1
|
Bush BJ, Mohamed A, Andrews EJ, Cain G, Fawole A, Johnson H, Arocho A, Qiao Z, Paul KN, Ehlen JC. Sleep patterns predicting stress resilience are dependent on sex. Neuropsychopharmacology 2025:10.1038/s41386-025-02124-0. [PMID: 40382501 DOI: 10.1038/s41386-025-02124-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/20/2025]
Abstract
Sleep disturbances and stress have a well-established link with neuropsychiatric illness; however, the nature of this relationship remains unclear. Recently, studies using the mouse social-defeat stress model revealed a causal role for non-rapid eye movement (NREM) sleep in the maladaptive behavioral responses to stress. These results suggest a novel function for NREM sleep; as a response by cortical neurons to mitigate the maladaptive effects of stress. A major limitation in many social defeat studies has been the exclusion of females. Women exhibit a greater prevalence of both affective disorders and sleep disturbances compared to men, thus there is a clear need to understand sleep-stress interactions in females. The present study adapts recently developed female social-defeat stress models to allow social-defeat and EEG in male-female pairs. Our findings duplicate the behavioral responses that occur in other female, nondiscriminatory, and male models of social-defeat stress. Analysis of electroencephalographic (EEG) recordings, before exposure to stress, reveal that resilience is associated with differences in both NREM and REM sleep that are dependent on sex. After social defeat stress, NREM sleep was increased only in resilient males. In females, susceptibility to stress was associated with increased durations in NREM-sleep bouts. A potential cause of these sleep differences was also identified prior to stress exposure, sex differences in recovery from NREM-sleep loss; thus, suggesting an underlying sex-difference in the homeostatic process regulating sleep interactions with social-defeat stress. These findings suggest that NREM sleep quality is lower in resilient females, whereas the amount of REM sleep is decreased in susceptible females-when compared to males of the same behavioral phenotype. Overall, our findings reveal sexual dimorphism in both the sleep characteristics predicting resilience and sleep changes induced by social-defeat stress.
Collapse
Affiliation(s)
- Brittany J Bush
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Affra Mohamed
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Eva-Jeneé Andrews
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Gabrielle Cain
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Ayobami Fawole
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Hadiya Johnson
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Ashton Arocho
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Zhimei Qiao
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Ketema N Paul
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
2
|
González-Portilla M, Montagud-Romero S, Rodríguez de Fonseca F, Rodríguez-Arias M. Oleoylethanolamide restores stress-induced prepulse inhibition deficits and modulates inflammatory signaling in a sex-dependent manner. Psychopharmacology (Berl) 2025; 242:913-928. [PMID: 37314479 PMCID: PMC12043760 DOI: 10.1007/s00213-023-06403-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
RATIONALE Social stress contributes to the development of depressive and anxiety symptomatology and promotes pro-inflammatory signaling in the central nervous system. In this study, we explored the effects of a lipid messenger with anti-inflammatory properties - oleoylethanolamide (OEA) - on the behavioral deficits caused by social stress in both male and female mice. METHODS Adult mice were assigned to an experimental group according to the stress condition (control or stress) and treatment (vehicle or OEA, 10 mg/kg, i.p.). Male mice in the stress condition underwent a protocol consisting of four social defeat (SD) encounters. In the case of female mice, we employed a procedure of vicarious SD. After the stress protocol resumed, anxiety, depressive-like behavior, social interaction, and prepulse inhibition (PPI) were assessed. In addition, we characterized the stress-induced inflammatory profile by measuring IL-6 and CX3CL1 levels in the striatum and hippocampus. RESULTS Our results showed that both SD and VSD induced behavioral alterations. We found that OEA treatment restored PPI deficits in socially defeated mice. Also, OEA affected differently stress-induced anxiety and depressive-like behavior in male and female mice. Biochemical analyses showed that both male and female stressed mice showed increased levels of IL-6 in the striatum compared to control mice. Similarly, VSD female mice exhibited increased striatal CX3CL1 levels. These neuroinflammation-associated signals were not affected by OEA treatment. CONCLUSIONS In summary, our results confirm that SD and VSD induced behavioral deficits together with inflammatory signaling in the striatum and hippocampus. We observed that OEA treatment reverses stress-induced PPI alterations in male and female mice. These data suggest that OEA can exert a buffering effect on stress-related sensorimotor gating behavioral processing.
Collapse
Affiliation(s)
- Macarena González-Portilla
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Sandra Montagud-Romero
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad Clínica de Neurología, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
- Atención Primaria, Cronicidad Y Promoción de La Salud. Red de Investigación en Atención Primaria de Adicciones (RIAPAD) Rd21/0009/0005, Málaga, Spain
| | - Marta Rodríguez-Arias
- Department of Psychobiology, Faculty of Psychology, Universitat de València, Avda. Blasco Ibáñez 21, 46010, Valencia, Spain.
- Atención Primaria, Cronicidad Y Promoción de La Salud. Red de Investigación en Atención Primaria de Adicciones (RIAPAD) Rd21/0009/0005, Málaga, Spain.
| |
Collapse
|
3
|
Cramoisy S, Cabeza L, Ramadan B, Houdayer C, Haffen E, Belin D, Peterschmitt Y, Bourasset F. Cumulative effect of stress on decisional exploration-to-exploitation switch assessed through a gambling task in female mice. Brain Res 2025; 1854:149546. [PMID: 40043786 DOI: 10.1016/j.brainres.2025.149546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025]
Abstract
Survival and well-being hinge on an organism's ability to evaluate options, weighing costs and benefits to make adaptive decisions. It has long been shown that stress influences cognition and reward-related behaviour, the nature of which depends on the stressor's type and duration as well as gene x environment interactions. However, how stress influence decision-making in females has not been completely elucidated. Here, we have developed a new mouse gambling task (mGT) adapted to assess decision-making under uncertainty and risk. Adult female C57BL/6JRj mice administered with corticosterone (CORT) for 5 or 8 weeks reached similar final performance in the mGT as vehicle-treated controls. All groups tended to learn to maximize gain as the task progressed. Our results revealed that individual choice kinetics is impacted by chronic exposure to CORT, showing an accentuated sensitivity to penalties in female mice. These results confirm the suitability of our new mGT to assess decision-making under uncertainty and risk and are in line with previous reports of the effect of chronic CORT treatment on decision-making in male mice. Thereby this study provides new insights into the influence of sex and stress on decision-making.
Collapse
Affiliation(s)
- Stéphanie Cramoisy
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France.
| | - Lidia Cabeza
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Bahrie Ramadan
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Christophe Houdayer
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Emmanuel Haffen
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, service de psychiatrie de l'adulte, CIC-1431 INSERM, CHU de Besançon F-25030, France
| | - David Belin
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - Yvan Peterschmitt
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France
| | - Fanchon Bourasset
- Université Marie et Louis Pasteur, UMR INSERM 1322 LINC, F-25000 Besançon, France.
| |
Collapse
|
4
|
Geiger LT, Balouek JAR, Barrett MR, Thompson JM, Fang LZ, Farrelly LA, Chen AS, Tang M, Bennett SN, Garcia BA, Maze I, Creed MC, Peña CJ. Early-life stress alters chromatin modifications in VTA to prime stress sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.14.584631. [PMID: 38559030 PMCID: PMC10980038 DOI: 10.1101/2024.03.14.584631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Early-life stress increases sensitivity to subsequent stress, which has been observed at behavioral, neural activity, and gene expression levels. However, the molecular mechanisms underlying such long-lasting sensitivity are poorly understood. We tested the hypothesis that persistent changes in transcription and transcriptional potential were maintained at the level of the epigenome, through changes in chromatin. We used a combination of bottom-up mass spectrometry, viral-mediated epigenome-editing, RNA-sequencing, patch clamp electrophysiology of dopamine neurons, and behavioral quantification in a mouse model of early-life stress, focusing on the ventral tegmental area (VTA), a dopaminergic brain region critically implicated in motivation, reward learning, stress response, and mood and drug disorders. We found that early-life stress alters histone dynamics in VTA, including enrichment of histone-3 lysine-4 monomethylation - associated with open chromatin and primed or active enhancers - and the H3K4 monomethylase Setd7. Mimicking early-life stress through postnatal overexpression of Setd7 and enrichment of H3K4me1 in VTA sensitizes transcriptional, physiological, and behavioral response to adult stress. These findings link early-life stress experience to long-term stress hypersensitivity within the brain's dopaminergic circuitry, providing a mechanism by which early-life stress increases risk for mood and anxiety disorders later in life.
Collapse
|
5
|
Bazer A, Denney K, Chacona M, Montgomery C, Vinod S, Datta U, Samuels BA. Male and Female Mice Are Similarly Susceptible to Chronic Nondiscriminatory Social Defeat Stress Despite Differences in Attack Frequency from Aggressor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645316. [PMID: 40196656 PMCID: PMC11974878 DOI: 10.1101/2025.03.25.645316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Rationale Mood disorders are often precipitated by chronic stress and can result in an inability to adapt to the environment and increased vulnerability to challenging experiences. While diagnoses of mood disorders are diagnosed twice as frequently in women than in men, most preclinical chronic social defeat stress mouse models exclude females due to decreased aggression toward female intruders. Objectives We previously reported that the chronic non-discriminatory social defeat stress (CNSDS) paradigm is effective in both sexes, allowing for comparisons between male and female mice. We aimed to improve the screening protocol to identify CD-1 aggressors for use in CNSDS and the method for determining susceptibility to CNSDS. Finally, we aimed to determine whether susceptibility to CNSDS correlated with impaired performance in a satiety-based outcome devaluation task. Methods We analyzed CNSDS screening and social defeat sessions to determine appropriate parameters for selecting CD-1 aggressors and investigated aggressions toward male and female intruder mice. We also investigated CNSDS effects on a reward valuation task. Results We observed that despite receiving fewer attacks, female mice are equally susceptible to CNSDS as males and that CNSDS abolished satiety-based outcome devaluation in susceptible male and female mice, but not in resilient male and female mice. Conclusions These data suggest that CNSDS-defined susceptible and resilient phenotypes extend to reward behaviors.
