses of prevention and treatment can be investigated. We have successfully induced depressive-like behavior for male C57BL/6J mice with this model. Here, we reported that certain level of during-stress social interactions with single female or multiple male peer(s) exerted a positive role in preventing the development of depressive-like behavior induced by repeated social defeat stress. Our data suggested that the stress-susceptible mice may benefit from positive social interaction, which reduces the chance for depressive-like behavior development. Since numerous studies indicate that the metabotropic glutamate receptor 5 (mGluR5) plays an important role in various cognitive functions, we further investigate the treatment effect of 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) on the depressive-like behavior induced by repeated social defeat stress. Most importantly, robust anti-depressant effects have been achieved through modulating the mGluR5 function. We found that single oral dose administration of CDPPB (20 mg/kg), to some extent, alleviated the social avoidance behaviors for the stress-susceptible mice. Our data implies that the CDPPB, a positive allosteric modulator of mGluR5, is a promising anti-depressant candidate with limited side effect.
Collapse
Affiliation(s)
- Liangui Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Ying Wang
- Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Shengtian Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Satoru Otani
- Vision Institute, CNRS - INSERM - Sorbonne University, Paris 75012, France.
| | - Fujun Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
8
|
Zhai X, Ai L, Chen D, Zhou D, Han Y, Ji R, Hu M, Wang Q, Zhang M, Wang Y, Zhang C, Yang JX, Hu A, Liu H, Cao JL, Zhang H. Multiple integrated social stress induces depressive-like behavioral and neural adaptations in female C57BL/6J mice. Neurobiol Dis 2024; 190:106374. [PMID: 38097092 DOI: 10.1016/j.nbd.2023.106374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/25/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
Despite women representing most of those affected by major depression, preclinical studies have focused almost exclusively on male subjects, partially due to a lack of ideal animal paradigms. As the persistent need regarding the sex balance of neuroscience research and female-specific pathology of mental disorders surges, the establishment of natural etiology-based and systematically validated animal paradigms for depression with female subjects becomes an urgent scientific problem. This study aims to establish, characterize, and validate a "Multiple Integrated Social Stress (MISS)" model of depression in female C57BL/6J mice by manipulating and integrating daily social stressors that females are experiencing. Female C57BL/6J mice randomly experienced social competition failure in tube test, modified vicarious social defeat stress, unescapable overcrowding stress followed by social isolation on each day, for ten consecutive days. Compared with their controls, female MISS mice exhibited a relatively decreased preference for social interaction and sucrose, along with increased immobility in the tail suspension test, which could last for at least one month. These MISS mice also exhibited increased levels of blood serum corticosterone, interleukin-6 L and 1β. In the pharmacological experiment, MISS-induced dysfunctions in social interaction, sucrose preference, and tail suspension tests were amended by systematically administrating a single dose of sub-anesthetic ketamine, a rapid-onset antidepressant. Compared with controls, MISS females exhibited decreased c-Fos activation in their anterior cingulate cortex, prefrontal cortex, nucleus accumbens and some other depression-related brain regions. Furthermore, 24 h after the last exposure to the paradigm, MISS mice demonstrated a decreased center zone time in the open field test and decreased open arm time in the elevated plus-maze test, indicating anxiety-like behavioral phenotypes. Interestingly, MISS mice developed an excessive nesting ability, suggesting a likely behavioral phenotype of obsessive-compulsive disorder. These data showed that the MISS paradigm was sufficient to generate pathological profiles in female mice to mimic core symptoms, serum biochemistry and neural adaptations of depression in clinical patients. The present study offers a multiple integrated natural etiology-based animal model tool for studying female stress susceptibility.
Collapse
Affiliation(s)
- Xiaojing Zhai
- 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
| | - Lin Ai
- 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
| | - Dandan 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
| | - Dongyu Zhou
- 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
| | - Yi Han
- 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
| | - Ran Ji
- 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
| | - Mengfan Hu
- 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
| | - Qing 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
| | - Moruo Zhang
- 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
| | - Yuxin 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
| | - Chunyan Zhang
- 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
| | - Ankang Hu
- Laboratory Animal Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, PR China
| | - He Liu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine, Huzhou Central Hospital, Huzhou 313003, China; The Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313003, China; The Fifth School of Clinical Medicine, Zhejiang Chinese Medical University, Huzhou 313003, China; The Affiliated Central Hospital, Huzhou University, Huzhou 313003, 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.
| | - Hongxing Zhang
- 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
|
9
|
Gyles TM, Nestler EJ, Parise EM. Advancing preclinical chronic stress models to promote therapeutic discovery for human stress disorders. Neuropsychopharmacology 2024; 49:215-226. [PMID: 37349475 PMCID: PMC10700361 DOI: 10.1038/s41386-023-01625-0] [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: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023]
Abstract
There is an urgent need to develop more effective treatments for stress-related illnesses, which include depression, post-traumatic stress disorder, and anxiety. We view animal models as playing an essential role in this effort, but to date, such approaches have generally not succeeded in developing therapeutics with new mechanisms of action. This is partly due to the complexity of the brain and its disorders, but also to inherent difficulties in modeling human disorders in rodents and to the incorrect use of animal models: namely, trying to recapitulate a human syndrome in a rodent which is likely not possible as opposed to using animals to understand underlying mechanisms and evaluating potential therapeutic paths. Recent transcriptomic research has established the ability of several different chronic stress procedures in rodents to recapitulate large portions of the molecular pathology seen in postmortem brain tissue of individuals with depression. These findings provide crucial validation for the clear relevance of rodent stress models to better understand the pathophysiology of human stress disorders and help guide therapeutic discovery. In this review, we first discuss the current limitations of preclinical chronic stress models as well as traditional behavioral phenotyping approaches. We then explore opportunities to dramatically enhance the translational use of rodent stress models through the application of new experimental technologies. The goal of this review is to promote the synthesis of these novel approaches in rodents with human cell-based approaches and ultimately with early-phase proof-of-concept studies in humans to develop more effective treatments for human stress disorders.
Collapse
Affiliation(s)
- Trevonn M Gyles
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
10
|
Edem EE, Oguntala OA, Ikuelogbon DA, Nebo KE, Fafure AA, Akinluyi ET, Isaac GT, Kunlere OE. Prolonged ketamine therapy differentially rescues psychobehavioural deficits via modulation of nitro-oxidative stress and oxytocin receptors in the gut-brain-axis of chronically-stressed mice. Psychoneuroendocrinology 2023; 158:106370. [PMID: 37678086 DOI: 10.1016/j.psyneuen.2023.106370] [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: 01/18/2023] [Revised: 07/30/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Ketamine is an anaesthetic known to have short but rapid-acting anti-depressant effects; however, the neurobehavioural effects of its prolonged use and its role on the oxytocin system in the gut-brain axis are largely undetermined. Female BALB/c mice were either exposed to the chronic unpredictable mild stress (CUMS) paradigm for 21 days and then treated with ketamine in four doses for 14 days or exposed to CUMS and treated simultaneously in four doses of ketamine during the last two weeks of CUMS exposure. After each dose, the forced swim test was conducted to assess depressive-like behaviour. Before sacrifice, all the mice were subjected to behavioural tests to assess anxiety, memory, and social interaction. Prolonged treatment of depression with ketamine did not rescue depressive-like behaviour. It did, however, improve depression-associated anxiety-like behaviours, short-term memory and social interaction deficits when compared to the stressed untreated mice. Furthermore, ketamine treatment enhanced plasma oxytocin levels, expression of oxytocin receptors; as well as abrogated nitro-oxidative stress biomarkers in the intestinal and hippocampal tissues. Taken together, our findings indicate that while short-term use of ketamine has anti-depressant benefits, its prolonged therapeutic use does not seem to adequately resolve depressive-like behaviour in mice.
Collapse
Affiliation(s)
- Edem Ekpenyong Edem
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria; Department of Anatomy, College of Medicine, University of Lagos, Idi-Araba, Lagos State, Nigeria.
| | - Oluwatomisn Adeyosola Oguntala
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | | | - Kate Eberechukwu Nebo
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Adedamola Adediran Fafure
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Elizabeth Toyin Akinluyi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Godspower Tochukwu Isaac
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Oladunni Eunice Kunlere
- Neuroscience Unit, Department of Human Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
11
|
Hisey EE, Fritsch EL, Newman EL, Ressler KJ, Kangas BD, Carlezon WA. Early life stress in male mice blunts responsiveness in a translationally-relevant reward task. Neuropsychopharmacology 2023; 48:1752-1759. [PMID: 37258714 PMCID: PMC10579416 DOI: 10.1038/s41386-023-01610-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023]
Abstract
Early-life stress (ELS) leaves signatures upon the brain that persist throughout the lifespan and increase the risk of psychiatric illnesses including mood and anxiety disorders. In humans, myriad forms of ELS-including childhood abuse, bullying, poverty, and trauma-are increasingly prevalent. Understanding the signs of ELS, including those associated with psychiatric illness, will enable improved treatment and prevention. Here, we developed a novel procedure to model human ELS in mice and identify translationally-relevant biomarkers of mood and anxiety disorders. We exposed male mice (C57BL/6 J) to an early-life (juvenile) chronic social defeat stress (jCSDS) and examined social interaction and responsivity to reward during adulthood. As expected, jCSDS-exposed mice showed a socially avoidant phenotype in open-field social interaction tests. However, sucrose preference tests failed to demonstrate ELS-induced reductions in choice for the sweetened solution, suggesting no effect on reward function. To explore whether other tasks might be more sensitive to changes in motivation, we tested the mice in the Probabilistic Reward Task (PRT), a procedure often used in humans to study reward learning deficits associated with depressive illness. In a touchscreen PRT variant that was reverse-translated to maximize alignment with the version used in human subjects, mice exposed to jCSDS displayed significant reductions in the tendency to develop response biases for the more richly-rewarded stimulus, a hallmark sign of anhedonia when observed in humans. Our findings suggest that translationally-relevant procedures that utilize the same endpoints across species may enable the development of improved model systems that more accurately predict outcomes in humans.
