1
|
Cai A, Zheng D, Xu F, Wang F, Sajikumar S, Wang J. Variations of Aberrant Volume, Activity, and Network Connectivity of Hippocampus in Adolescent Male Rats Exposed to Juvenile Stress. Brain Sci 2025; 15:284. [PMID: 40149805 PMCID: PMC11940772 DOI: 10.3390/brainsci15030284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Childhood is a crucial period for brain development, and short-term juvenile stress has demonstrated long-lasting effects on cognitive and cellular functions in the hippocampus. However, the influence of such stress on the brain's overall network remains unclear. METHODS In this study, we employed functional magnetic resonance imaging (fMRI) to explore the effects of transient wild stress on juvenile male rats. Pregnant rats were purchased and housed in a specific pathogen-free (SPF) environment, with pups separated by sex on postnatal day 21 (PD21). From PD27 to PD29, male rats were subjected to transient wild stress, which included forced swimming, elevated platform exposure, and restraint stress. Following stress exposure, all animals were carefully maintained and scanned at 42 days of age (PD42) using fMRI. Structural analysis was performed using voxel-based morphometry (VBM) to assess changes in gray matter volume, while functional activity was evaluated through regional homogeneity (ReHo) and voxel-wise functional connectivity. RESULTS The results showed significant reductions in gray matter volume in several brain regions in the stress group, including the periaqueductal gray (PAG), entorhinal cortex (Ent), and dentate gyrus (DG). In terms of functional activity, cortical regions, particularly the primary somatosensory areas, exhibited decreased activity, whereas increased activity was observed in the PAG, DG, and medulla. Furthermore, functional connectivity analysis revealed a significant reduction in connectivity between the DG and entorhinal cortex, while the DG-PAG connectivity was significantly enhanced. CONCLUSIONS These findings suggest that juvenile stress leads to profound alterations in both brain structure and function, potentially disrupting emotional regulation and memory processing by affecting the development and connectivity of key brain regions.
Collapse
Affiliation(s)
- Aoling Cai
- Department of Radiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; (A.C.)
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210000, China
| | - Danhao Zheng
- Department of Radiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; (A.C.)
- Key Laboratory of Magnetic Resonance in Biological Systems, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fanyong Xu
- Department of Radiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; (A.C.)
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210000, China
- Department of Mental Health, School of Public Health, Nanjing Medical University, Nanjing 210000, China
| | - Sreedharan Sajikumar
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
- Life Sciences Institute Neurobiology Programme, Centre for Life Sciences, National University of Singapore, Singapore 117456, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Jie Wang
- Department of Radiology, Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai 201600, China; (A.C.)
| |
Collapse
|
2
|
Lupinsky D, Nasseef MT, Parent C, Craig K, Diorio J, Zhang TY, Meaney MJ. Resting-state fMRI reveals altered functional connectivity associated with resilience and susceptibility to chronic social defeat stress in mouse brain. Mol Psychiatry 2025:10.1038/s41380-025-02897-2. [PMID: 39984680 DOI: 10.1038/s41380-025-02897-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 02/23/2025]
Abstract
Chronic stress is a causal antecedent condition for major depressive disorder and associates with altered patterns of neural connectivity. There are nevertheless important individual differences in susceptibility to chronic stress. How functional connectivity (FC) amongst interconnected, depression-related brain regions associates with resilience and susceptibility to chronic stress is largely unknown. We used resting-state functional magnetic resonance imaging (rs-fMRI) to examine FC between established depression-related regions in susceptible (SUS) and resilient (RES) adult mice following chronic social defeat stress (CSDS). Seed-seed FC analysis revealed that the ventral dentate gyrus (vDG) exhibited the greatest number of FC group differences with other stress-related limbic brain regions. SUS mice showed greater FC between the vDG and subcortical regions compared to both control (CON) or RES groups. Whole brain vDG seed-voxel analysis supported seed-seed findings in SUS mice but also indicated significantly decreased FC between the vDG and anterior cingulate area compared to CON mice. Interestingly, RES mice exhibited enhanced FC between the vDG and anterior cingulate area compared to SUS mice. Moreover, RES mice showed greater FC between the infralimbic prefrontal cortex and the nucleus accumbens shell compared to CON mice. These findings indicate unique differences in FC patterns in phenotypically distinct SUS and RES mice that could represent a neurobiological basis for depression, anxiety, and negative-coping behaviors that are associated with exposure to chronic stress.
Collapse
Affiliation(s)
- Derek Lupinsky
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
| | - Md Taufiq Nasseef
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
- Department of Mathematics, College of Science and Humanity Studies, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Carine Parent
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
| | - Kelly Craig
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Josie Diorio
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Tie-Yuan Zhang
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada.
| | - Michael J Meaney
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada.