Collapse
Affiliation(s)
- Allyson Bazer
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Behavioral and Systems Neuroscience Graduate Program, Department of Psychology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Katherine Denney
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Maria Chacona
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Behavioral and Systems Neuroscience Graduate Program, Department of Psychology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Catherine Montgomery
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Shriya Vinod
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Urboshi Datta
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Benjamin Adam Samuels
- Department of Psychology, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
- Behavioral and Systems Neuroscience Graduate Program, Department of Psychology, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
6
|
McGraw M, Christensen C, Nelson H, Li AJ, Qualls-Creekmore E. Divergent changes in social stress-induced motivation in male and female mice. Physiol Behav 2025; 291:114787. [PMID: 39710132 DOI: 10.1016/j.physbeh.2024.114787] [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: 08/05/2024] [Revised: 11/01/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Exposure to stressors has been shown to dysregulate motivated behaviors in a bidirectional manner over time. The relationship between stress and motivation is relevant to psychological disorders, including depression, binge eating, and substance use disorder; however, this relationship is not well characterized, especially in females, despite their increased risk of these disorders. Social defeat stress is a common model to study stress-induced motivation changes, however, historically this model excluded females due to lack of female-to-female aggression and unreliable male-to-female aggression. Additionally, changes in motivation are often assessed well after stress exposure ends, potentially missing or occluding changes to motivation during stress. Recently, the chronic non-discriminatory social defeat stress (CNSDS) model has demonstrated social defeat of male and female C57BL/6J mice by simultaneously exposing both mice to an aggressive male CD-1 mouse. Here we use this model to directly compare changes in the motivated behavior of male and female mice during and following chronic stress. We hypothesized that motivated behavioral responses would be dysregulated during stress and that the effects would worsen as the stress exposure continued. To monitor motivated behavior, mice had access to a Feeding Experimental Device.3 (FED3), a home cage device for operant responding. Operant responding was monitored prior to, during, and after stress by measuring nose pokes for sucrose pellets on a modified progressive ratio schedule of reinforcement. Our results demonstrated divergent behavioral outcomes between males and female mice in response to stress; where male mice increased motivated behavior during stress only, whereas female mice exhibited a decrease in motivation during and after stress. This study highlights the need to investigate the effects of stress-induced motivation over time, as well as the increased need to understand differences in the stress response in females.
Collapse
Affiliation(s)
- Megan McGraw
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Cooper Christensen
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Hailey Nelson
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Ai-Jun Li
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Emily Qualls-Creekmore
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA.
| |
Collapse
|
7
|
Yabe T, Mitsui Y, Ohnishi M, Tanigawa R, Tanizaki M, Sugiyama R, Kiriyama N, Otsuka A, Munekazu K. Social-defeat stress exposure during pregnancy induces abnormalities in spontaneous activity, sociality, and resilience to stress in offspring of mice. Behav Brain Res 2025; 480:115367. [PMID: 39631504 DOI: 10.1016/j.bbr.2024.115367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Environmental stress during prenatal periods can lead to neurodevelopmental disorders. Psychosocial stress can be studied using the social-defeat stress (SDS) animal model. However, the effects of prenatal exposure to SDS on the behavior of mature offspring mice have not been clarified. The present study assessed the spontaneous activity and social interaction of pups born to mothers exposed to SDS during gestation, as well as their post-maturity responses to environmental stimuli, focusing on changes in anxiety-like behavior following restraint stress exposure. METHODS Pregnant C57BL/6 J mice were subjected to SDS for 4 days, from E12.5-E15.5, using aggressive male ICR mice. We assessed the mature offspring (after 10 weeks of age) born to these mothers for spontaneous activity, anxiety-like behavior, and social interactions, and evaluated their activity levels post-maturity following restraint stress exposure. RESULTS The open field test (OF) indicated reduced travel distance and duration in the SDS group versus controls, whereas home-cage monitoring showed increased area traveled. In a novel environment, the SDS group showed a decrease in interest in stranger mice. In a multiple-animal rearing environment, the SDS group showed an increase in the frequency and number of contact with other individuals. Movement duration in the OF following restraint stress reduced significantly from 30 min to 4 h in the control versus SDS group. CONCLUSIONS Prenatal exposure to SDS can result in behavior resembling developmental disorders, impacting spontaneous activity and social interactions. Altered responses to stress suggest potential brain function abnormalities in offspring after maturation due to maternal SDS exposure.
Collapse
Affiliation(s)
- Tamaki Yabe
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Yuko Mitsui
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Momoka Ohnishi
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Rena Tanigawa
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Mizuki Tanizaki
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Rei Sugiyama
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Niina Kiriyama
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Airi Otsuka
- Laboratory of Nutrition and Health Science, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan
| | - Komada Munekazu
- Mammalian Embryology Laboratory, Department of Life Science, Faculty of Science and Engineering, Kindai University, Kowakae 3-4-1, Higashiosaka, Osaka 577-8502, Japan.
| |
Collapse
|
8
|
Parise LF, Joseph Burnett C, Russo SJ. Early life stress and altered social behaviors: A perspective across species. Neurosci Res 2025; 211:65-74. [PMID: 37992997 PMCID: PMC11102940 DOI: 10.1016/j.neures.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/21/2023] [Accepted: 11/04/2023] [Indexed: 11/24/2023]
Abstract
Childhood and adolescent affiliations guide how individuals engage in social relationships throughout their lifetime and adverse experiences can promote biological alterations that facilitate behavioral maladaptation. Indeed, childhood victims of abuse are more likely to be diagnosed with conduct or mood disorders which are both characterized by altered social engagement. A key domain particularly deserving of attention is aggressive behavior, a hallmark of many disorders characterized by deficits in reward processing. Animal models have been integral in identifying both the short- and long-term consequences of stress exposure and suggest that whether it is disruption to parental care or social isolation, chronic exposure to early life stress increases corticosterone, changes the expression of neurotransmitters and neuromodulators, and facilitates structural alterations to the hypothalamus, hippocampus, and amygdala, influencing how these brain regions communicate with other reward-related substrates. Herein, we describe how adverse early life experiences influence social behavioral outcomes across a wide range of species and highlight the long-term biological mechanisms that are most relevant to maladaptive aggressive behavior.
Collapse
Affiliation(s)
- Lyonna F Parise
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA.
| | - C Joseph Burnett
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Scott J Russo
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA.
| |
Collapse
|
9
|
Zhukovskaya A, Zimmerman CA, Willmore L, Pan-Vazquez A, Janarthanan SR, Lynch LA, Falkner AL, Witten IB. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. Neuron 2024; 112:3940-3956.e10. [PMID: 39393349 PMCID: PMC11624084 DOI: 10.1016/j.neuron.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 07/04/2024] [Accepted: 09/08/2024] [Indexed: 10/13/2024]
Abstract
Some individuals are susceptible to chronic stress, and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After (but not before) chronic social defeat stress, the LHb is active when susceptible mice are in proximity of the aggressor strain. During stress, activity is higher in susceptible mice during aggressor interactions, and activation biases mice toward susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical vs. cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
Affiliation(s)
- Anna Zhukovskaya
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | | | - Laura A Lynch
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Annegret L Falkner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA; Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
10
|
Schuler H, Eid RS, Wu S, Tse YC, Cvetkovska V, Lopez J, Quinn R, Zhou D, Meccia J, Dion-Albert L, Bennett SN, Newman EL, Trainor BC, Peña CJ, Menard C, Bagot RC. Data-Driven Analysis Identifies Novel Modulation of Social Behavior in Female Mice Witnessing Chronic Social Defeat Stress. Biol Psychiatry 2024:S0006-3223(24)01786-4. [PMID: 39638223 DOI: 10.1016/j.biopsych.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Chronic social defeat stress is a widely used depression model in male mice. Several proposed adaptations extend this model to females with variable, often marginal effects. We examined if the widely used male-defined metrics of stress are suboptimal in females witnessing defeat. METHODS Using a data-driven method, we comprehensively classified social interaction behavior in 761 male and female mice after chronic social witness/defeat stress, examining social modulation of behavior and associations with conventional metrics (i.e., social interaction ratio). RESULTS Social stress induced distinct behavioral adaptation patterns in defeated males and witness females. Social interaction ratio led to underpowered analyses in witness females with limited utility to differentiate susceptibility/resilience. Data-driven analyses revealed changes in social adaptation in witness females that were captured in attenuated velocity change from no target to target trials. We explored the utility of this metric in 4 female social stress models and in male witnesses. Combining social interaction ratio and velocity change optimally differentiated susceptibility/resilience in witness females and revealed resilient-specific adaptation in a resilience-associated neural circuit in female mice. CONCLUSIONS Chronic witness stress induced behavioral changes in females that were qualitatively distinct from those observed in defeated males and not adequately sampled by standard male-defined metrics. Modulation of locomotion is a robust and easily implementable metric for rigorous research in witness female mice. Overall, our findings highlight the need to critically evaluate sex differences in behavior and implement sex-based considerations in preclinical model design.
Collapse
Affiliation(s)
- Heike Schuler
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montréal, Québec, Canada
| | - Serena Wu
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Yiu-Chung Tse
- Department of Psychology, McGill University, Montréal, Québec, Canada
| | | | - Joëlle Lopez
- Department of Psychology, McGill University, Montréal, Québec, Canada
| | - Rosalie Quinn
- Department of Psychology, McGill University, Montréal, Québec, Canada
| | - Delong Zhou
- Department of Psychology, McGill University, Montréal, Québec, Canada
| | - Juliet Meccia
- Integrated Program in Neuroscience, McGill University, Montréal, Québec, Canada
| | - Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Université Laval and CERVO Brain Research Centre, Québec City, Québec, Canada
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Emily L Newman
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Division of Depression and Anxiety Disorders, Neurobiology of Fear Laboratory, McLean Hospital, Belmont, Massachusetts
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, Davis, California
| | - Catherine J Peña
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Université Laval and CERVO Brain Research Centre, Québec City, Québec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montréal, Québec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Montréal, Québec, Canada.
| |
Collapse
|
11
|
Navarrete J, Schneider KN, Smith BM, Goodwin NL, Zhang YY, Salazar AS, Gonzalez YE, Anumolu P, Gross E, Tsai VS, Heshmati M, Golden SA. Individual Differences in Volitional Social Self-Administration and Motivation in Male and Female Mice Following Social Stress. Biol Psychiatry 2024; 96:309-321. [PMID: 38244753 PMCID: PMC11255129 DOI: 10.1016/j.biopsych.2024.01.007] [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: 07/14/2023] [Revised: 12/18/2023] [Accepted: 01/06/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND A key challenge in developing treatments for neuropsychiatric illness is the disconnect between preclinical models and the complexity of human social behavior. We integrate voluntary social self-administration into a rodent model of social stress as a platform for the identification of fundamental brain and behavior mechanisms underlying stress-induced individual differences in social motivation. METHODS Here, we introduced an operant social stress procedure in male and female mice composed of 3 phases: 1) social self-administration training, 2) social stress exposure concurrent with reinforced self-administration testing, and 3) poststress operant testing under nonreinforced and reinforced conditions. We used social-defeat and witness-defeat stress in male and female mice. RESULTS Social defeat attenuated social reward seeking in males but not females, whereas witness defeat had no effect in males but promoted seeking behavior in females. We resolved social stress-induced changes to social motivation by aggregating z-scored operant metrics into a cumulative social index score to describe the spectrum of individual differences exhibited during operant social stress. Clustering does not adequately describe the relative distributions of social motivation following stress and is better described as a nonbinary behavioral distribution defined by the social index score, capturing a dynamic range of stress-related alterations in social motivation inclusive of sex as a biological variable. CONCLUSIONS We demonstrated that operant social stress can detect stable individual differences in stress-induced changes to social motivation. The inclusion of volitional behavior in social procedures may enhance the understanding of behavioral adaptations that promote stress resiliency and their mechanisms under more naturalistic conditions.