Collapse
Affiliation(s)
- Erin E Hisey
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Emma L Fritsch
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Emily L Newman
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Brian D Kangas
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| |
Collapse
|
12
|
Giovanniello J, Bravo-Rivera C, Rosenkranz A, Matthew Lattal K. Stress, associative learning, and decision-making. Neurobiol Learn Mem 2023; 204:107812. [PMID: 37598745 DOI: 10.1016/j.nlm.2023.107812] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/02/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Exposure to acute and chronic stress has significant effects on the basic mechanisms of associative learning and memory. Stress can both impair and enhance associative learning depending on type, intensity, and persistence of the stressor, the subject's sex, the context that the stress and behavior is experienced in, and the type of associative learning taking place. In some cases, stress can cause or exacerbate the maladaptive behavior that underlies numerous psychiatric conditions including anxiety disorders, obsessive-compulsive disorder, post-traumatic stress disorder, substance use disorder, and others. Therefore, it is critical to understand how the varied effects of stress, which may normally facilitate adaptive behavior, can also become maladaptive and even harmful. In this review, we highlight several findings of associative learning and decision-making processes that are affected by stress in both human and non-human subjects and how they are related to one another. An emerging theme from this work is that stress biases behavior towards less flexible strategies that may reflect a cautious insensitivity to changing contingencies. We consider how this inflexibility has been observed in different associative learning procedures and suggest that a goal for the field should be to clarify how factors such as sex and previous experience influence this inflexibility.
Collapse
Affiliation(s)
| | - Christian Bravo-Rivera
- Departments of Psychiatry and Anatomy & Neurobiology, University of Puerto Rico School of Medicine, San Juan, PR 00935, United States.
| | - Amiel Rosenkranz
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Chicago Medical School, Rosalind Franklin University of Medicine and Science, United States.
| | - K Matthew Lattal
- Department of Behavioral Neuroscience, Oregon Health & Science University, United States.
| |
Collapse
|
13
|
Boyle CC, Bower JE, Eisenberger NI, Irwin MR. Stress to inflammation and anhedonia: Mechanistic insights from preclinical and clinical models. Neurosci Biobehav Rev 2023; 152:105307. [PMID: 37419230 DOI: 10.1016/j.neubiorev.2023.105307] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
Anhedonia, as evidenced by impaired pleasurable response to reward, reduced reward motivation, and/or deficits in reward-related learning, is a common feature of depression. Such deficits in reward processing are also an important clinical target as a risk factor for depression onset. Unfortunately, reward-related deficits remain difficult to treat. To address this gap and inform the development of effective prevention and treatment strategies, it is critical to understand the mechanisms that drive impairments in reward function. Stress-induced inflammation is a plausible mechanism of reward deficits. The purpose of this paper is to review evidence for two components of this psychobiological pathway: 1) the effects of stress on reward function; and 2) the effects of inflammation on reward function. Within these two areas, we draw upon preclinical and clinical models, distinguish between acute and chronic effects of stress and inflammation, and address specific domains of reward dysregulation. By addressing these contextual factors, the review reveals a nuanced literature which might be targeted for additional scientific inquiry to inform the development of precise interventions.
Collapse
Affiliation(s)
- Chloe C Boyle
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA.
| | - Julienne E Bower
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA; Department of Psychology, UCLA, Los Angeles, CA, USA
| | | | - Michael R Irwin
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, UCLA, USA
| |
Collapse
|
14
|
Bordes J, Miranda L, Reinhardt M, Narayan S, Hartmann J, Newman EL, Brix LM, van Doeselaar L, Engelhardt C, Dillmann L, Mitra S, Ressler KJ, Pütz B, Agakov F, Müller-Myhsok B, Schmidt MV. Automatically annotated motion tracking identifies a distinct social behavioral profile following chronic social defeat stress. Nat Commun 2023; 14:4319. [PMID: 37463994 PMCID: PMC10354203 DOI: 10.1038/s41467-023-40040-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
Severe stress exposure increases the risk of stress-related disorders such as major depressive disorder (MDD). An essential characteristic of MDD is the impairment of social functioning and lack of social motivation. Chronic social defeat stress is an established animal model for MDD research, which induces a cascade of physiological and behavioral changes. Current markerless pose estimation tools allow for more complex and naturalistic behavioral tests. Here, we introduce the open-source tool DeepOF to investigate the individual and social behavioral profile in mice by providing supervised and unsupervised pipelines using DeepLabCut-annotated pose estimation data. Applying this tool to chronic social defeat in male mice, the DeepOF supervised and unsupervised pipelines detect a distinct stress-induced social behavioral pattern, which was particularly observed at the beginning of a novel social encounter and fades with time due to habituation. In addition, while the classical social avoidance task does identify the stress-induced social behavioral differences, both DeepOF behavioral pipelines provide a clearer and more detailed profile. Moreover, DeepOF aims to facilitate reproducibility and unification of behavioral classification by providing an open-source tool, which can advance the study of rodent individual and social behavior, thereby enabling biological insights and, for example, subsequent drug development for psychiatric disorders.
Collapse
Affiliation(s)
- Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Lucas Miranda
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Maya Reinhardt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Emily L Newman
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Lea Maria Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Lotte van Doeselaar
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Clara Engelhardt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Larissa Dillmann
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Shiladitya Mitra
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Benno Pütz
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Felix Agakov
- Pharmatics Limited, Edinburgh, EH16 4UX, Scotland, UK
| | - Bertram Müller-Myhsok
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
15
|
Bordes J, Miranda L, Müller-Myhsok B, Schmidt MV. Advancing social behavioral neuroscience by integrating ethology and comparative psychology methods through machine learning. Neurosci Biobehav Rev 2023; 151:105243. [PMID: 37225062 DOI: 10.1016/j.neubiorev.2023.105243] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/21/2023] [Accepted: 05/20/2023] [Indexed: 05/26/2023]
Abstract
Social behavior is naturally occurring in vertebrate species, which holds a strong evolutionary component and is crucial for the normal development and survival of individuals throughout life. Behavioral neuroscience has seen different influential methods for social behavioral phenotyping. The ethological research approach has extensively investigated social behavior in natural habitats, while the comparative psychology approach was developed utilizing standardized and univariate social behavioral tests. The development of advanced and precise tracking tools, together with post-tracking analysis packages, has recently enabled a novel behavioral phenotyping method, that includes the strengths of both approaches. The implementation of such methods will be beneficial for fundamental social behavioral research but will also enable an increased understanding of the influences of many different factors that can influence social behavior, such as stress exposure. Furthermore, future research will increase the number of data modalities, such as sensory, physiological, and neuronal activity data, and will thereby significantly enhance our understanding of the biological basis of social behavior and guide intervention strategies for behavioral abnormalities in psychiatric disorders.
Collapse
Affiliation(s)
- Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Lucas Miranda
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany; International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Bertram Müller-Myhsok
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
16
|
Larosa A, Wong TP. The hippocampus in stress susceptibility and resilience: Reviewing molecular and functional markers. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110601. [PMID: 35842073 DOI: 10.1016/j.pnpbp.2022.110601] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/22/2022] [Accepted: 07/10/2022] [Indexed: 10/17/2022]
Abstract
Understanding the individual variability that comes with the likelihood of developing stress-related psychopathologies is of paramount importance when addressing mechanisms of their neurobiology. This article focuses on the hippocampus as a region that is highly influenced by chronic stress exposure and that has strong ties to the development of related disorders, such as depression and post-traumatic stress disorder. We first outline three commonly used animal models that have been used to separate animals into susceptible and resilient cohorts. Next, we review molecular and functional hippocampal markers of susceptibility and resilience. We propose that the hippocampus plays a crucial role in the differences in the processing and storage of stress-related information in animals with different stress susceptibilities. These hippocampal markers not only help us attain a more comprehensive understanding of the various facets of stress-related pathophysiology, but also could be targeted for the development of new treatments.