Collapse
Affiliation(s)
- Jovana Navarrete
- Department of Biological Structure, University of Washington, Seattle, Washington; Graduate Program in Neuroscience, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington
| | - Kevin N Schneider
- Department of Biological Structure, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington
| | - Briana M Smith
- Department of Biological Structure, University of Washington, Seattle, Washington
| | - Nastacia L Goodwin
- Department of Biological Structure, University of Washington, Seattle, Washington; Graduate Program in Neuroscience, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington
| | - Yizhe Y Zhang
- Department of Biological Structure, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington
| | - Axelle S Salazar
- Department of Biological Structure, University of Washington, Seattle, Washington
| | - Yahir E Gonzalez
- Department of Biological Structure, University of Washington, Seattle, Washington; Undergraduate Neuroscience Program, University of Washington, Seattle, Washington
| | - Pranav Anumolu
- Department of Biological Structure, University of Washington, Seattle, Washington; Undergraduate Neuroscience Program, University of Washington, Seattle, Washington
| | - Ethan Gross
- Department of Biological Structure, University of Washington, Seattle, Washington
| | - Valerie S Tsai
- Department of Biological Structure, University of Washington, Seattle, Washington; Undergraduate Neuroscience Program, University of Washington, Seattle, Washington
| | - Mitra Heshmati
- Department of Biological Structure, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Sam A Golden
- Department of Biological Structure, University of Washington, Seattle, Washington; Graduate Program in Neuroscience, University of Washington, Seattle, Washington; Center of Excellence in Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, Washington.
| |
Collapse
|
12
|
Zhukovskaya A, Christopher Z, Willmore L, Pan Vazquez A, Janarthanan S, Falkner A, Witten I. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565681. [PMID: 39005438 PMCID: PMC11244933 DOI: 10.1101/2023.11.06.565681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Some individuals are susceptible to the experience of chronic stress and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After, but not before, chronic social defeat stress (CSDS), the LHb is active when susceptible mice are in the proximity of the aggressor strain. During stress itself, LHb activity is higher in susceptible mice during aggressor proximity, and activation of the LHb during stress biases mice towards susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical versus cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
|
13
|
Poitras M, Lebeau M, Plamondon H. The cycle of stress: A systematic review of the impact of chronic psychological stress models on the rodent estrous cycle. Neurosci Biobehav Rev 2024; 162:105730. [PMID: 38763179 DOI: 10.1016/j.neubiorev.2024.105730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Stress is known to impair reproduction through interactions between the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes. However, while it is well accepted that stress can alter estrous cycle regularity, a key indicator of female's HPG axis function, effects of different types of psychological stress have been inconsistent. This systematic review evaluated the impact of rodent models of psychological stress on estrous cyclicity, while reporting biological parameters pertaining to HPA or HPG axis function assessed within these studies. We performed a systematic database search and included articles that implemented a psychological stress model in rodents and reported estrous cyclicity for at least two cycles after initiation of stress. Of the 32 studies included, 62.5% reported post-stress alterations to estrous cyclicity, with Chronic Mild Stress (CMS) models showing the most conclusive effects. Twenty-five studies measured HPG or HPA axis markers, with cycle disruptions being commonly observed in parallel with altered estradiol and increased corticosterone levels. Our review highlights gaps in reporting estrous cyclicity assessments and makes recommendations to improve comparability between studies.
Collapse
Affiliation(s)
- Marilou Poitras
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - Madison Lebeau
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada
| | - Hélène Plamondon
- Cerebro Vascular Accidents and Behavioural Recovery Laboratory, School of Psychology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
14
|
Kalisch R, Russo SJ, Müller MB. Neurobiology and systems biology of stress resilience. Physiol Rev 2024; 104:1205-1263. [PMID: 38483288 PMCID: PMC11381009 DOI: 10.1152/physrev.00042.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 05/16/2024] Open
Abstract
Stress resilience is the phenomenon that some people maintain their mental health despite exposure to adversity or show only temporary impairments followed by quick recovery. Resilience research attempts to unravel the factors and mechanisms that make resilience possible and to harness its insights for the development of preventative interventions in individuals at risk for acquiring stress-related dysfunctions. Biological resilience research has been lagging behind the psychological and social sciences but has seen a massive surge in recent years. At the same time, progress in this field has been hampered by methodological challenges related to finding suitable operationalizations and study designs, replicating findings, and modeling resilience in animals. We embed a review of behavioral, neuroimaging, neurobiological, and systems biological findings in adults in a critical methods discussion. We find preliminary evidence that hippocampus-based pattern separation and prefrontal-based cognitive control functions protect against the development of pathological fears in the aftermath of singular, event-type stressors [as found in fear-related disorders, including simpler forms of posttraumatic stress disorder (PTSD)] by facilitating the perception of safety. Reward system-based pursuit and savoring of positive reinforcers appear to protect against the development of more generalized dysfunctions of the anxious-depressive spectrum resulting from more severe or longer-lasting stressors (as in depression, generalized or comorbid anxiety, or severe PTSD). Links between preserved functioning of these neural systems under stress and neuroplasticity, immunoregulation, gut microbiome composition, and integrity of the gut barrier and the blood-brain barrier are beginning to emerge. On this basis, avenues for biological interventions are pointed out.
Collapse
Affiliation(s)
- Raffael Kalisch
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
- Neuroimaging Center (NIC), Focus Program Translational Neuroscience (FTN), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Scott J Russo
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Marianne B Müller
- Leibniz Institute for Resilience Research (LIR), Mainz, Germany
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
15
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. Horm Behav 2024; 159:105472. [PMID: 38141539 PMCID: PMC10922504 DOI: 10.1016/j.yhbeh.2023.105472] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 12/25/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons, Otx2, shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
Affiliation(s)
| | - Austin B Chang
- Princeton Neuroscience Institute, Princeton University, USA
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, USA; Department of Molecular Biology, Princeton University, USA
| | - Parker Jones
- Princeton Neuroscience Institute, Princeton University, USA
| | | |
Collapse
|
16
|
Ito M, Ito H, Miyoshi K, Kanai-Azuma M. Chronic non-discriminatory social defeat stress during the perinatal period induces depressive-like outcomes in female mice. Brain Res 2024; 1825:148734. [PMID: 38110072 DOI: 10.1016/j.brainres.2023.148734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/13/2023] [Accepted: 12/15/2023] [Indexed: 12/20/2023]
Abstract
Depression is more prevalent in women than in men. Perinatal stress is one of the main risk factors for depression in women. However, there is no suitable female model for perinatal depression that uses the social defeat stress (SDS) paradigm. The standard chronic SDS protocol, which is the most useful method for developing a depression-like model, is effective only in male mice. Thus, this study aimed to characterize a novel SDS method for producing a perinatal depression-like model mouse. We induced chronic SDS in perinatal female mice, wherein chronic non-discriminatory SDS (ND-SDS) was used to induce substantial stress in female mice. The female mice were placed in aggressive ICR mouse cages with sentinel male mice under ND-SDS conditions. Stressed female mice subjected to ND-SDS during the perinatal period efficiently exhibited stress-susceptible phenotypes, such as a social avoidance phenotype and anhedonic behavior, whereas stressed female mice subjected to SDS did not show depressive-like behaviors. These results indicate that chronic ND-SDS in perinatal females could be used to develop a female perinatal depression-like model that can be used to study women's health.
Collapse
Affiliation(s)
- Masumi Ito
- Department of Basic Life Science, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan; Research Facility Center for Science and Technology, Kagawa University, 1750-1, Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Hikaru Ito
- Department of Basic Life Science, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan; Research Facility Center for Science and Technology, Kagawa University, 1750-1, Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan; Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan; Center for Experimental Animals, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan.
| | - Kaori Miyoshi
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan; Center for Experimental Animals, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan; Center for Experimental Animals, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan
| |
Collapse
|
17
|
Pantoja-Urbán AH, Richer S, Mittermaier A, Giroux M, Nouel D, Hernandez G, Flores C. Gains and Losses: Resilience to Social Defeat Stress in Adolescent Female Mice. Biol Psychiatry 2024; 95:37-47. [PMID: 37355003 PMCID: PMC10996362 DOI: 10.1016/j.biopsych.2023.06.014] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Adolescence is a unique period of psychosocial growth during which social adversity can negatively influence mental health trajectories. Understanding how adolescent social stress impacts males and females and why some individuals are particularly affected is becoming increasingly urgent. Social defeat stress models for adolescent male mice have been effective in reproducing some physical/psychological aspects of bullying. Designing a model suitable for females has proven challenging. METHODS We report a version of the adolescent male accelerated social defeat stress (AcSD) paradigm adapted for females. Early adolescent C57BL/6J female mice (N = 107) were exposed to our modified AcSD procedure twice a day for 4 days and categorized as resilient or susceptible based on a social interaction test 24 hours later. Mice were then assessed for changes in Netrin-1/DCC guidance cue expression in dopamine systems, for inhibitory control in adulthood using the Go/No-Go task, or for alterations in dopamine connectivity organization in the matured prefrontal cortex. RESULTS Most adolescent females showed protection against stress-induced social avoidance, but in adulthood, these resilient females developed inhibitory control deficits and showed diminution of prefrontal cortex presynaptic dopamine sites. Female mice classified as susceptible were protected against cognitive and dopaminergic alterations. AcSD did not alter Netrin-1/DCC in early adolescent females, contrary to previous findings with males. CONCLUSIONS Preserving prosocial behavior in adolescent females may be important for survival advantage but seems to come at the price of developing persistent cognitive and dopamine deficiencies. The female AcSD paradigm produced findings comparable to those found in males, allowing mechanistic investigation in both sexes.
Collapse
Affiliation(s)
- Andrea Harée Pantoja-Urbán
- Integrated Program in Neuroscience, McGill University, Montreal, Québec, Canada; Douglas Mental Health University Institute, Montreal, Québec, Canada
| | - Samuel Richer
- Integrated Program in Neuroscience, McGill University, Montreal, Québec, Canada; Douglas Mental Health University Institute, Montreal, Québec, Canada
| | | | - Michel Giroux
- Douglas Mental Health University Institute, Montreal, Québec, Canada
| | - Dominique Nouel
- Douglas Mental Health University Institute, Montreal, Québec, Canada
| | | | - Cecilia Flores
- Douglas Mental Health University Institute, Montreal, Québec, Canada; Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
18
|
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
|
19
|
Parel ST, Bennett SN, Cheng CJ, Timmermans OC, Fiori LM, Turecki G, Peña CJ. Transcriptional signatures of early-life stress and antidepressant treatment efficacy. Proc Natl Acad Sci U S A 2023; 120:e2305776120. [PMID: 38011563 DOI: 10.1073/pnas.2305776120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 11/29/2023] Open
Abstract
Individuals with a history of early-life stress (ELS) tend to have an altered course of depression and lower treatment response rates. Research suggests that ELS alters brain development, but the molecular changes in the brain following ELS that may mediate altered antidepressant response have not been systematically studied. Sex and gender also impact the risk of depression and treatment response. Here, we leveraged existing RNA sequencing datasets from 1) blood samples from depressed female- and male-identifying patients treated with escitalopram or desvenlafaxine and assessed for treatment response or failure; 2) the nucleus accumbens (NAc) of female and male mice exposed to ELS and/or adult stress; and 3) the NAc of mice after adult stress, antidepressant treatment with imipramine or ketamine, and assessed for treatment response or failure. We find that transcriptomic signatures of adult stress after a history of ELS correspond with transcriptomic signatures of treatment nonresponse, across species and multiple classes of antidepressants. Transcriptomic correspondence with treatment outcome was stronger among females and weaker among males. We next pharmacologically tested these predictions in our mouse model of early-life and adult social defeat stress and treatment with either chronic escitalopram or acute ketamine. Among female mice, the strongest predictor of behavior was an interaction between ELS and ketamine treatment. Among males, however, early experience and treatment were poor predictors of behavior, mirroring our bioinformatic predictions. These studies provide neurobiological evidence for molecular adaptations in the brain related to sex and ELS that contribute to antidepressant treatment response.
Collapse
Affiliation(s)
- Sero Toriano Parel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Cindy J Cheng
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | | | - Laura M Fiori
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | - Gustavo Turecki
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | | |
Collapse
|
20
|
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: 3.0] [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
|
21
|
Jin H, Xu G, Lu Y, Niu C, Zhang X, Kan T, Cao J, Yang X, Cheng Q, Zhang J, Dong J. Fluoxetine partially alleviates inflammation in the kidney of socially stressed male C57 BL/6 mice. FEBS Open Bio 2023; 13:1723-1736. [PMID: 37400956 PMCID: PMC10476569 DOI: 10.1002/2211-5463.13670] [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: 01/27/2023] [Revised: 06/19/2023] [Accepted: 07/01/2023] [Indexed: 07/05/2023] Open
Abstract
Stress-related illnesses are linked to the onset and progression of renal diseases and depressive disorders. To investigate stress-induced changes in the renal transcriptome associated with the development of depressive behaviors, we generated here a chronic social defeat stress (CSDS) model of C57 BL/6 male mice and then performed RNA sequencing of the kidneys to obtain an inflammation-related transcriptome. Administration of the antidepressant drug fluoxetine (10 mg·kg-1 ·day-1 ) during CSDS induction could partially alleviate renal inflammation and reverse CSDS-induced depression-like behaviors. Moreover, fluoxetine also modulated gene expression of stress-related hormone receptors, including prolactin and melanin-concentrating hormone. These results suggest that CSDS can induce gene expression changes associated with inflammation in the kidney of C57 BL/6 male mice, and this inflammation can be treated effectively by fluoxetine.
Collapse
Affiliation(s)
- Hailong Jin
- The Third CenterPLA General HospitalBeijingChina
| | - Guanglei Xu
- Beijing Institute of Basic Medical SciencesChina
| | - Yuchen Lu
- Beijing Institute of Basic Medical SciencesChina
| | - Chunxiao Niu
- Beijing Institute of Basic Medical SciencesChina
| | | | - Tongtong Kan
- Beijing Institute of Basic Medical SciencesChina
| | - Junxia Cao
- Beijing Institute of Basic Medical SciencesChina
| | - Xiqin Yang
- Beijing Institute of Basic Medical SciencesChina
| | | | - Jiyan Zhang
- Beijing Institute of Basic Medical SciencesChina
| | - Jie Dong
- Beijing Institute of Basic Medical SciencesChina
| |
Collapse
|
22
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554785. [PMID: 37662236 PMCID: PMC10473690 DOI: 10.1101/2023.08.25.554785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
|
23
|
Balouek JA, Mclain CA, Minerva AR, Rashford RL, Bennett SN, Rogers FD, Peña CJ. Reactivation of Early-Life Stress-Sensitive Neuronal Ensembles Contributes to Lifelong Stress Hypersensitivity. J Neurosci 2023; 43:5996-6009. [PMID: 37429717 PMCID: PMC10451005 DOI: 10.1523/jneurosci.0016-23.2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/01/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023] Open
Abstract
Early-life stress (ELS) is one of the strongest lifetime risk factors for depression, anxiety, suicide, and other psychiatric disorders, particularly after facing additional stressful events later in life. Human and animal studies demonstrate that ELS sensitizes individuals to subsequent stress. However, the neurobiological basis of such stress sensitization remains largely unexplored. We hypothesized that ELS-induced stress sensitization would be detectable at the level of neuronal ensembles, such that cells activated by ELS would be more reactive to adult stress. To test this, we leveraged transgenic mice to genetically tag, track, and manipulate experience-activated neurons. We found that in both male and female mice, ELS-activated neurons within the nucleus accumbens (NAc), and to a lesser extent the medial prefrontal cortex, were preferentially reactivated by adult stress. To test whether reactivation of ELS-activated ensembles in the NAc contributes to stress hypersensitivity, we expressed hM4Dis receptor in control or ELS-activated neurons of pups and chemogenetically inhibited their activity during experience of adult stress. Inhibition of ELS-activated NAc neurons, but not control-tagged neurons, ameliorated social avoidance behavior following chronic social defeat stress in males. These data provide evidence that ELS-induced stress hypersensitivity is encoded at the level of corticolimbic neuronal ensembles.SIGNIFICANCE STATEMENT Early-life stress enhances sensitivity to stress later in life, yet the mechanisms of such stress sensitization are largely unknown. Here, we show that neuronal ensembles in corticolimbic brain regions remain hypersensitive to stress across the life span, and quieting these ensembles during experience of adult stress rescues stress hypersensitivity.
Collapse
Affiliation(s)
- Julie-Anne Balouek
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Christabel A Mclain
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Rebekah L Rashford
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|
24
|
Zhu X, Sakamoto S, Ishii C, Smith MD, Ito K, Obayashi M, Unger L, Hasegawa Y, Kurokawa S, Kishimoto T, Li H, Hatano S, Wang TH, Yoshikai Y, Kano SI, Fukuda S, Sanada K, Calabresi PA, Kamiya A. Dectin-1 signaling on colonic γδ T cells promotes psychosocial stress responses. Nat Immunol 2023; 24:625-636. [PMID: 36941398 DOI: 10.1038/s41590-023-01447-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 02/02/2023] [Indexed: 03/23/2023]
Abstract
The intestinal immune system interacts with commensal microbiota to maintain gut homeostasis. Furthermore, stress alters the microbiome composition, leading to impaired brain function; yet how the intestinal immune system mediates these effects remains elusive. Here we report that colonic γδ T cells modulate behavioral vulnerability to chronic social stress via dectin-1 signaling. We show that reduction in specific Lactobacillus species, which are involved in T cell differentiation to protect the host immune system, contributes to stress-induced social-avoidance behavior, consistent with our observations in patients with depression. Stress-susceptible behaviors derive from increased differentiation in colonic interleukin (IL)-17-producing γδ T cells (γδ17 T cells) and their meningeal accumulation. These stress-susceptible cellular and behavioral phenotypes are causally mediated by dectin-1, an innate immune receptor expressed in γδ T cells. Our results highlight the previously unrecognized role of intestinal γδ17 T cells in the modulation of psychological stress responses and the importance of dectin-1 as a potential therapeutic target for the treatment of stress-induced behaviors.
Collapse
Affiliation(s)
- Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shinji Sakamoto
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Chiharu Ishii
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koki Ito
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Mizuho Obayashi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa Unger
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Yuto Hasegawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shunya Kurokawa
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Taishiro Kishimoto
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
- Hills Joint Research Laboratory for Future Preventive Medicine and Wellness, Keio University School of Medicine, Tokyo, Japan
| | - Hui Li
- Departments of Mechanical Engineering and Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- School of Electrical, Computer and Biomedical Engineering, Southern Illinois University, Carbondale, IL, USA
| | - Shinya Hatano
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tza-Huei Wang
- Departments of Mechanical Engineering and Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yasunobu Yoshikai
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shin-Ichi Kano
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Japan
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan
- Laboratory for Regenerative Microbiology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kenji Sanada
- Department of Psychiatry, School of Medicine, Showa University, Tokyo, Japan
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Mahmud A, Avramescu RG, Niu Z, Flores C. Awakening the dormant: Role of axonal guidance cues in stress-induced reorganization of the adult prefrontal cortex leading to depression-like behavior. Front Neural Circuits 2023; 17:1113023. [PMID: 37035502 PMCID: PMC10079902 DOI: 10.3389/fncir.2023.1113023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic and disabling disorder affecting roughly 280 million people worldwide. While multiple brain areas have been implicated, dysfunction of prefrontal cortex (PFC) circuitry has been consistently documented in MDD, as well as in animal models for stress-induced depression-like behavioral states. During brain development, axonal guidance cues organize neuronal wiring by directing axonal pathfinding and arborization, dendritic growth, and synapse formation. Guidance cue systems continue to be expressed in the adult brain and are emerging as important mediators of synaptic plasticity and fine-tuning of mature neural networks. Dysregulation or interference of guidance cues has been linked to depression-like behavioral abnormalities in rodents and MDD in humans. In this review, we focus on the emerging role of guidance cues in stress-induced changes in adult prefrontal cortex circuitry and in precipitating depression-like behaviors. We discuss how modulating axonal guidance cue systems could be a novel approach for precision medicine and the treatment of depression.