Collapse
Affiliation(s)
- Amanda Larosa
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Tak Pan Wong
- Neuroscience Division, Douglas Research Centre, Montreal, QC, Canada; Dept. of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
17
|
McCullough KM, Katrinli S, Hartmann J, Lori A, Klengel C, Missig G, Klengel T, Langford NA, Newman EL, Anderson KJ, Smith AK, Carroll FI, Ressler KJ, Carlezon WA. Blood levels of T-Cell Receptor Excision Circles (TRECs) provide an index of exposure to traumatic stress in mice and humans. Transl Psychiatry 2022; 12:423. [PMID: 36192377 PMCID: PMC9530209 DOI: 10.1038/s41398-022-02159-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Exposure to stress triggers biological changes throughout the body. Accumulating evidence indicates that alterations in immune system function are associated with the development of stress-associated illnesses such as major depressive disorder and post-traumatic stress disorder, increasing interest in identifying immune markers that provide insight into mental health. Recombination events during T-cell receptor rearrangement and T-cell maturation in the thymus produce circular DNA fragments called T-cell receptor excision circles (TRECs) that can be utilized as indicators of thymic function and numbers of newly emigrating T-cells. Given data suggesting that stress affects thymus function, we examined whether blood levels of TRECs might serve as a quantitative peripheral index of cumulative stress exposure and its physiological correlates. We hypothesized that chronic stress exposure would compromise thymus function and produce corresponding decreases in levels of TRECs. In male mice, exposure to chronic social defeat stress (CSDS) produced thymic involution, adrenal hypertrophy, and decreased levels of TRECs in blood. Extending these studies to humans revealed robust inverse correlations between levels of circulating TRECs and childhood emotional and physical abuse. Cell-type specific analyses also revealed associations between TREC levels and blood cell composition, as well as cell-type specific methylation changes in CD4T + and CD8T + cells. Additionally, TREC levels correlated with epigenetic age acceleration, a common biomarker of stress exposure. Our findings demonstrate alignment between findings in mice and humans and suggest that blood-borne TRECs are a translationally-relevant biomarker that correlates with, and provides insight into, the cumulative physiological and immune-related impacts of stress exposure in mammals.
Collapse
Affiliation(s)
- Kenneth M McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Adriana Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Claudia Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Torsten Klengel
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Nicole A Langford
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Emily L Newman
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| | - Kerry J Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - William A Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
18
|
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: 5.5] [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
|
19
|
Yoshida M, Hasegawa S, Taniguchi M, Mouri A, Suzuki C, Yoshimi A, Mamiya T, Ozaki N, Noda Y. Memantine ameliorates the impairment of social behaviors induced by a single social defeat stress as juveniles. Neuropharmacology 2022; 217:109208. [PMID: 35926580 DOI: 10.1016/j.neuropharm.2022.109208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
Clinically, juveniles are more sensitive to stress than adults, and exposure to stress as juveniles prolongs psychiatric symptoms and causes treatment resistance. However, the efficacy of antidepressants for juveniles with psychiatric disorders is unknown. In the present study, we investigated whether the expression or development of impaired social behavior was attenuated by memantine, a NMDA receptor antagonist. In addition, we clarified the molecular mechanisms related to intracellular signal transduction through NMDA receptors and the ameliorating effect of memantine in mice with impaired social behavior. Acute administration of memantine before the social interaction test, but not before exposure to social defeat stress, attenuated social behavioral impairment. A single social defeat stress increased the phosphorylation of NMDA receptor subunit GluN2A and extracellular-signal-related kinase 1/2 (ERK1/2). Memantine inhibited the increase of phosphorylated GluN2A and ERK1/2 resulting from social interaction behavior. In both GluN2A deficient and pharmacological blockaded mice, social behavioral impairment was not observed in the social interaction test through regulation of ERK1/2 phosphorylation. These findings suggest that memantine ameliorates social behavioral impairment in mice exposed to a single social defeat stress as juveniles by regulating the NMDA receptor and subsequent ERK1/2 signaling activation. Memantine may constitute a novel therapeutic drug for stress-related psychiatric disorders in juveniles with adverse juvenile experiences.
Collapse
Affiliation(s)
- Mikio Yoshida
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Sho Hasegawa
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Masayuki Taniguchi
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Akihiro Mouri
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Chiharu Suzuki
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Akira Yoshimi
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan
| | - Takayoshi Mamiya
- Department of Chemical Pharmacology, Graduate School of Pharmacy, Meijo University, Nagoya, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Noda
- Division of Clinical Sciences and Neuropsychopharmacology, Meijo University Faculty and Graduate School of Pharmacy, Nagoya, Japan.
| |
Collapse
|
20
|
Nicotinic receptors promote susceptibility to social stress in female mice linked with neuroadaptations within VTA dopamine neurons. Neuropsychopharmacology 2022; 47:1587-1596. [PMID: 35459925 PMCID: PMC9283477 DOI: 10.1038/s41386-022-01314-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
Abstract
There are about twice as many women as men who experience depression during their lifetime. Although life circumstances and especially exposure to stressful situations constitute a major risk factor to develop depression, the underlying mechanisms have yet to be unraveled. We employed the chronic social defeat procedure to elicit depressive-like symptoms in females and ketamine to validate the model. We performed ex-vivo patch clamp recordings to assess cellular adaptations and used pharmacological agents to dissect these deregulations. Chronic social defeat exposure triggers a hyperactivity of VTA putative dopamine (DA) neurons in females susceptible to stress but not resilient ones. This hyperactivity was fully reversed by a single administration of ketamine. In virally-identified brain circuits of both susceptible and resilient females, we found a hypercholinergic tone to the VTA arising from the laterodorsal tegmentum. Application of puffs of nicotine revealed a decreased sensitivity of DA neurons in resilient mice when compared to naive or susceptible ones. The in vivo acute administration of the positive allosteric modulator for α7 nicotinic acetylcholine receptors (nAChRs) not only increased susceptibility to stress by enhancing activity of VTA DA neurons, but also triggered a switch in phenotype from resilient to susceptible. Our data unravel dysregulations of VTA DA neurons activity exclusively in females exhibiting depressive-like symptoms and identify VTA nAChRs as key molecular substrates that exacerbate susceptibility to stress.
Collapse
|
21
|
The effect of ketamine on anhedonia: improvements in dimensions of anticipatory, consummatory, and motivation-related reward deficits. Psychopharmacology (Berl) 2022; 239:2011-2039. [PMID: 35292831 DOI: 10.1007/s00213-022-06105-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 02/23/2022] [Indexed: 10/18/2022]
Abstract
Anhedonia is a common, persistent, and disabling condition. However, available therapeutics primarily focus on the reduction of depressive and negative symptoms rather than amelioration of deficits in positive affect. As such, extant drug treatments remain largely ineffective in treating symptoms of anhedonia. Ketamine is a rapid-acting and novel therapeutic treatment for treatment-resistant depression, which has also been demonstrated to attenuate symptoms of anhedonia. However, the literature on the anti-anhedonic effects of ketamine is limited-especially within independent dimensions of this symptom domain. Herein, this review examined the impact of ketamine treatment on anhedonia and its dimensions on anticipatory, consummatory, and motivation-related reward deficits. Overall, the findings have shown a trend towards symptom reduction and/or improvements in anhedonia and their respective subdomains, in both human and preclinical studies, as well as its potential to provide additional benefit in reducing suicidality and improving quality-of-life. Although further research is required in understanding the long-term efficacy and mechanism, ketamine may provide an effective and rapid-acting therapeutic in an otherwise unmet domain.
Collapse
|
22
|
Yu G, Cao F, Hou T, Cheng Y, Jia B, Yu L, Chen W, Xu Y, Chen M, Wang Y. Astrocyte reactivation in medial prefrontal cortex contributes to obesity-promoted depressive-like behaviors. J Neuroinflammation 2022; 19:166. [PMID: 35761401 PMCID: PMC9235218 DOI: 10.1186/s12974-022-02529-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 06/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Little is known about how the obesogenic environment influences emotional states associated with glial responses and neuronal function. Here, we investigated glial reactivation and neuronal electrophysiological properties in emotion-related brain regions of high-fat diet (HFD) and ob/ob mice under chronic stress. METHODS The glial reactivation and neuronal activities in emotion-related brain regions were analyzed among normal diet mice (ND), HFD mice, wild-type mice, and ob/ob mice. To further activate or inhibit astrocytes in medial prefrontal cortex (mPFC), we injected astrocytes specific Gq-AAV or Gi-AAV into mPFC and ongoing treated mice with CNO. RESULTS The results showed that obesogenic factors per se had no significant effect on neuronal activities in emotion-related brain regions, or on behavioral performance. However, exposure to a chronic stressor profoundly reduced the frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) and spontaneous excitatory postsynaptic currents (sEPSCs) in the mPFC; depressive-like behaviors were seen, accompanied by significant upregulation of astrocyte reactivation. We identified resilient and susceptible mice among chronic social defeat stress-exposed HFD mice. As expected, astrocyte reactivity was upregulated, while neuronal activity was depressed, in the mPFC of susceptible compared to resilient mice. Furthermore, activating astrocytes resulted in similar levels of neuronal activity and depressive-like behaviors between resilient and susceptible mice. Additionally, inhibiting astrocyte reactivation in the mPFC of HFD mice upregulated neuronal activities and inhibited depressive-like behaviors. CONCLUSIONS These observations indicate that obesogenic factors increase the risk of depression, and improve our understanding of the pathological relationship between obesity and depression.
Collapse
Affiliation(s)
- Gang Yu
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Feng Cao
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Tingting Hou
- College of Pharmacy, Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China.,Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Yunsheng Cheng
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Benli Jia
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Liang Yu
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Wanjing Chen
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yanyan Xu
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China.,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Mingming Chen
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China. .,Department of Neurology, Yale University School of Medicine, New Haven, 06536, USA.
| | - Yong Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, China. .,Bariatric Center, the Second Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
23
|
Effects of stress on endophenotypes of suicide across species: A role for ketamine in risk mitigation. Neurobiol Stress 2022; 18:100450. [PMID: 35685678 PMCID: PMC9170747 DOI: 10.1016/j.ynstr.2022.100450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
Suicide is a leading cause of death and morbidity worldwide, yet few interventions are available to mitigate its risk. Barriers to effective treatments involve a limited understanding of factors that predict the onset of suicidal thoughts and behaviors. In the context of suicide risk, stress is a precipitating factor that is largely overlooked in the literature. Indeed, the pathophysiology of stress and suicide are heavily interconnected, underscoring the need to target the stress system in suicide prevention. In this review, we integrate findings from the preclinical and clinical literature that links stress and suicide. We focus specifically on the effects of stress on underlying biological functions and processes associated with suicide, allowing for the review of research using animal models. Owing to the rapid anti-suicidal effects of (R,S)-ketamine, we discuss its ability to modulate various stress-related endophenotypes of suicide, as well as its potential role in preventing suicide in those with a history of chronic life stress (e.g., early life adversity). We highlight future research directions that could advance our understanding of stress-related effects on suicide risk, advocating a dimensional, endophenotype approach to suicide research. Suicide and chronic stress pathophysiology are interconnected. Chronic stress has profound impacts on several endophenotypes of suicide. Animal and human research points to stress as a precipitating factor in suicide. Ketamine modulates specific biological processes associated with stress and suicide. Suicide research into endophenotypes can help inform risk-mitigation strategies.