Collapse
Affiliation(s)
- Ashraf Mahmud
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | | | - Zhipeng Niu
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Cecilia Flores
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Department of Psychiatry, Neurology, and Neurosurgery, McGill University, Montréal, QC, Canada
| |
Collapse
|
26
|
Ecological validity of social defeat stressors in mouse models of vulnerability and resilience. Neurosci Biobehav Rev 2023; 145:105032. [PMID: 36608919 DOI: 10.1016/j.neubiorev.2023.105032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Laboratory mouse models offer opportunities to bridge the gap between basic neuroscience and applied stress research. Here we consider the ecological validity of social defeat stressors in mouse models of emotional vulnerability and resilience. Reports identified in PubMed from 1980 to 2020 are reviewed for the ecological validity of social defeat stressors, sex of subjects, and whether results are discussed in terms of vulnerability alone, resilience alone, or both vulnerability and resilience. Most of the 318 reviewed reports (95%) focus on males, and many reports (71%) discuss vulnerability and resilience. Limited ecological validity is associated with increased vulnerability and decreased resilience. Elements of limited ecological validity include frequent and repeated exposure to defeat stressors without opportunities to avoid or escape from unfamiliar conspecifics that are pre-screened and selected for aggressive behavior. These elements ensure defeat and may be required to induce vulnerability, but they are not representative of naturalistic conditions. Research aimed at establishing causality is needed to determine whether ecologically valid stressors build resilience in both sexes of mice.
Collapse
|
27
|
Kraeuter AK. The use of integrated behavioural z-scoring in behavioural neuroscience - A perspective article. J Neurosci Methods 2023; 384:109751. [PMID: 36435327 DOI: 10.1016/j.jneumeth.2022.109751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Complex pathophysiology in psychiatric disorders results in difficulties interpreting pre-clinical data. Guilloux et al. (2011b), proposed an integrated behavioural z-scoring procedure to improve the predictive validity of animal models by converging evidence similarly used to diagnose mental health conditions in humans. Here, I set out to give a brief review of the current methodology and literature using integrated behavioural z-scoring. Secondly, I will discuss the benefits and downfalls of integrated behavioural z-scoring and its potential future applications. Integrated behavioural z-scoring is a methodology used most frequently within animal models of depression and anxiety. Here, I am suggesting broadening the application of integrated behavioural z-scoring beyond the field of depression and anxiety to a three-step methodology to obtain disease-specific behavioural z-scores (i.e Schizophrenia index, Alzheimer's disease index) to aid translatability and interpretation of data. Lastly, I suggest integrating not only behaviour but also biological variables to create converging psychological and physiological evidence to sustain face and construct validity, while improving predict validity.
Collapse
Affiliation(s)
- Ann-Katrin Kraeuter
- Faculty of Health and Life Sciences, Psychology, Northumbria University, Newcastle upon Tyne, UK.
| |
Collapse
|
28
|
Willmore L, Cameron C, Yang J, Witten IB, Falkner AL. Behavioural and dopaminergic signatures of resilience. Nature 2022; 611:124-132. [PMID: 36261520 PMCID: PMC10026178 DOI: 10.1038/s41586-022-05328-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/07/2022] [Indexed: 12/14/2022]
Abstract
Chronic stress can have lasting adverse consequences in some individuals, yet others are resilient to the same stressor1,2. Susceptible and resilient individuals exhibit differences in the intrinsic properties of mesolimbic dopamine (DA) neurons after the stressful experience is over3-8. However, the causal links between DA, behaviour during stress and individual differences in resilience are unknown. Here we recorded behaviour in mice simultaneously with DA neuron activity in projections to the nucleus accumbens (NAc) (which signals reward9-12) and the tail striatum (TS) (which signals threat13-16) during social defeat. Supervised and unsupervised behavioural quantification revealed that during stress, resilient and susceptible mice use different behavioural strategies and have distinct activity patterns in DA terminals in the NAc (but not the TS). Neurally, resilient mice have greater activity near the aggressor, including at the onset of fighting back. Conversely, susceptible mice have greater activity at the offset of attacks and onset of fleeing. We also performed optogenetic stimulation of NAc-projecting DA neurons in open loop (randomly timed) during defeat or timed to specific behaviours using real-time behavioural classification. Both open-loop and fighting-back-timed activation promoted resilience and reorganized behaviour during defeat towards resilience-associated patterns. Together, these data provide a link between DA neural activity, resilience and resilience-associated behaviour during the experience of stress.
Collapse
Affiliation(s)
- Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Courtney Cameron
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - John Yang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
- Department of Psychology, Princeton University, Princeton, NJ, USA.
| | - Annegret L Falkner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
29
|
Bush BJ, Donnay C, Andrews EJA, Lewis-Sanders D, Gray CL, Qiao Z, Brager AJ, Johnson H, Brewer HCS, Sood S, Saafir T, Benveniste M, Paul KN, Ehlen JC. Non-rapid eye movement sleep determines resilience to social stress. eLife 2022; 11:e80206. [PMID: 36149059 PMCID: PMC9586557 DOI: 10.7554/elife.80206] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Resilience, the ability to overcome stressful conditions, is found in most mammals and varies significantly among individuals. A lack of resilience can lead to the development of neuropsychiatric and sleep disorders, often within the same individual. Despite extensive research into the brain mechanisms causing maladaptive behavioral-responses to stress, it is not clear why some individuals exhibit resilience. To examine if sleep has a determinative role in maladaptive behavioral-response to social stress, we investigated individual variations in resilience using a social-defeat model for male mice. Our results reveal a direct, causal relationship between sleep amount and resilience-demonstrating that sleep increases after social-defeat stress only occur in resilient mice. Further, we found that within the prefrontal cortex, a regulator of maladaptive responses to stress, pre-existing differences in sleep regulation predict resilience. Overall, these results demonstrate that increased NREM sleep, mediated cortically, is an active response to social-defeat stress that plays a determinative role in promoting resilience. They also show that differences in resilience are strongly correlated with inter-individual variability in sleep regulation.
Collapse
Affiliation(s)
- Brittany J Bush
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Caroline Donnay
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | | | | | - Cloe L Gray
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Zhimei Qiao
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Allison J Brager
- Behavioral Biology Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of ResearchSilver SpringUnited States
| | - Hadiya Johnson
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Hamadi CS Brewer
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Sahil Sood
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Talib Saafir
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Morris Benveniste
- Neuroscience Institute, Morehouse School of MedicineAtlantaUnited States
| | - Ketema N Paul
- Department of Integrative Biology and Physiology, University of California, Los AngelesLos AngelesUnited States
| | | |
Collapse
|
30
|
Maita I, Roepke TA, Samuels BA. Chronic stress-induced synaptic changes to corticotropin-releasing factor-signaling in the bed nucleus of the stria terminalis. Front Behav Neurosci 2022; 16:903782. [PMID: 35983475 PMCID: PMC9378865 DOI: 10.3389/fnbeh.2022.903782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
The sexually dimorphic bed nucleus of the stria terminalis (BNST) is comprised of several distinct regions, some of which act as a hub for stress-induced changes in neural circuitry and behavior. In rodents, the anterodorsal BNST is especially affected by chronic exposure to stress, which results in alterations to the corticotropin-releasing factor (CRF)-signaling pathway, including CRF receptors and upstream regulators. Stress increases cellular excitability in BNST CRF+ neurons by potentiating miniature excitatory postsynaptic current (mEPSC) amplitude, altering the resting membrane potential, and diminishing M-currents (a voltage-gated K+ current that stabilizes membrane potential). Rodent anterodorsal and anterolateral BNST neurons are also critical regulators of behavior, including avoidance of aversive contexts and fear learning (especially that of sustained threats). These rodent behaviors are historically associated with anxiety. Furthermore, BNST is implicated in stress-related mood disorders, including anxiety and Post-Traumatic Stress Disorders in humans, and may be linked to sex differences found in mood disorders.
Collapse
Affiliation(s)
- Isabella Maita
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Troy A. Roepke
- Roepke Laboratory, Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Benjamin A. Samuels
- Samuels Laboratory, Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
31
|
Bhatti DL, Medrihan L, Chen MX, Jin J, McCabe KA, Wang W, Azevedo EP, Ledo JH, Kim Y. Molecular and Cellular Adaptations in Hippocampal Parvalbumin Neurons Mediate Behavioral Responses to Chronic Social Stress. Front Mol Neurosci 2022; 15:898851. [PMID: 35813065 PMCID: PMC9268684 DOI: 10.3389/fnmol.2022.898851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin-expressing interneurons (PV neurons) maintain inhibitory control of local circuits implicated in behavioral responses to environmental stressors. However, the roles of molecular and cellular adaptations in PV neurons in stress susceptibility or resilience have not been clearly established. Here, we show behavioral outcomes of chronic social defeat stress (CSDS) are mediated by differential neuronal activity and gene expression in hippocampal PV neurons in mice. Using in vivo electrophysiology and chemogenetics, we find increased PV neuronal activity in the ventral dentate gyrus is required and sufficient for behavioral susceptibility to CSDS. PV neuron-selective translational profiling indicates mitochondrial oxidative phosphorylation is the most significantly altered pathway in stress-susceptible versus resilient mice. Among differentially expressed genes associated with stress-susceptibility and resilience, we find Ahnak, an endogenous regulator of L-type calcium channels which are implicated in the regulation of mitochondrial function and gene expression. Notably, Ahnak deletion in PV neurons impedes behavioral susceptibility to CSDS. Altogether, these findings indicate behavioral effects of chronic stress can be controlled by selective modulation of PV neuronal activity or a regulator of L-type calcium signaling in PV neurons.
Collapse
Affiliation(s)
- Dionnet L. Bhatti
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Lucian Medrihan
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Michelle X. Chen
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Junghee Jin
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Kathryn A. McCabe
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Wei Wang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Bioinformatics Resource Center, The Rockefeller University, New York, NY, United States
| | - Estefania P. Azevedo
- Laboratory of Molecular Genetics, The Rockefeller University, New York, NY, United States
| | - Jose H. Ledo
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Yong Kim
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Yong Kim,
| |
Collapse
|
32
|
Trainor BC, Falkner AL. Quantifying Sex Differences in Behavior in the Era of "Big" Data. Cold Spring Harb Perspect Biol 2022; 14:a039164. [PMID: 34607831 PMCID: PMC9159265 DOI: 10.1101/cshperspect.a039164] [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] [Indexed: 11/25/2022]
Abstract
Sex differences are commonly observed in behaviors that are closely linked to adaptive function, but sex differences can also be observed in behavioral "building blocks" such as locomotor activity and reward processing. Modern neuroscientific inquiry, in pursuit of generalizable principles of functioning across sexes, has often ignored these more subtle sex differences in behavioral building blocks that may result from differences in these behavioral building blocks. A frequent assumption is that there is a default (often male) way to perform a behavior. This approach misses fundamental drivers of individual variability within and between sexes. Incomplete behavioral descriptions of both sexes can lead to an overreliance on reduced "single-variable" readouts of complex behaviors, the design of which may be based on male-biased samples. Here, we advocate that the incorporation of new machine-learning tools for collecting and analyzing multimodal "big behavior" data allows for a more holistic and richer approach to the quantification of behavior in both sexes. These new tools make behavioral description more robust and replicable across laboratories and species, and may open up new lines of neuroscientific inquiry by facilitating the discovery of novel behavioral states. Having more accurate measures of behavioral diversity in males and females could serve as a hypothesis generator for where and when we should look in the brain for meaningful neural differences.