Collapse
|
24
|
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: 20] [Impact Index Per Article: 10.0] [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
|
25
|
Williams ES, Mazei-Robison M, Robison AJ. Sex Differences in Major Depressive Disorder (MDD) and Preclinical Animal Models for the Study of Depression. Cold Spring Harb Perspect Biol 2022; 14:a039198. [PMID: 34404738 PMCID: PMC8886985 DOI: 10.1101/cshperspect.a039198] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Depression and related mood disorders constitute an enormous burden on health, quality of life, and the global economy, and women have roughly twice the lifetime risk of men for experiencing depression. Here, we review sex differences in human brain physiology that may be connected to the increased susceptibility of women to major depressive disorder (MDD). Moreover, we summarize decades of preclinical research using animal models for the study of mood dysfunction that uncover some of the potential molecular, cellular, and circuit-level mechanisms that may underlie sex differences and disease etiology. We place particular emphasis on a series of recent studies demonstrating the central contribution of the circuit projecting from ventral hippocampus to nucleus accumbens and how inherent sex differences in the excitability of this circuit may predict and drive depression-related behaviors. The findings covered in this review underscore the continued need for studies using preclinical models and circuit-specific strategies for uncovering molecular and physiological mechanisms that could lead to potential sex-specific diagnosis, prognosis, prevention, and/or treatments for MDD and other mood disorders.
Collapse
Affiliation(s)
- Elizabeth S Williams
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | | - A J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
26
|
Modifying the maternal microbiota alters the gut-brain metabolome and prevents emotional dysfunction in the adult offspring of obese dams. Proc Natl Acad Sci U S A 2022; 119:2108581119. [PMID: 35197280 PMCID: PMC8892342 DOI: 10.1073/pnas.2108581119] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity disturbs brain-gut-microbiota interactions and induces negative affect in the offspring, but its impact on gut and brain metabolism in the offspring (F1) are unknown. Here, we tested whether perinatal intake of a multispecies probiotic could mitigate the abnormal emotional behavior in the juvenile and adult offspring of obese dams. Untargeted NMR-based metabolomic profiling and gene-expression analysis throughout the gut-brain axis were then used to investigate the biology underpinning behavioral changes in the dams and their offspring. Prolonged high-fat diet feeding reduced maternal gut short-chain fatty acid abundance, increased markers of peripheral inflammation, and decreased the abundance of neuroactive metabolites in maternal milk during nursing. Both juvenile (postnatal day [PND] 21) and adult (PND112) offspring of obese dams exhibited increased anxiety-like behavior, which were prevented by perinatal probiotic exposure. Maternal probiotic treatment increased gut butyrate and brain lactate in the juvenile and adult offspring and increased the expression of prefrontal cortex PFKFB3, a marker of glycolytic metabolism in astrocytes. PFKFB3 expression correlated with the increase in gut butyrate in the juvenile and adult offspring. Maternal obesity reduced synaptophysin expression in the adult offspring, while perinatal probiotic exposure increased expression of brain-derived neurotrophic factor. Finally, we showed that the resilience of juvenile and adult offspring to anxiety-like behavior was most prominently associated with increased brain lactate abundance, independent of maternal group. Taken together, we show that maternal probiotic supplementation exerts a long-lasting effect on offspring neuroplasticity and the offspring gut-liver-brain metabolome, increasing resilience to emotional dysfunction induced by maternal obesity.
Collapse
|
27
|
Huang SH, Liu WZ, Qin X, Guo CY, Xiong QC, Wang Y, Hu P, Pan BX, Zhang WH. Association of Increased Amygdala Activity with Stress-Induced Anxiety but not Social Avoidance Behavior in Mice. Neurosci Bull 2022; 38:16-28. [PMID: 34494228 PMCID: PMC8782949 DOI: 10.1007/s12264-021-00762-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/16/2021] [Indexed: 01/03/2023] Open
Abstract
Chronic stress leads to many psychiatric disorders, including social and anxiety disorders that are associated with over-activation of neurons in the basolateral amygdala (BLA). However, not all individuals develop psychiatric diseases, many showing considerable resilience against stress exposure. Whether BLA neuronal activity is involved in regulating an individual's vulnerability to stress remains elusive. In this study, using a mouse model of chronic social defeat stress (CSDS), we divided the mice into susceptible and resilient subgroups based on their social interaction behavior. Using in vivo fiber photometry and in vitro patch-clamp recording, we showed that CSDS persistently (after 20 days of recovery from stress) increased BLA neuronal activity in all the mice regardless of their susceptible or resilient nature, although impaired social interaction behavior was only observed in susceptible mice. Increased anxiety-like behavior, on the other hand, was evident in both groups. Notably, the CSDS-induced increase of BLA neuronal activity correlated well with the heightened anxiety-like but not the social avoidance behavior in mice. These findings provide new insight to our understanding of the role of neuronal activity in the amygdala in mediating stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Shou-He Huang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Wei-Zhu Liu
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xia Qin
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Chen-Yi Guo
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Qing-Cheng Xiong
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Yu Wang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China
| | - Ping Hu
- Institute of Translational Medicine, Nanchang University, Nanchang, 330001, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Wen-Hua Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
28
|
Benham RS, Choi C, Hodgson NW, Hewage NB, Kastli R, Donahue RJ, Muschamp JW, Engin E, Carlezon WA, Hensch TK, Rudolph U. α2-containing γ-aminobutyric acid type A receptors promote stress resiliency in male mice. Neuropsychopharmacology 2021; 46:2197-2206. [PMID: 34408277 PMCID: PMC8505491 DOI: 10.1038/s41386-021-01144-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/11/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
Brain α2-containing GABAA receptors play a critical role in the modulation of anxiety- and fear-like behavior. However, it is unknown whether these receptors also play a role in modulating resilience to chronic stress, and in which brain areas and cell types such an effect would be mediated. We evaluated the role of α2-containing GABAA receptors following chronic social defeat stress using male mice deficient in the α2 subunit globally or conditionally in dopamine D1- or D2-receptor-expressing neurons, e.g., within the nucleus accumbens (NAc). In addition, we examined the effect of the lack of the α2 subunit on intermediates of the glutathione synthesis pathway. We found that α2-containing GABAA receptors on D2-receptor-positive but not on D1-receptor-positive neurons promote resiliency to chronic social defeat stress, as reflected in social interaction tests. The pro-resiliency effects of α2-containing GABAA receptors on D2-receptor-positive neurons do not appear to be directly related to alterations in anxiety-like behavior, as reflected in the elevated plus-maze, light-dark box, and novel open field tests. Increases in indices of oxidative stress-reflected by increases in cystathionine levels and reductions in GSH/GSSG ratios-were found in the NAc and prefrontal cortex but not in the hippocampus of mice lacking α2-containing GABAA receptors. We conclude that α2-containing GABAA receptors within specific brain areas and cell populations promote stress resiliency independently of direct effects on anxiety-like behaviors. A potential mechanism contributing to this increased resiliency is the protection that α2-containing GABAA receptors provide against oxidative stress in NAc and the prefrontal cortex.
Collapse
Affiliation(s)
- Rebecca S Benham
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Catherine Choi
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Nathaniel W Hodgson
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Nishani B Hewage
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rahel Kastli
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Rachel J Donahue
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Laboratory of Behavioral Genetics, McLean Hospital, Belmont, MA, USA
| | - John W Muschamp
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Laboratory of Behavioral Genetics, McLean Hospital, Belmont, MA, USA
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Elif Engin
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Laboratory of Behavioral Genetics, McLean Hospital, Belmont, MA, USA
| | - Takao K Hensch
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
29
|
Lopez J, Bagot RC. Defining Valid Chronic Stress Models for Depression With Female Rodents. Biol Psychiatry 2021; 90:226-235. [PMID: 33965195 DOI: 10.1016/j.biopsych.2021.03.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
Women are twice as likely to experience depression than men, yet until recently, preclinical studies in rodents have focused almost exclusively on males. As interest in sex differences and sex-specific mechanisms of stress susceptibility increases, chronic stress models for inducing depression-relevant behavioral and physiological changes in male rodents are being applied to females, and several new models have emerged to include both males and females, yet not all models have been systematically validated in females. An increasing number of researchers seek to include female rodents in their experimental designs, asking the question "what is the ideal chronic stress model for depression in females?" We review criteria for assessing female model validity in light of key research questions and the fundamental distinction between studying sex differences and studying both sexes. In overviewing current models, we explore challenges inherent to establishing an ideal female chronic stress model, with particular emphasis on the need for standardization and adoption of validated behavioral tests sensitive to stress effects in females. Taken together, these considerations will empower female chronic stress models to provide a better understanding of stress susceptibility and allow the development of efficient sex-specific treatments.