Collapse
Affiliation(s)
- Brian C Trainor
- Department of Psychology, University of California, Davis, California 95616, USA
| | | |
Collapse
|
33
|
Wang W, Liu WZ, Wang ZL, Duan DX, Wang XY, Liu SJ, Wang ZJ, Xing GG, Xing Y. Spinal microglial activation promotes perioperative social defeat stress-induced prolonged postoperative pain in a sex-dependent manner. Brain Behav Immun 2022; 100:88-104. [PMID: 34808295 DOI: 10.1016/j.bbi.2021.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/08/2021] [Accepted: 11/13/2021] [Indexed: 11/25/2022] Open
Abstract
Prolonged postsurgical pain, which is associated with multiple risk factors in the perioperative stage, is a common medical and social problem worldwide. Suitable animal models should be established to elucidate the mechanisms underlying the perioperative prolonged postsurgical pain. In this study, standard and modified social defeat stress mice models, including chronic social defeat stress (CSDS), chronic nondiscriminatory social defeat stress (CNSDS) and vicarious social defeat stress (VSDS), were applied to explore the effect of perioperative social defeat stress on postsurgical pain in male and female mice. Our results showed that exposure to preoperative CSDS could induce prolonged postsurgical pain in defeated mice regardless of susceptibility or resilience differentiated by the social interaction test. Similar prolongation of incision-induced mechanical hypersensitivity was also observed in both sexes upon exposing to CNSDS or VSDS in the preoperative period. Moreover, we found that using the modified CNSDS or VSDS models at different recovery stages after surgery could still promote abnormal pain without sex differences. Further studies revealed the key role of spinal microglial activation in the stress-induced transition from acute to prolonged postoperative pain in male but not female mice. Together, these data indicate that perioperative social defeat stress is a vital risk factor for developing prolonged postoperative pain in both sexes, but the promotion of stress-induced prolonged postoperative pain by spinal microglial activation is sexually dimorphic in mice.
Collapse
Affiliation(s)
- Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; The Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Wei-Zhen Liu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; The Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zi-Liang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; The Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Dong-Xiao Duan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xue-Yun Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; The Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Shi-Jin Liu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; The Academy of Medical Sciences of Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhi-Ju Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Guo-Gang Xing
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing 100191, China.
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
34
|
Abstract
Social stressors are known to have strong negative impacts on mental health. There is a long history of preclinical social defeat stress studies in rodents focusing on males that has produced important insights into the neural mechanisms that modulate depression- and anxiety-related behavior. Despite these impressive results, a historical weakness of rodent social stress models has been an under-representation of studies in females. This is problematic because rates of depression and anxiety are higher in women versus men. Recently there has been a surge of interest in adapting social stress methods for female rodents. Here we review new rodent models that have investigated numerous facets of social stress in females. The different models have different strengths and weaknesses, with some model systems having stronger ethological validity with other models having better access to molecular tools to manipulate neural circuits. Continued use and refinement of these complementary models will be critical for addressing gaps in understanding the function of neural circuits modulating depression- and anxiety-related behavior in females.
Collapse
Affiliation(s)
- Jace X Kuske
- Department of Psychology, University of California, Davis, CA, USA
| | - Brian C Trainor
- Department of Psychology, University of California, Davis, CA, USA.
| |
Collapse
|
35
|
Miczek KA, DiLeo A, Newman EL, Akdilek N, Covington HE. Neurobiological Bases of Alcohol Consumption After Social Stress. Curr Top Behav Neurosci 2022; 54:245-281. [PMID: 34964935 PMCID: PMC9698769 DOI: 10.1007/7854_2021_273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The urge to seek and consume excessive alcohol is intensified by prior experiences with social stress, and this cascade can be modeled under systematically controlled laboratory conditions in rodents and non-human primates. Adaptive coping with intermittent episodes of social defeat stress often transitions to maladaptive responses to traumatic continuous stress, and alcohol consumption may become part of coping responses. At the circuit level, the neural pathways subserving stress coping intersect with those for alcohol consumption. Increasingly discrete regions and connections within the prefrontal cortex, the ventral and dorsal striatum, thalamic and hypothalamic nuclei, tegmental areas as well as brain stem structures begin to be identified as critical for reacting to and coping with social stress while seeking and consuming alcohol. Several candidate molecules that modulate signals within these neural connections have been targeted in order to reduce excessive drinking and relapse. In spite of some early clinical failures, neuropeptides such as CRF, opioids, or oxytocin continue to be examined for their role in attenuating stress-escalated drinking. Recent work has focused on neural sites of action for peptides and steroids, most likely in neuroinflammatory processes as a result of interactive effects of episodic social stress and excessive alcohol seeking and drinking.
Collapse
Affiliation(s)
- Klaus A. Miczek
- Department of Psychology, Tufts University, Medford, MA, USA,Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Alyssa DiLeo
- Department of Neuroscience, Tufts University, Boston, MA, USA
| | - Emily L. Newman
- Department of Psychiatry, Harvard Medical School, Belmont, MA, USA
| | - Naz Akdilek
- Department of Psychology, Tufts University, Medford, MA, USA
| | | |
Collapse
|
36
|
Parise LF, Parise EM, Sial OK, Bolaños-Guzmán CA. Social Buffering is Dependent on Mutual Experience in Adolescent Male Mice Exposed to Social Defeat Stress. CHRONIC STRESS 2022; 6:24705470221111094. [PMID: 35874910 PMCID: PMC9305802 DOI: 10.1177/24705470221111094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022]
Abstract
Background Individuals who experience emotional, physical, or sexual abuse as children suffer from higher rates of major depressive disorder, drug abuse, and suicide. Early life interventions such as peer support groups can be beneficial to adolescents who experience trauma, suggesting that social support is important in facilitating rehabilitation and promoting resiliency to stress. Although there are some animal paradigms that can model how peer-peer interactions influence stress-reactivity, less is known about how individual stress experiences influence the effectiveness of social buffering. Methods The vicarious social defeat stress (VSDS) paradigm allows for the assessment of two different stress modalities, physical (PS) and emotional (ES) stress, which confer different levels of stress with similar biological and behavioral outcomes. Using a modified VSDS paradigm in which pairs of mice experience ES and PS together we can begin to evaluate how stress exposure influences the buffering efficacy of social relationships. Adolescent mice (postnatal day 35) were randomly combined into dyads and were allocated into either mutual experience or cohabitation pairs. Within each dyad, one mouse was assigned to the physically stressed (PS) condition and was repeatedly exposed to an aggressive CD1 mouse while the other mouse was designated as the partner. In the mutual experience dyads the partner mice witnessed the defeat bout (ES) while in the cohabitation dyads the partner was separated from the PS mouse and returned after the 10 min defeat bout was terminated (non-stressed). After 10 days of defeat, mice were tested in the social interaction test (SIT), the elevated plus maze (EPM), and the forced swim test (FST). Results PS-exposed mice in the cohabitation dyads, but not those in the mutual experience dyads, showed significantly more avoidance of a novel CD1 aggressor or c57BL/6 mouse, in the SIT. Surprisingly, both partner conditions showed avoidance to a CD1. Interestingly, non-stressed partner mice spent less time in the open arms of the EPM, suggesting increased anxiety; only PS-exposed mice in cohabitation dyads showed more time spent immobile in the FST, indicative of increased learned helplessness. Conclusions These data suggest that the efficacy of social buffering can be mediated by individual stress experience.
Collapse
Affiliation(s)
- Lyonna F. Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M. Parise
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Omar K. Sial
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, USA
| | | |
Collapse
|
37
|
Kim J, Pokharel K, Sandali M, Kim CS. Establishment of the Mouse Model of Social Avoidance Induced by Female-Directed Female Aggression. CHRONIC STRESS 2022; 6:24705470221129288. [PMID: 36187211 PMCID: PMC9523834 DOI: 10.1177/24705470221129288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
Background Most preclinical research on the effects of stress has been done on male subjects, even though women are more prone than men to experience stress-related problems. Chronic social defeat stress (CSDS) is a rodent model of psychosocial stress. However, this model has been challenged in female mouse studies since neither male nor female resident mice attack intruder females. A female-to-female CSDS model is needed to investigate the physiological and behavioral aspects. Methods The intruders were either male or female C57BL/6J mice, whereas the residents were male or ovariectomized (OVX) female CD-1 mice. The CD-1 aggressor mice had direct physical contact with the C57BL/6J mice for 10 min before initiating sensory contact with them for 24 h. Jump escape and freezing were evaluated during the social defeat of days 1 and 12. Experimental C57BL/6J mice underwent a social interaction test after suffering social defeat for 12 days. Results We found that the number of attack bites and attack latency had a significant negative correlation during the selection of aggressors. In the single-housed OVX mice, 34% of mice met the criterion of the selection of aggressors. However, single-housed OVX mice did not show sustained aggressive behavior (eg, attack bites) through the 12-day CSDS. As a result, we did not find susceptible mice during the social interaction test. In contrast, during the selection of aggressors, 42% of OVX mice housed with partners satisfied the criterion and displayed consistently aggressive behavior. CSDS produced susceptible (50%) and resilient (50%) phenotypes during the social interaction test. Notably, male and OVX female CD-1 mice housed with partners had similar amounts of attack bites and attack rates over the 12-day CSDS. Finally, we found that chronically socially defeated male and female mice displayed different coping behaviors (eg, active vs passive) with social defeat. Conclusions Our study demonstrates that OVX CD-1 mice housed with mates exhibited territorial aggression toward female intruders, producing susceptibility and resilience to social avoidance. Additionally, socially defeated male and female mice displayed different behavioral susceptibility to social defeat.