Collapse
Affiliation(s)
- Joëlle Lopez
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montréal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Montréal, Quebec, Canada.
| |
Collapse
|
30
|
Lee CW, Fang YP, Chu MC, Chung YJ, Chi H, Tang CW, So EC, Lin HC, Lin HC. Differential mechanisms of synaptic plasticity for susceptibility and resilience to chronic social defeat stress in male mice. Biochem Biophys Res Commun 2021; 562:112-118. [PMID: 34049204 DOI: 10.1016/j.bbrc.2021.05.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
Mood dysregulation refers to the inability of a person to control their negative emotions, and it is linked to various stressful experiences. Dysregulated neural synaptic plasticity and actin-filament dynamics are important regulators of stress response in animal models. However, until now, there is no evidence to differential the mechanisms of synaptic plasticity and actin-filament dynamics in stress susceptibility and stress-resistant. Here we found that depression-like behaviour was observed in the susceptible group following chronic social defeat stress (CSDS) exposure, but not in stress-resistant mice. High-frequency stimulation-induced long-term potentiation (LTP) was impaired in the CSDS-induced depression-susceptible group. Further, the levels of pro-brain derived neurotrophic factor (BDNF), mature BDNF, PSD-95, phosphorylated CaMKII, and phosphorylated Cofilin, an actin-filament dynamics regulator, were reduced in CSDS-induced depression-susceptible mice unlike in stress-resistant mice. These results demonstrate that synaptic plasticity-related molecules, such as BDNF and phosphorylated Cofilin, are important for maintaining synaptic functions and structure in mice that experience more stress.
Collapse
Affiliation(s)
- Chi-Wei Lee
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, Taiwan
| | - Yen-Po Fang
- Department of Orthopaedic Surgery, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Ming-Chia Chu
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yueh-Jung Chung
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsiang Chi
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Wei Tang
- Department of Neurology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Edmund Cheung So
- Department of Anesthesia and Medical Research, An Nan Hospital, China Medical University, Tainan City, Taiwan; Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan City, Taiwan
| | - Hsin-Chuan Lin
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hui-Ching Lin
- Department and Institute of Physiology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institute, Taipei, Taiwan; Brain Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
31
|
McCullough KM, Missig G, Robbie MA, Foilb AR, Wells AM, Hartmann J, Anderson KJ, Neve RL, Nestler EJ, Ressler KJ, Carlezon WA. Nucleus Accumbens Medium Spiny Neuron Subtypes Differentially Regulate Stress-Associated Alterations in Sleep Architecture. Biol Psychiatry 2021; 89:1138-1149. [PMID: 33715826 PMCID: PMC8178228 DOI: 10.1016/j.biopsych.2020.12.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/17/2020] [Accepted: 12/20/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Stress is implicated in the pathophysiology of major depression and posttraumatic stress disorder. These conditions share core features, including motivational deficits, heighted anxiety, and sleep dysregulation. Chronic stress produces these same features in rodents, with some individuals being susceptible or resilient, as seen in humans. While stress-induced neuroadaptations within the nucleus accumbens are implicated in susceptibility-related dysregulation of motivational and emotional behaviors, their effects on sleep are unclear. METHODS We used chemogenetics (DREADDs [designer receptors exclusively activated by designer drugs]) to examine the effects of selective alterations in activity of nucleus accumbens medium spiny neurons expressing dopamine D1 receptors (D1-MSNs) or dopamine D2 receptors (D2-MSNs) on sleep-related end points. Mice were implanted with wireless transmitters enabling continuous collection of data to quantify vigilance states over a 20-day test period. Parallel cohorts were examined in behavioral tests assessing stress susceptibility. RESULTS D1- and D2-MSNs play dissociable roles in sleep regulation. Stimulation of inhibitory or excitatory DREADDs expressed in D1-MSNs exclusively affects rapid eye movement sleep, whereas targeting D2-MSNs affects slow wave sleep. The combined effects of D1-MSN inhibition and D2-MSN activation on sleep resemble those of chronic social defeat stress. Alterations in D1-MSN function also affect stress susceptibility in social behavior tests. Elevation of CREB (cAMP response element-binding protein) within D1-MSNs is sufficient to produce stress-like effects on rapid eye movement sleep. CONCLUSIONS In addition to regulation of motivational and emotional behaviors, the nucleus accumbens also influences sleep, an end point with high translational relevance. These findings provide a neural basis for comorbidity in key features of stress-related illness.
Collapse
Affiliation(s)
- Kenneth M. McCullough
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Galen Missig
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Mykel A. Robbie
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Allison R. Foilb
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Audrey M. Wells
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Jakob Hartmann
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Kasey J. Anderson
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Rachael L. Neve
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eric J. Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai. New York, NY 10029, USA
| | - Kerry J. Ressler
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - William A. Carlezon
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Corresponding Author: William A. Carlezon, Jr., Ph.D., Department of Psychiatry, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA,
| |
Collapse
|
32
|
Life-course effects of early life adversity exposure on eating behavior and metabolism. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 97:237-273. [PMID: 34311901 DOI: 10.1016/bs.afnr.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Environmental variations in early life influence brain development, making individuals more vulnerable to psychiatric and metabolic disorders. Early life stress (ELS) has a strong impact on the development of eating behavior. However, eating is a complex behavior, determined by an interaction between signals of energy homeostasis, neuronal circuits involved in its regulation, and circuits related to rewarding properties of the food. Although mechanisms underlying ELS-induced altered feeding behavior are not completely understood, evidence suggest that the effects of ELS on metabolic, mood, and emotional disorders, as well as reward system dysfunctions can contribute directly or indirectly to altered feeding behavior. The focus of this chapter is to discuss the effects of ELS on eating behavior and metabolism, considering different factors that control appetite such as energy homeostasis, hedonic properties of the food, emotional and cognitive status. After highlighting classic studies on the association between ELS and eating behavior alterations, we discuss how exposure to adversity can interact with genetics characteristics to predict variable outcomes.
Collapse
|
33
|
Li HJ, Su X, Zhang LW, Zhang CY, Wang L, Li WQ, Yang YF, Lv LX, Li M, Xiao X. Transcriptomic analyses of humans and mice provide insights into depression. Zool Res 2021; 41:632-643. [PMID: 32987454 PMCID: PMC7671914 DOI: 10.24272/j.issn.2095-8137.2020.174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accumulating studies have been conducted to identify risk genes and relevant biological mechanisms underlying major depressive disorder (MDD). In particular, transcriptomic analyses in brain regions engaged in cognitive and emotional processes, e.g., the dorsolateral prefrontal cortex (DLPFC), have provided essential insights. Based on three independent DLPFC RNA-seq datasets of 79 MDD patients and 75 healthy controls, we performed differential expression analyses using two alternative approaches for cross-validation. We also conducted transcriptomic analyses in mice undergoing chronic variable stress (CVS) and chronic social defeat stress (CSDS). We identified 12 differentially expressed genes (DEGs) through both analytical methods in MDD patients, the majority of which were also dysregulated in stressed mice. Notably, the mRNA level of the immediate early gene FOS ( Fos proto-oncogene) was significantly decreased in both MDD patients and CVS-exposed mice, and CSDS-susceptible mice exhibited a greater reduction in Fos expression compared to resilient mice. These findings suggest the potential key roles of this gene in the pathogenesis of MDD related to stress exposure. Altered transcriptomes in the DLPFC of MDD patients might be, at least partially, the result of stress exposure, supporting that stress is a primary risk factor for MDD.
Collapse
Affiliation(s)
- Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xi Su
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Lu-Wen Zhang
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Wen-Qiang Li
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Yong-Feng Yang
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Lu-Xian Lv
- Henan Mental Hospital, Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Key Lab of Biological Psychiatry, International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang Medical University, Xinxiang, Henan 453002, China.,Henan Province People's Hospital, Zhengzhou, Henan 450003, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
34
|
Liao C, Kwan AC. Applying Reinforcement Learning to Rodent Stress Research. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2021; 5:2470547020984732. [PMID: 33598593 PMCID: PMC7863143 DOI: 10.1177/2470547020984732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 11/16/2022]
Abstract
Rodent models are an invaluable tool for studying the pathophysiological mechanisms underlying stress and depressive disorders. However, the widely used behavioral assays to measure depressive-like states in rodents have serious limitations. In this commentary, we suggest that learning tasks, particularly those that can be analyzed with the framework of reinforcement learning, are ideal for assaying reward processing deficits relevant to depression. The key advantages of these tasks are their repeatable, quantifiable nature and the link to clinical studies. By optimizing the behavioral readout of stress-induced phenotypes in rodents, a reinforcement learning-based approach may help bridge the translational gap and advance antidepressant discovery.