Collapse
Affiliation(s)
- Jiwon Kim
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kritika Pokharel
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Michael Sandali
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Chung Sub Kim
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
38
|
Wu PF, Han QQ, Chen FF, Shen TT, Li YH, Cao Y, Chen JG, Wang F. Erasing m 6A-dependent transcription signature of stress-sensitive genes triggers antidepressant actions. Neurobiol Stress 2021; 15:100390. [PMID: 34527794 PMCID: PMC8430387 DOI: 10.1016/j.ynstr.2021.100390] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/08/2021] [Accepted: 09/03/2021] [Indexed: 11/29/2022] Open
Abstract
Emerging evidence has shown that stress responsivity and psychiatric diseases are associated with alterations in N6-methyladenosine (m6A) mRNA epigenetic modifications. Fat mass and obesity-associated protein (FTO) is an m6A demethylase that has been linked to increased body mass and obesity. Here, we show that tricyclic antidepressants (TCAs) with weight-gain side effects, such as imipramine and amitriptyline, directly increased FTO expression and activated its epigenetic function in the ventral tegmental area (VTA). VTA-specific genetic disruption of FTO increased stress vulnerability and abolished the antidepressant activity of TCAs, whereas erasing m6A modification in the VTA by FTO overexpression or cycloleucine led to significant antidepressant activity. Mechanistically, both transcriptome sequencing and quantitative PCR revealed that overexpression of FTO in the VTA decreased the transcription of stress-related neuropeptides, such as cocaine- and amphetamine-regulated transcript peptide and urocortin, in the social defeat model, which was mimicked by imipramine, suggesting an m6A-dependent transcription mechanism of stress-related neuropeptides may underlie the responses to antidepressant. Collectively, our results demonstrate that inhibiting m6A-dependent transcription of stress-related genes may work as a novel antidepressant strategy and highlight a previously unrecognized activator of FTO-dependent epigenetic function that may be used for the treatment of other neurological diseases. TCAs erase m6A epigenetic modification by activating FTO. FTO mediates the antidepressant activity of TCAs. FTO in the VTA confers stress resistance. FTO in the VTA limits m6A-dependent transcription of stress-sensitive genes.
Collapse
Affiliation(s)
- Peng-Fei Wu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian-Qian Han
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Fu-Feng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Tian-Tian Shen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Yi-Heng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Yu Cao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei, 430030, China.,The Research Center for Depression, Tongji Medical College, Huazhong University of Science, 430030, Wuhan, China.,Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan City, Hubei, 430030, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan City, Hubei, 430030, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
39
|
Vitamin A5/X controls stress-adaptation and prevents depressive-like behaviors in a mouse model of chronic stress. Neurobiol Stress 2021; 15:100375. [PMID: 34401411 PMCID: PMC8355947 DOI: 10.1016/j.ynstr.2021.100375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 11/21/2022] Open
Abstract
9-cis-13,14-dihydroretinoic acid (9CDHRA), acts as an endogenous ligand of the retinoid X receptors (RXRs), and is an active form of a suggested new vitamin, vitamin A5/X. Nutritional-relevance of this pathway as well as its detailed role in vertebrate physiology, remain largely unknown. Since recent GWAS data and experimental studies associated RXR-mediated signaling with depression, we explored here the relevance of RXR and vitamin A5/X-mediated signaling in the control of stress adaptation and depressive-like behaviors in mice. We found that compromised availability of 9CDHRA in Rbp1−/− mice was associated with increased despair in the forced swim and anhedonia in the sucrose preference test. 9CDHRA similarly to synthetic RXR agonist, BMS649, normalized despair behaviors in Rbp1−/− but not Rxrγ−/− mice, supporting involvement of RXR signaling in anti-despair activity of these ligands. Importantly, similarly to BMS649, the 9CDHRA and its nutritional-precursor, 9-cis-13,14-dihydroretinol (vitamin A5/X alcohol), prevented development of depressive-like behaviors in mice exposed to chronic social defeat stress, revealing the beneficial role of RXRs and its endogenous ligand in stress adaptation process. These data point to the need for relevant nutritional, biochemical and pharmacological studies of this signaling pathway in human, both in physiological conditions and in pathologies of stress-related disorders.
Collapse
|
40
|
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: 54] [Impact Index Per Article: 13.5] [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
|
41
|
Phan ML, Liu TT, Vollbrecht MS, Mansour MH, Nikodijevic I, Jadav N, Patibanda N, Dang J, Shekaran G, Reisler RC, Kim WS, Zhou X, DiCicco-Bloom E, Samuels BA. Engrailed 2 deficiency and chronic stress alter avoidance and motivation behaviors. Behav Brain Res 2021; 413:113466. [PMID: 34271036 DOI: 10.1016/j.bbr.2021.113466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/18/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorder (ASD) is a pervasive neurodevelopmental disorder characterized by impairments in social interaction, cognition, and communication, as well as the presence of repetitive or stereotyped behaviors and interests. ASD is most often studied as a neurodevelopmental disease, but it is a lifelong disorder. Adults with ASD experience more stressful life events and greater perceived stress, and frequently have comorbid mood disorders such as anxiety and depression. It remains unclear whether adult exposure to chronic stress can exacerbate the behavioral and neurodevelopmental phenotypes associated with ASD. To address this issue, we first investigated whether adult male and female Engrailed-2 deficient (En2-KO, En2-/-) mice, which display behavioral disturbances in avoidance tasks and dysregulated monoaminergic neurotransmitter levels, also display impairments in instrumental behaviors associated with motivation, such as the progressive ratio task. We then exposed adult En2-KO mice to chronic environmental stress (CSDS, chronic social defeat stress), to determine if stress exacerbated the behavioral and neuroanatomical effects of En2 deletion. En2-/- mice showed impaired instrumental acquisition and significantly lower breakpoints in a progressive ratio test, demonstrating En2 deficiency decreases motivation to exert effort for reward. Furthermore, adult CSDS exposure increased avoidance behaviors in En2-KO mice. Interestingly, adult CSDS exposure also exacerbated the deleterious effects of En2 deficiency on forebrain-projecting monoaminergic fibers. Our findings thus suggest that adult exposure to stress may exacerbate behavioral and neuroanatomical phenotypes associated with developmental effects of genetic En2 deficiency.
Collapse
Affiliation(s)
- Mimi L Phan
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Tonia T Liu
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Mallory S Vollbrecht
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Mark H Mansour
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Ivana Nikodijevic
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Nikita Jadav
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Neeharika Patibanda
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Jenny Dang
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Gopna Shekaran
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Robert C Reisler
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Won S Kim
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA
| | - Xiaofeng Zhou
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, 08854, USA
| | - Benjamin A Samuels
- Behavioral and Systems Neuroscience Area, Department of Psychology, Rutgers University-New Brunswick, Piscataway, NJ, 08854, USA.
| |
Collapse
|
42
|
van Doeselaar L, Yang H, Bordes J, Brix L, Engelhardt C, Tang F, Schmidt MV. Chronic social defeat stress in female mice leads to sex-specific behavioral and neuroendocrine effects. Stress 2021; 24:168-180. [PMID: 33322989 DOI: 10.1080/10253890.2020.1864319] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the years, it has become increasingly clear that males and females respond differently towards environmental stressors, highlighting the importance of including both sexes when studying the effects of stress. This study aims to provide further insight into the detailed consequences of exposing female mice to 21 days of chronic social defeat stress (CSDS). We used a protocol that relies on the ability of odorants and pheromones in male urine to trigger male mouse aggressive behavior. Collected male C57Bl/6n urine was applied to female C57Bl/6n mice who were then attacked by a novel male CD1 mouse each day according to the CDSD protocol. Control females were pair-housed and handled daily. Physiological, neuroendocrine and behavioral changes were evaluated during the experiment. CSDS exposure resulted in number of physiological changes, such as body weight gain, enlarged adrenals and reduced thymus weight, exaggerated HPA-axis negative feedback and increased anxiety-like behavior. However, no generalized social avoidance behavior was observed. This study provides important insights in the physiological, neuroendocrine and behavioral responses of female mice to CSDS, which are partially dependent on estrous cycle stage. This protocol will allow direct comparison of male and female responses to CSDS and enable sex-specific study of mechanisms underlying individual stress resilience.Lay summaryIn this study we found that there are differences in the way that female and male mice respond towards chronic social stress conditions when it comes to behavior and hormonal changes.
Collapse
Affiliation(s)
- Lotte van Doeselaar
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Huanqing Yang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lea Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Clara Engelhardt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Fiona Tang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
43
|
Dieterich A, Floeder J, Stech K, Lee J, Srivastava P, Barker DJ, Samuels BA. Activation of Basolateral Amygdala to Nucleus Accumbens Projection Neurons Attenuates Chronic Corticosterone-Induced Behavioral Deficits in Male Mice. Front Behav Neurosci 2021; 15:643272. [PMID: 33716685 PMCID: PMC7943928 DOI: 10.3389/fnbeh.2021.643272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
The basolateral amygdala (BLA) is critical for reward behaviors via a projection to the nucleus accumbens (NAc). Specifically, BLA-NAc projections are involved in reinforcement learning, reward-seeking, sustained instrumental responding, and risk behaviors. However, it remains unclear whether chronic stress interacts with BLA-NAc projection neurons to result in maladaptive behaviors. Here we take a chemogenetic, projection-specific approach to clarify how NAc-projecting BLA neurons affect avoidance, reward, and feeding behaviors in male mice. Then, we examine whether chemogenetic activation of NAc-projecting BLA neurons attenuates the maladaptive effects of chronic corticosterone (CORT) administration on these behaviors. CORT mimics the behavioral and neural effects of chronic stress exposure. We found a nuanced role of BLA-NAc neurons in mediating reward behaviors. Surprisingly, activation of BLA-NAc projections rescues CORT-induced deficits in the novelty suppressed feeding, a behavior typically associated with avoidance. Activation of BLA-NAc neurons also increases instrumental reward-seeking without affecting free-feeding in chronic CORT mice. Taken together, these data suggest that NAc-projecting BLA neurons are involved in chronic CORT-induced maladaptive reward and motivation behaviors.
Collapse
Affiliation(s)
- Andrew Dieterich
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Joseph Floeder
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Karina Stech
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Jay Lee
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Prachi Srivastava
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - David J. Barker
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Benjamin A. Samuels
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Psychology, Behavioral and Systems Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
44
|
Chronic non-discriminatory social defeat stress reduces effort-related motivated behaviors in male and female mice. Transl Psychiatry 2021; 11:125. [PMID: 33589585 PMCID: PMC7884699 DOI: 10.1038/s41398-021-01250-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/07/2021] [Accepted: 01/20/2021] [Indexed: 01/31/2023] Open
Abstract
Reward and motivation deficits are prominent symptoms in many mood disorders, including depression. Similar reward and effort-related choice behavioral tasks can be used to study aspects of motivation in both rodents and humans. Chronic stress can precipitate mood disorders in humans and maladaptive reward and motivation behaviors in male rodents. However, while depression is more prevalent in women, there is relatively little known about whether chronic stress elicits maladaptive behaviors in female rodents in effort-related motivated tasks and whether there are any behavioral sex differences. Chronic nondiscriminatory social defeat stress (CNSDS) is a variation of chronic social defeat stress that is effective in both male and female mice. We hypothesized that CNSDS would reduce effort-related motivated and reward behaviors, including reducing sensitivity to a devalued outcome, reducing breakpoint in progressive ratio, and shifting effort-related choice behavior. Separate cohorts of adult male and female C57BL/6 J mice were divided into Control or CNSDS groups, exposed to the 10-day CNSDS paradigm, and then trained and tested in instrumental reward or effort-related behaviors. CNSDS reduced motivation to lever press in progressive ratio and shifted effort-related choice behavior from a high reward to a more easily attainable low reward in both sexes. CNSDS caused more nuanced impairments in outcome devaluation. Taken together, CNSDS induces maladaptive shifts in effort-related choice and reduces motivated lever pressing in both sexes.