Collapse
Affiliation(s)
- Clara Liao
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Alex C. Kwan
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
35
|
Fonseca R, Madeira N, Simoes C. Resilience to fear: The role of individual factors in amygdala response to stressors. Mol Cell Neurosci 2020; 110:103582. [PMID: 33346000 DOI: 10.1016/j.mcn.2020.103582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 11/13/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022] Open
Abstract
Resilience to stress is an adaptive process that varies individually. Resilience refers to the adaptation, or the ability to maintain or regain mental health, despite being subject to adverse situation. Resilience is a dynamic concept that reflects a combination of internal individual factors, including age and gender interacting with external factors such as social, cultural and environmental factors. In the last decade, we have witnessed an increase in the prevalence of anxiety disorders, including post-traumatic stress disorder. Given that stress in unavoidable, it is of great interest to understand the neurophysiological mechanisms of resilience, the individual factors that may contribute to susceptibility and promote efficacious approaches to improve resilience. Here, we address this complex question, attempting at defining clear and operational definitions that may allow us to improve our analysis of behavior incorporating individuality. We examine how individual perception of the stressor can alter the outcome of an adverse situation using as an example, the fear-conditioning paradigm and discuss how individual differences in the reward system can contribute to resilience. Given the central role of the endocannabinoid system in regulating fear responses and anxiety, we discuss the evidence that polymorphisms in several molecules of this signaling system contribute to different anxiety phenotypes. The endocannabinoid system is highly interconnected with the serotoninergic and dopaminergic modulatory systems, contributing to individual differences in stress perception and coping mechanisms. We review how the individual variability in these modulatory systems can be used towards a multivariable assessment of stress risk. Incorporating individuality in our research will allow us to define biomarkers of anxiety disorders as well as assess prognosis, towards a personalized clinical approach to mental health.
Collapse
Affiliation(s)
- Rosalina Fonseca
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal.
| | - Natália Madeira
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| | - Carla Simoes
- Cellular and Systems Neurobiology, Chronic Diseases Research Center (CEDOC), NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 130 1169-056 Lisboa, Portugal
| |
Collapse
|
36
|
Calarco CA, Lobo MK. Depression and substance use disorders: Clinical comorbidity and shared neurobiology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:245-309. [PMID: 33648671 DOI: 10.1016/bs.irn.2020.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mood disorders, including major depressive disorder (MDD), are the most prevalent psychiatric illnesses, and pose an incredible burden to society, both in terms of disability and in terms of costs associated with medical care and lost work time. MDD has extremely high rates of comorbidity with substance use disorders (SUD) as many of the same neurobiological circuits and molecular mechanisms regulate the reward pathways disrupted in both conditions. MDD may induce SUDs, SUD may contribute to MDD development, or underlying vulnerabilities and common life experience may confer risk to developing both conditions. In this chapter we explore theories of MDD and SUD comorbidity, the neurobiological underpinnings of depression, overlapping cellular and molecular pathways for both conditions, and current treatment approaches for these comorbid conditions.
Collapse
Affiliation(s)
- Cali A Calarco
- 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.
| |
Collapse
|
37
|
Hamilton PJ, Chen EY, Tolstikov V, Peña CJ, Picone JA, Shah P, Panagopoulos K, Strat AN, Walker DM, Lorsch ZS, Robinson HL, Mervosh NL, Kiraly DD, Sarangarajan R, Narain NR, Kiebish MA, Nestler EJ. Chronic stress and antidepressant treatment alter purine metabolism and beta oxidation within mouse brain and serum. Sci Rep 2020; 10:18134. [PMID: 33093530 PMCID: PMC7582177 DOI: 10.1038/s41598-020-75114-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Major depressive disorder (MDD) is a complex condition with unclear pathophysiology. Molecular disruptions within limbic brain regions and the periphery contribute to depression symptomatology and a more complete understanding the diversity of molecular changes that occur in these tissues may guide the development of more efficacious antidepressant treatments. Here, we utilized a mouse chronic social stress model for the study of MDD and performed metabolomic, lipidomic, and proteomic profiling on serum plus several brain regions (ventral hippocampus, nucleus accumbens, and medial prefrontal cortex) of susceptible, resilient, and unstressed control mice. To identify how commonly used tricyclic antidepressants impact the molecular composition in these tissues, we treated stress-exposed mice with imipramine and repeated our multi-OMIC analyses. Proteomic analysis identified three serum proteins reduced in susceptible animals; lipidomic analysis detected differences in lipid species between resilient and susceptible animals in serum and brain; and metabolomic analysis revealed dysfunction of purine metabolism, beta oxidation, and antioxidants, which were differentially associated with stress susceptibility vs resilience by brain region. Antidepressant treatment ameliorated stress-induced behavioral abnormalities and affected key metabolites within outlined networks, most dramatically in the ventral hippocampus. This work presents a resource for chronic social stress-induced, tissue-specific changes in proteins, lipids, and metabolites and illuminates how molecular dysfunctions contribute to individual differences in stress sensitivity.
Collapse
Affiliation(s)
- Peter J Hamilton
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA. .,Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| | - Emily Y Chen
- BERG LLC, 500 Old Connecticut Path, Framingham, MA, 01701, USA
| | | | - Catherine J Peña
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | - Joseph A Picone
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Punit Shah
- BERG LLC, 500 Old Connecticut Path, Framingham, MA, 01701, USA
| | | | - Ana N Strat
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | - Deena M Walker
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | - Zachary S Lorsch
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | - Hannah L Robinson
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Nicholas L Mervosh
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| | | | - Niven R Narain
- BERG LLC, 500 Old Connecticut Path, Framingham, MA, 01701, USA
| | | | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, 10029, USA
| |
Collapse
|
38
|
Barthas F, Hu MY, Siniscalchi MJ, Ali F, Mineur YS, Picciotto MR, Kwan AC. Cumulative Effects of Social Stress on Reward-Guided Actions and Prefrontal Cortical Activity. Biol Psychiatry 2020; 88:541-553. [PMID: 32276717 PMCID: PMC7434704 DOI: 10.1016/j.biopsych.2020.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/27/2020] [Accepted: 02/09/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND When exposed to chronic social stress, animals display behavioral changes that are relevant to depressive-like phenotypes. However, the cascading relationship between incremental stress exposure and neural dysfunctions over time remains incompletely understood. METHODS We characterized the longitudinal effect of social defeat on goal-directed actions and prefrontal cortical activity in mice using a novel head-fixed sucrose preference task and two-photon calcium imaging. RESULTS Behaviorally, stress-induced loss of reward sensitivity intensifies over days. Motivational anhedonia, the failure to translate positive reinforcements into future actions, requires multiple sessions of stress exposure to become fully established. For neural activity, individual layer 2/3 pyramidal neurons in the cingulate and medial secondary motor subregions of the medial prefrontal cortex have heterogeneous responses to stress. Changes in ensemble activity differ significantly between susceptible and resilient mice after the first defeat session and continue to diverge following successive stress episodes before reaching persistent abnormal levels. CONCLUSIONS Collectively, these results demonstrate that the cumulative impact of an ethologically relevant stress can be observed at the level of cellular activity of individual prefrontal neurons. The distinct neural responses associated with resilience versus susceptibility suggests the hypothesis that the negative impact of social stress is neutralized in resilient animals, in part through an adaptive reorganization of prefrontal cortical activity.
Collapse
Affiliation(s)
- Florent Barthas
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Melody Y. Hu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Michael J. Siniscalchi
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA,Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Farhan Ali
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Yann S. Mineur
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Alex C. Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA,Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
39
|
Choi KH, Berman RY, Zhang M, Spencer HF, Radford KD. Effects of Ketamine on Rodent Fear Memory. Int J Mol Sci 2020; 21:ijms21197173. [PMID: 32998470 PMCID: PMC7582895 DOI: 10.3390/ijms21197173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 11/25/2022] Open
Abstract
Ketamine, a multimodal anesthetic drug, has become increasingly popular in the treatment of pain following traumatic injury as well as treatment-resistant major depressive disorders. However, the psychological impact of this dissociative medication on the development of stress-related disorders such as post-traumatic stress disorder (PTSD) remains controversial. To address these concerns, preclinical studies have investigated the effects of ketamine administration on fear memory and stress-related behaviors in laboratory animals. Despite a well-documented line of research examining the effects of ketamine on fear memory, there is a lack of literature reviews on this important topic. Therefore, this review article summarizes the current preclinical literature on ketamine and fear memory with a particular emphasis on the route, dose, and timing of ketamine administration in rodent fear conditioning studies. Additionally, this review describes the molecular mechanisms by which ketamine may impact fear memory and stress-related behaviors. Overall, findings from previous studies are inconsistent in that fear memory may be increased, decreased, or unaltered following ketamine administration in rodents. These conflicting results can be explained by factors such as the route, dose, and timing of ketamine administration; the interaction between ketamine and stress; and individual variability in the rodent response to ketamine. This review also recommends that future preclinical studies utilize a clinically relevant route of administration and account for biological sex differences to improve translation between preclinical and clinical investigations.
Collapse
Affiliation(s)
- Kwang H. Choi
- Department of Psychiatry, Uniformed Services University, Bethesda, MD 20814, USA; (K.H.C.); (R.Y.B.); (M.Z.)
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA
| | - Rina Y. Berman
- Department of Psychiatry, Uniformed Services University, Bethesda, MD 20814, USA; (K.H.C.); (R.Y.B.); (M.Z.)
| | - Michael Zhang
- Department of Psychiatry, Uniformed Services University, Bethesda, MD 20814, USA; (K.H.C.); (R.Y.B.); (M.Z.)
- Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD 20814, USA
| | - Haley F. Spencer
- Program in Neuroscience, Uniformed Services University, Bethesda, MD 20814, USA;
| | - Kennett D. Radford
- Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA
- Correspondence:
| |
Collapse
|
40
|
Słupski J, Cubała WJ, Górska N, Słupska A, Gałuszko-Węgielnik M. Copper and anti-anhedonic effect of ketamine in treatment-resistant depression. Med Hypotheses 2020; 144:110268. [PMID: 33254572 DOI: 10.1016/j.mehy.2020.110268] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 12/28/2022]
Abstract
Treatment-resistant depression (TRD) remains one of the major psychiatric challenges worldwide. Search for non-monoaminergic agents, possibly effective in treatment of TRD is of prime importance. One of those agents is ketamine - a dissociative, anesthetic drug exhibiting a fast antidepressant action in patients with TRD. Concurrently, anhedonia appears to be significant symptom domain with far-reaching impact on course of treatment. There is data demonstrating that abnormal copper levels might be associated with symptoms of depression. As there is common denominator in ketamine and copper role in neurotransmission this paper is to explore the associations of blood copper levels and psychometric measures in patients with TRD in course of major depressive disorder (MDD) and bipolar disorder (BP), focusing on anhedonia measured with Snaith-Hamilton Pleasure Scale (SHAPS) score.
Collapse
Affiliation(s)
- Jakub Słupski
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Wiesław Jerzy Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Górska
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Anita Słupska
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
41
|
|
42
|
Gold MS, Baron D, Bowirrat A, Blum K. Neurological correlates of brain reward circuitry linked to opioid use disorder (OUD): Do homo sapiens acquire or have a reward deficiency syndrome? J Neurol Sci 2020; 418:117137. [PMID: 32957037 PMCID: PMC7490287 DOI: 10.1016/j.jns.2020.117137] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/19/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The extant literature confirms that an array of polymorphic genes related to- neurotransmitters and second messengers govern the net release of dopamine in the Nucleus Accumbens (NAc) in the mesolimbic region of the brain. They are linked predominantly to motivation, anti-stress, incentive salience (wanting), and wellbeing. Notably, in 2000 the Nobel Prize was awarded to Carlsson, Greengard, and Kandel for their work on the molecular and cellular function of dopaminergic activity at neurons. This historical psychopharmacological work involved neurotransmission of serotonin, endorphins, glutamate, and dopamine, and the seminal work of Blum, Gold, Volkow, Nestler, and others related to neurotransmitter function and related behaviors. Currently, Americans are facing their second and worst opioid epidemic, prescribed opioids, and easy access drive this epidemic of overdoses, and opioid use disorders (OUDs). Presently the clinical consensus is to treat OUD, as if it were an opioid deficiency syndrome, with long-term to life-long opioid substitution therapy. Opioid agonist administration is seen as necessary to replace missing opioids, treat OUD, and prevent overdoses, like insulin is used to treat diabetes. Treatment of OUD and addiction, in general, is similar to the endocrinopathy conceptualization in that it views opioid agonist MATs as an essential core to therapy. Is this approach logical? Other than as harm reduction, is using opioids to treat OUD therapeutic or harmful in the long term? This historical Trieste provides a molecular framework to understand the current underpinnings of endorphinergic/dopaminergic mechanisms related to opioid deficiency syndrome and generalized reward processing depletion. WC 249.
Collapse
Affiliation(s)
- Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.
| | - David Baron
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| | - Abdalla Bowirrat
- Department of Neuroscience and Genetics, Interdisciplinary Center Herzliya, Israel
| | - Kenneth Blum
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| |
Collapse
|
43
|
Warren BL, Mazei-Robison MS, Robison AJ, Iñiguez SD. Can I Get a Witness? Using Vicarious Defeat Stress to Study Mood-Related Illnesses in Traditionally Understudied Populations. Biol Psychiatry 2020; 88:381-391. [PMID: 32228871 PMCID: PMC7725411 DOI: 10.1016/j.biopsych.2020.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 12/17/2022]
Abstract
The chronic social defeat stress model has been instrumental in shaping our understanding of neurobiology relevant to affect-related illnesses, including major depressive disorder. However, the classic chronic social defeat stress procedure is limited by its exclusive application to adult male rodents. We have recently developed a novel vicarious social defeat stress procedure wherein one mouse witnesses the physical defeat bout of a conspecific from the safety of an adjacent compartment. This witness mouse develops a similar behavioral phenotype to that of the mouse that physically experiences social defeat stress, modeling multiple aspects of major depressive disorder. Importantly, this new procedure allows researchers to perform vicarious social defeat stress in males or females and in juvenile mice, which typically are excluded from classic social defeat experiments. Here we discuss several recent advances made using this procedure and how its application provides a new preclinical approach to study the neurobiology of psychological stress-induced phenotypes.
Collapse
Affiliation(s)
- Brandon L Warren
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | | | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Sergio D Iñiguez
- Department of Psychology, The University of Texas at El Paso, El Paso, Texas.
| |
Collapse
|
44
|
Garcia-Carachure I, Flores-Ramirez FJ, Castillo SA, Themann A, Arenivar MA, Preciado-Piña J, Zavala AR, Lobo MK, Iñiguez SD. Enduring effects of adolescent ketamine exposure on cocaine- and sucrose-induced reward in male and female C57BL/6 mice. Neuropsychopharmacology 2020; 45:1536-1544. [PMID: 32165718 PMCID: PMC7360558 DOI: 10.1038/s41386-020-0654-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022]
Abstract
Ketamine has shown promising antidepressant efficacy for adolescent treatment-resistant depression. However, the potential enduring consequences of ketamine exposure have not been thoroughly evaluated. Thus, we examined if juvenile ketamine treatment results in long-lasting changes for the rewarding properties of sucrose and cocaine in adulthood, across three separate experiments. In Experiment 1, adolescent male and female C57BL/6 mice received ketamine (20 mg/kg) for 15 consecutive days (Postnatal Day [PD] 35-49). Twenty-one days later (PD70; adulthood) we examined their behavioral responsivity to sucrose (1%) on a two-bottle choice design, or cocaine (0, 5, 10 mg/kg) using the conditioned place preference (CPP) test. We found that juvenile ketamine-pretreatment increased preference for sucrose and environments paired with cocaine in male, but not female, adult mice. This long-term outcome was not observed when male and female mice received ketamine as adults (PD70-84) and tested for sucrose and cocaine preference 21-days later (Experiment 2). Similarly, in Experiment 3, no long-lasting differences in these measures were observed when adolescent male mice were exposed to concomitant ketamine and social stressors (PD35-44), namely the social defeat or vicarious defeat stress paradigms-procedures that mediated a depression-related phenotype (along with a ketamine antidepressant-like response). Collectively, we demonstrate that in the absence of physical or psychological stress, adolescent ketamine exposure increases later life preference for the rewarding properties of sucrose and cocaine in a sex- and age-specific manner. As such, this preclinical work provides awareness for the potential long-term behavioral consequences associated with juvenile ketamine exposure.
Collapse
Affiliation(s)
- Israel Garcia-Carachure
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Francisco J. Flores-Ramirez
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Samuel A. Castillo
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Anapaula Themann
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Miguel A. Arenivar
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Joshua Preciado-Piña
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| | - Arturo R. Zavala
- 0000 0000 9093 6830grid.213902.bDepartment of Psychology, California State University, Long Beach, CA USA
| | - Mary Kay Lobo
- 0000 0001 2175 4264grid.411024.2Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD USA
| | - Sergio D. Iñiguez
- 0000 0001 0668 0420grid.267324.6Department of Psychology, The University of Texas at El Paso, El Paso, TX USA
| |
Collapse
|
45
|
Whittaker AL, Hickman DL. The Impact of Social and Behavioral Factors on Reproducibility in Terrestrial Vertebrate Models. ILAR J 2020; 60:252-269. [PMID: 32720675 DOI: 10.1093/ilar/ilaa005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The use of animal models remains critical in preclinical and translational research. The reliability of the animal models and aspects of their validity is likely key to effective translation of findings to medicine. However, despite considerable uniformity in animal models brought about by control of genetics, there remain a number of social as well as innate and acquired behavioral characteristics of laboratory animals that may impact on research outcomes. These include the effects of strain and genetics, age and development, sex, personality and affective states, and social factors largely brought about by housing and husbandry. In addition, aspects of the testing environment may also influence research findings. A number of considerations resulting from the animals' innate and acquired behavioral characteristics as well as their social structures are described. Suggestions for minimizing the impact of these factors on research are provided.
Collapse
Affiliation(s)
- Alexandra L Whittaker
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, South Australia, Australia
| | - Debra L Hickman
- Laboratory Animal Resource Center, Indiana University, Indianapolis, Indiana
| |
Collapse
|
46
|
Parker KE, Sugiarto E, Taylor AMW, Pradhan AA, Al-Hasani R. Pain, Motivation, Migraine, and the Microbiome: New Frontiers for Opioid Systems and Disease. Mol Pharmacol 2020; 98:433-444. [PMID: 32958571 DOI: 10.1124/mol.120.119438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
For decades the broad role of opioids in addiction, neuropsychiatric disorders, and pain states has been somewhat well established. However, in recent years, with the rise of technological advances, not only is the existing dogma being challenged, but we are identifying new disease areas in which opioids play a critical role. This review highlights four new areas of exploration in the opioid field. The most recent addition to the opioid family, the nociceptin receptor system, shows promise as the missing link in understanding the neurocircuitry of motivation. It is well known that activation of the kappa opioid receptor system modulates negative affect and dysphoria, but recent studies now implicate the kappa opioid system in the modulation of negative affect associated with pain. Opioids are critical in pain management; however, the often-forgotten delta opioid receptor system has been identified as a novel therapeutic target for headache disorders and migraine. Lastly, changes to the gut microbiome have been shown to directly contribute to many of the symptoms of chronic opioid use and opioid related behaviors. This review summarizes the findings from each of these areas with an emphasis on identifying new therapeutic targets. SIGNIFICANCE STATEMENT: The focus of this minireview is to highlight new disease areas or new aspects of disease in which opioids have been implicated; this includes pain, motivation, migraine, and the microbiome. In some cases, this has resulted in the pursuit of a novel therapeutic target and resultant clinical trial. We believe this is very timely and will be a refreshing take on reading about opioids and disease.
Collapse
Affiliation(s)
- Kyle E Parker
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Elizabeth Sugiarto
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Anna M W Taylor
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Amynah A Pradhan
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Ream Al-Hasani
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| |
Collapse
|
47
|
Han M, Luo H, Bai Y, Zheng S, Li F, Fu J, Jiang S, Liu Z, Zheng X. The effect of traumatic-like stress exposure on alterations in the temporal social behavior of a rodent population. Stress 2020; 23:393-404. [PMID: 31814486 DOI: 10.1080/10253890.2019.1702642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Though the relationship between traumatic stress and social behavior, which has been explored for years, is dynamic and largely estimated between dyads, little is known about the causal effects of traumatic stress exposure on the time-dependent dynamic alterations in the social behaviors on a large-group level. We thus investigated the effect of a single prolonged stress (SPS) exposure, a classical animal model that recapitulates posttraumatic stress disorder (PTSD)-like symptoms in rodents, on the spatiotemporal, social behavior changes within a large group of cohabiting rats. One-half of thirty-two Sprague-Dawley rats were assigned to the experimental group and subjected to SPS treatment administered two weeks after baseline social behavior recording; the other half served as the controls. Each group of rats (n = 16) was housed in one of two large custom-made cylinders. We used an automatic tracking system to record the behavioral indices of social behavior of the rats before SPS exposure, on the SPS exposure day, during a 7-day-long quiescent period after SPS treatment, as well as during subsequent behavioral test days. In addition to SPS-induced PTSD-like behaviors, SPS induced a time-dependent, oscillating change in active/passive social behaviors that lasted for 3 weeks. SPS treatment decreased active social behaviors (especially affiliative behaviors) but increased passive social behaviors (e.g. huddling) immediately following stress exposure. Increased active social interactions were observed during the early phase after SPS treatment; while increased passive social behaviors were observed during the late phase after SPS treatment. These dynamic changes were repeatedly observed when the rats underwent subsequent stressful behavioral tests and challenges. SPS induced a long-term, time-dependent oscillating change in indices of the social behavior. These changes may serve as an adaptive mechanism, and their manifestations critically depended on the time course following the traumatic stress exposure.
Collapse
Affiliation(s)
- Mengfei Han
- School of Psychology, Beijing Sport University, Beijing, China
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Haoshuang Luo
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yunjing Bai
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Shichun Zheng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fenghua Li
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Juan Fu
- College of Biological and Environmental Engineering, Binzhou University, Shandong, China
| | - Shaofei Jiang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhengkui Liu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xigeng Zheng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Kokane SS, Armant RJ, Bolaños-Guzmán CA, Perrotti LI. Overlap in the neural circuitry and molecular mechanisms underlying ketamine abuse and its use as an antidepressant. Behav Brain Res 2020; 384:112548. [PMID: 32061748 PMCID: PMC7469509 DOI: 10.1016/j.bbr.2020.112548] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/18/2020] [Accepted: 02/07/2020] [Indexed: 02/09/2023]
Abstract
Ketamine, a dissociative anesthetic and psychedelic compound, has revolutionized the field of psychopharmacology by showing robust, and rapid-acting antidepressant activity in patients suffering from major depressive disorder (MDD), suicidal tendencies, and treatment-resistant depression (TRD). Ketamine's efficacy, however, is transient, and patients must return to the clinic for repeated treatment as they experience relapse. This is cause for concern because ketamine is known for its abuse liability, and repeated exposure to drugs of abuse often leads to drug abuse/dependence. Though the mechanism(s) underlying its antidepressant activity is an area of current intense research, both clinical and preclinical evidence shows that ketamine's effects are mediated, at least in part, by molecular adaptations resulting in long-lasting synaptic changes in mesolimbic brain regions known to regulate natural and drug reward. This review outlines our limited knowledge of ketamine's neurobiological and biochemical underpinnings mediating its antidepressant effects and correlates them to its abuse potential. Depression and addiction share overlapping neural circuitry and molecular mechanisms, and though speculative, repeated use of ketamine for the treatment of depression could lead to the development of substance use disorder/addiction, and thus should be tempered with caution. There is much that remains to be known about the long-term effects of ketamine, and our lack of understanding of neurobiological mechanisms underlying its antidepressant effects is a clear limiting factor that needs to be addressed systematically before using repeated ketamine in the treatment of depressed patients.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, The University of Texas at Arlington, United States
| | - Ross J Armant
- Department of Psychology, The University of Texas at Arlington, United States
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences, Institute for Neuroscience, Texas A&M University, College Station, TX 77840, United States
| | - Linda I Perrotti
- Department of Psychology, The University of Texas at Arlington, United States.
| |
Collapse
|
49
|
Calpe-López C, García-Pardo MP, Martínez-Caballero MA, Santos-Ortíz A, Aguilar MA. Behavioral Traits Associated With Resilience to the Effects of Repeated Social Defeat on Cocaine-Induced Conditioned Place Preference in Mice. Front Behav Neurosci 2020; 13:278. [PMID: 31998090 PMCID: PMC6962131 DOI: 10.3389/fnbeh.2019.00278] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/06/2019] [Indexed: 12/25/2022] Open
Abstract
The relationship between stress and drug use is well demonstrated. Stress-induced by repeated social defeat (RSD) enhances the conditioned place preference (CPP) induced by cocaine in mice. The phenomenon of resilience understood as the ability of subjects to overcome the negative effects of stress is the focus of increasing interest. Our aim is to characterize the behavior of resilient animals with respect to the effects of RSD on the CPP induced by cocaine. To this end, 25 male C57BL/6 mice were exposed to stress by RSD during late adolescence, while other 15 male mice did not undergo stress (controls). On the 2 days following the last defeat, all the animals carried out the elevated plus maze (EPM) and Hole Board, Social Interaction, Tail Suspension and Splash tests. Three weeks later, all the animals performed the CPP paradigm with a low dose of cocaine (1 mg/kg). Exposure to RSD decreased all measurements related to the open arms of the EPM. It also reduced social interaction, immobility in the tail suspension test (TST) and grooming in the splash test. RSD exposure also increased the sensitivity of the mice to the rewarding effects of cocaine, since only defeated animals acquired CPP. Several behavioral traits were related to resilience to the potentiating effect of RSD on cocaine CPP. Mice that showed less submission during defeat episodes, a lower percentage of time in the open arms of the EPM, low novelty-seeking, high social interaction, greater immobility in the TST and a higher frequency of grooming were those that were resilient to the long-term effects of social defeat on cocaine reward since they behaved like controls and did not develop CPP. These results suggest that the behavioral profile of resilient defeated mice is characterized by an active coping response during episodes of defeat, a greater concern for potential dangers, less reactivity in a situation of inevitable moderate stress and fewer depressive-like symptoms after stress. Determining the neurobehavioral substrates of resilience is the first step towards developing behavioral or pharmacological interventions that increase resilience in individuals at a high risk of suffering from stress.
Collapse
Affiliation(s)
- Claudia Calpe-López
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Maria Pilar García-Pardo
- Department of Psychology and Sociology, Faculty of Social Sciences, University of Zaragoza, Teruel, Spain
| | - Maria Angeles Martínez-Caballero
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Alejandra Santos-Ortíz
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Maria Asunción Aguilar
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behavior Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| |
Collapse
|
50
|
Antidepressant effects of ketamine on depression-related phenotypes and dopamine dysfunction in rodent models of stress. Behav Brain Res 2019; 379:112367. [PMID: 31739001 DOI: 10.1016/j.bbr.2019.112367] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
Depression, the most prevalent psychiatric disorder, is characterized by increased negative affect (i.e. depressed mood) and reduced positive affect (i.e. anhedonia). Stress is a risk factor for depression in humans, and animal models of chronic stress are typically used to study neurobehavioral alterations relevant to depression. Common behavioral outcomes in rodent models of chronic stress include anhedonia, social dysfunction and behavioral despair. For example, chronically stressed rodents exhibit reduced reward preference, as measured by a loss of preference for sucrose solutions and time spent interacting with a novel conspecific, while also exhibiting less time struggling against inescapable stressors (e.g. forced swim, tail suspension). In both humans and rodents, anhedonia is associated with dysfunction of the dopamine (DA) system. Unlike traditional antidepressants, which are limited by inadequate efficacy and delayed therapeutic response, acute ketamine administration rapidly alleviates depressive symptoms in humans and reverses stress-induced changes in animal models. These effects are partially mediated via actions on the DA system. This review summarizes the clinical effects of ketamine, the neurobiological underpinnings of depression with a focus on DA dysfunction, as well as antidepressant effects of ketamine on depression-related endophenotypes (i.e. anhedonia, despair) and ventral tegmental area (VTA) activity in rodent models of repeated stress. Moreover, we discuss evidence regarding sex differences in ketamine's antidepressant effects, wherein females appear to be more sensitive to lower dose ketamine, as well as novel findings suggesting that ketamine has prophylactic effects with regard to protection against the neurobehavioral impact of future stressors.
Collapse
|