Collapse
|
45
|
Dentate gyrus activin signaling mediates the antidepressant response. Transl Psychiatry 2021; 11:7. [PMID: 33414389 PMCID: PMC7791138 DOI: 10.1038/s41398-020-01156-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Antidepressants that target monoaminergic systems, such as selective serotonin reuptake inhibitors (SSRIs), are widely used to treat neuropsychiatric disorders including major depressive disorder, several anxiety disorders, and obsessive-compulsive disorder. However, these treatments are not ideal because only a subset of patients achieve remission. The reasons why some individuals remit to antidepressant treatments while others do not are unknown. Here, we developed a paradigm to assess antidepressant treatment resistance in mice. Exposure of male C57BL/6J mice to either chronic corticosterone administration or chronic social defeat stress induces maladaptive affective behaviors. Subsequent chronic treatment with the SSRI fluoxetine reverses these maladaptive affective behavioral changes in some, but not all, of the mice, permitting stratification into persistent responders and non-responders to fluoxetine. We found several differences in expression of Activin signaling-related genes between responders and non-responders in the dentate gyrus (DG), a region that is critical for the beneficial behavioral effects of fluoxetine. Enhancement of Activin signaling in the DG converted behavioral non-responders into responders to fluoxetine treatment more effectively than commonly used second-line antidepressant treatments, while inhibition of Activin signaling in the DG converted responders into non-responders. Taken together, these results demonstrate that the behavioral response to fluoxetine can be bidirectionally modified via targeted manipulations of the DG and suggest that molecular- and neural circuit-based modulations of DG may provide a new therapeutic avenue for more effective antidepressant treatments.
Collapse
|
46
|
Maita I, Bazer A, Blackford JU, Samuels BA. Functional anatomy of the bed nucleus of the stria terminalis-hypothalamus neural circuitry: Implications for valence surveillance, addiction, feeding, and social behaviors. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:403-418. [PMID: 34225978 DOI: 10.1016/b978-0-12-819975-6.00026-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is a medial basal forebrain structure that modulates the hypothalamo-pituitary-adrenal (HPA) axis. The heterogeneous subnuclei of the BNST integrate inputs from mood and reward-related areas and send direct inhibitory projections to the hypothalamus. The connections between the BNST and hypothalamus are conserved across species, promote activation of the HPA axis, and can increase avoidance of aversive environments, which is historically associated with anxiety behaviors. However, BNST-hypothalamus circuitry is also implicated in motivated behaviors, drug seeking, feeding, and sexual behavior. These complex and diverse roles, as well its sexual dimorphism, indicate that the BNST-hypothalamus circuitry is an essential component of the neural circuitry that may underlie various psychiatric diseases, ranging from anorexia to anxiety to addiction. The following review is a cross-species exploration of BNST-hypothalamus circuitry. First, we describe the BNST subnuclei, microcircuitry and complex reciprocal connections with the hypothalamus. We will then discuss the behavioral functions of BNST-hypothalamus circuitry, including valence surveillance, addiction, feeding, and social behavior. Finally, we will address sex differences in morphology and function of the BNST and hypothalamus.
Collapse
Affiliation(s)
- Isabella Maita
- Department of Psychology, Rutgers University, Piscataway, NJ, United States
| | - Allyson Bazer
- Department of Psychology, Rutgers University, Piscataway, NJ, United States
| | - Jennifer Urbano Blackford
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, United States; Research Health Scientist, Tennessee Valley HealthCare System, US Department of Veterans Affairs, Nashville, TN, United States
| | | |
Collapse
|
47
|
Takahashi A. Toward Understanding the Sex Differences in the Biological Mechanism of Social Stress in Mouse Models. Front Psychiatry 2021; 12:644161. [PMID: 33664686 PMCID: PMC7921148 DOI: 10.3389/fpsyt.2021.644161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Significant sex differences in terms of prevalence, symptomatic profiles, severity, and comorbidities of psychiatric disorders are quite common. Women have been shown to be more vulnerable to stress and are nearly twice as likely as men to develop stress-related disorders such as depression and anxiety. Therefore, understanding sex differences with respect to the neurobiological mechanisms underlying stress-related disorders is important for developing more efficient pharmacological interventions for women. However, most preclinical studies on stress-related disorders have focused heavily on male rodents. Here, recent developments in the study of repeated social defeat stress models in female mice are summarized. Our findings suggest that a variety of factors need to be considered when employing this model.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
48
|
Adult-Born Neurons in the Hippocampus Are Essential for Social Memory Maintenance. eNeuro 2020; 7:ENEURO.0182-20.2020. [PMID: 33060182 PMCID: PMC7768285 DOI: 10.1523/eneuro.0182-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023] Open
Abstract
Throughout adulthood, the dentate gyrus continues to produce new granule cells, which integrate into the hippocampal circuitry. New neurons have been linked to several known functions of the hippocampus, including learning and memory, anxiety and stress regulation, and social behavior. We explored whether transgenic reduction of adult-born neurons in mice would impair social memory and the formation of social dominance hierarchies. We used a conditional transgenic mouse strain [thymidine kinase (TK) mice] that selectively reduces adult neurogenesis by treatment with the antiviral drug valganciclovir (VGCV). TK mice treated with VGCV were unable to recognize conspecifics as familiar 24 h after initial exposure. We then explored whether reducing new neurons completely impaired their ability to acquire or retrieve a social memory and found that TK mice treated with VGCV were able to perform at control levels when the time between exposure (acquisition) and reexposure (retrieval) was brief. We next explored whether adult-born neurons are involved in dominance hierarchy formation by analyzing their home cage behavior as well as their performance in the tube test, a social hierarchy test, and did not find any consistent alterations in behavior between control and TK mice treated with VGCV. These data suggest that adult neurogenesis is essential for social memory maintenance, but not for acquisition nor retrieval over a short time frame, with no effect on social dominance hierarchy. Future work is needed to explore whether the influence of new neurons on social memory is mediated through connections with the CA2, an area involved in social recognition.
Collapse
|
49
|
Dieterich A, Yohn CN, Samuels BA. Chronic Stress Shifts Effort-Related Choice Behavior in a Y-Maze Barrier Task in Mice. J Vis Exp 2020. [PMID: 32865538 PMCID: PMC7646533 DOI: 10.3791/61548] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Mood disorders, including major depressive disorder, can be precipitated by chronic stress. The Y-maze barrier task is an effort-related choice test that measures motivation to expend effort and obtain reward. In mice, chronic stress exposure significantly impacts motivation to work for a higher value reward when a lesser value reward is freely available compared to unstressed mice. Here we describe the chronic corticosterone administration paradigm, which produces a shift in effortful responding in the Y-maze barrier task. In the Y-maze task, one arm contains 4 food pellets, while the other arm contains only 2 pellets. After mice learn to select the high reward arm, barriers with progressively increasing height are then introduced into the high reward arm over multiple test sessions. Unfortunately, most chronic stress paradigms (including corticosterone and social defeat) were developed in male mice and are less effective in female mice. Therefore, we also discuss chronic non-discriminatory social defeat stress (CNSDS), a stress paradigm we developed that is effective in both male and female mice. Repeating results with multiple distinct chronic stressors in male and female mice combined with increased usage of translationally relevant behavior tasks will help to advance the understanding of how chronic stress can precipitate mood disorders.
Collapse
Affiliation(s)
- Andrew Dieterich
- Department of Psychology, Behavioral and Systems Neuroscience Area, The State University of New Jersey; Graduate Program in Neuroscience, Rutgers, The State University of New Jersey;
| | - Christine N Yohn
- Department of Psychology, Behavioral and Systems Neuroscience Area, The State University of New Jersey
| | - Benjamin Adam Samuels
- Department of Psychology, Behavioral and Systems Neuroscience Area, The State University of New Jersey; Graduate Program in Neuroscience, Rutgers, The State University of New Jersey;
| |
Collapse
|
50
|
Depression and Social Defeat Stress Are Associated with Inhibitory Synaptic Changes in the Nucleus Accumbens. J Neurosci 2020; 40:6228-6233. [PMID: 32561672 DOI: 10.1523/jneurosci.2568-19.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/28/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic stress in both humans and rodents induces a robust downregulation of neuroligin-2, a key component of the inhibitory synapse, in the NAc that modifies behavioral coping mechanisms and stress resiliency in mice. Here we extend this observation by examining the role of two other inhibitory synapse constituents, vesicular GABA transporter (vGAT) and gephyrin, in the NAc of male mice that underwent chronic social defeat stress (CSDS) and in patients with major depressive disorder (MDD). We first performed transcriptional profiling of vGAT and gephyrin in postmortem NAc samples from a cohort of healthy controls, medicated, and nonmedicated MDD patients. In parallel, we conducted whole-cell electrophysiology recordings in the NAc of stress-susceptible and stress-resilient male mice following 10 d of CSDS. Finally, we used immunohistochemistry to analyze protein levels of vGAT and gephyrin in the NAc of mice after CSDS. We found that decreased vGAT and gephyrin mRNA in the NAc of nonmedicated MDD patients is paralleled by decreased inhibitory synapse markers and decreased frequency of mini inhibitory postsynaptic currents (mIPSC) in the NAc of susceptible mice, indicating a reduction in the number of NAc inhibitory synapses that is correlated with depression-like behavior. Overall, these findings suggest a common state of reduced inhibitory tone in the NAc in depression and stress susceptibility.SIGNIFICANCE STATEMENT Existing studies focus on excitatory synaptic changes after social stress, although little is known about stress-induced inhibitory synaptic plasticity and its relevance for neuropsychiatric disease. These results extend our previous findings on the critical role of impaired inhibitory tone in the NAc following stress and provide new neuropathological evidence for reduced levels of inhibitory synaptic markers in human NAc from nonmedicated major depressive disorder patients. This finding is corroborated in stress-susceptible male mice that have undergone chronic social defeat stress, a mouse model of depression, at both the level of synaptic function and protein expression. These data support the hypothesis that reduced inhibitory synaptic transmission within the NAc plays a critical role in the stress response.
Collapse
|