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Brain-Body Initiative, Agency for Science, Technology & Research, Singapore, Singapore.
| |
Collapse
|
3
|
Hamden JE, Salehzadeh M, Bajaj H, Li MX, Soma KK. Lipopolysaccharide differentially alters systemic and brain glucocorticoid levels in neonatal and adult mice. J Neuroendocrinol 2025; 37:e13481. [PMID: 39694531 PMCID: PMC11791005 DOI: 10.1111/jne.13481] [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: 04/01/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024]
Abstract
Glucocorticoids (GCs) are secreted by the adrenal glands and increase in response to stressors (e.g., infection). The brain regulates local GC levels via GC synthesis, regeneration and/or metabolism. Little is known about local GC regulation within discrete brain regions at baseline or in response to stress. We treated male and female C57BL/6J mice at postnatal day 5 (PND5) or PND90 with lipopolysaccharide (LPS; 50 μg/kg bw i.p.) or vehicle and collected blood and brain after 4 h. We microdissected the prefrontal cortex, hippocampus, hypothalamus and amygdala. We measured seven steroids, including corticosterone, via liquid chromatography-tandem mass spectrometry and measured transcripts for key steroidogenic enzymes (Cyp11b1, Hsd11b1, Hsd11b2) via qPCR. At both ages, LPS increased GC levels in blood and all brain regions; however, the increases were much greater at PND90 than at PND5. Interestingly, PND5 corticosterone levels were lower in prefrontal cortex than in blood, but higher in amygdala than in blood. These changes in corticosterone levels align with local changes in steroidogenic enzyme expression, demonstrating robust regional heterogeneity and a possible mechanism for the region-specific effects of early-life stress. In contrast, PND90 corticosterone levels were lower in all brain regions than in blood and similar among regions, and steroidogenic enzyme mRNA levels were generally not affected by LPS. Together, these data indicate that local GC levels within discrete brain regions are more heterogeneous at baseline and in response to LPS at PND5 than at PND90, as a result of increased local GC production and metabolism in the neonatal brain.
Collapse
Affiliation(s)
- Jordan E. Hamden
- Department of Biochemistry and Molecular BiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Melody Salehzadeh
- Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of ZoologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Hitasha Bajaj
- Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of PsychologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Graduate Program in NeuroscienceUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Michael X. Li
- Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kiran K. Soma
- Djavad Mowafaghian Centre for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of ZoologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of PsychologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Graduate Program in NeuroscienceUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
4
|
Wang ZY, Hu SX, Lu J, Shang W, Chen T, Zhang RT. Dimensional early life adversity and anxiety symptoms: A network analysis and longitudinal study. CHILD ABUSE & NEGLECT 2025; 160:107201. [PMID: 39731875 DOI: 10.1016/j.chiabu.2024.107201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/13/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Accumulating literature has found a close relation between early life adversity (ELA) and anxiety. However, previous studies did not rule out the high co-occurrence of different types of ELA when exploring the association of ELA and anxiety. In the present study, we carried out network analysis based on a cross-sectional sample and longitudinal sample to investigate the relationship between ELA and anxiety symptoms in non-clinical populations over time. METHODS Online advertisement was carried out to recruit participants. The cross-sectional sample included 871 Chinese participants (Mage = 19.11, SDage = 1.57), and the longitudinal sample involved 440 Chinese participants (Mage = 18.93, SDage = 0.75). Three dimensions of ELA were assessed. The Threat/Harm dimension was assessed by subscales of physical abuse, emotional abuse, and sexual abuse of Childhood Trauma Questionnaire (CTQ). The Deprivation dimension of ELA was measured by subscales of physical neglect, and emotional neglect of CTQ. The Unpredictability dimension of ELA was evaluated by the Childhood Unpredictability scale. Anxiety symptoms were captured by the Generalized Anxiety Disorder-7 (GAD-7). Regularized partial correlation networks were constructed, and the expected influence (EI) as well as predictability of each node were calculated. Stability within the network was tested and the network comparison test was conducted to examine the difference between the cross-sectional network and the longitudinal network. RESULTS The cross-sectional network was relatively tight, and nodes within the dimension of ELA clustered together. Childhood unpredictability and emotional abuse revealed stronger associations with anxiety symptoms than other ELAs. Emotional abuse showed the highest EI in the network. These findings were replicated in the longitudinal network. The network comparison test indicated no significant difference between the cross-sectional network and the longitudinal network. CONCLUSION Childhood unpredictability and emotional abuse were strong predictors of anxiety symptoms, and the prediction was reliable and persistent. Reduce the experience of childhood emotional abuse can serve as a target to prevent anxiety.
Collapse
Affiliation(s)
- Zhao-Ying Wang
- Department of Psychology, Hunan Normal University, Changsha, China; Cognition and Human Behavior Key Laboratory of Hunan Province, Hunan Normal University, Changsha, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, China; Center for Mind & Brain Sciences, Hunan Normal University, Changsha, China
| | - Shi-Xiong Hu
- Shuda College, Hunan Normal University, Changsha, China
| | - Jian Lu
- Counseling and Psychological Services, Hunan Normal University, Changsha, China
| | - Wen Shang
- Institute of Marxism, Central South University, Changsha, China
| | - Tao Chen
- The University of Sydney, Brain and Mind Centre, Sydney, Australia
| | - Rui-Ting Zhang
- Department of Psychology, Hunan Normal University, Changsha, China; Cognition and Human Behavior Key Laboratory of Hunan Province, Hunan Normal University, Changsha, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, China; Center for Mind & Brain Sciences, Hunan Normal University, Changsha, China.
| |
Collapse
|
5
|
Ahmed S, Polis B, Jamwal S, Sanganahalli BG, MacDowell Kaswan Z, Islam R, Kim D, Bowers C, Giuliano L, Biederer T, Hyder F, Kaffman A. Transient impairment in microglial function causes sex-specific deficits in synaptic maturity and hippocampal function in mice exposed to early adversity. Brain Behav Immun 2024; 122:95-109. [PMID: 39134183 PMCID: PMC11402597 DOI: 10.1016/j.bbi.2024.08.010] [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: 02/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Abnormal development and function of the hippocampus are two of the most consistent findings in humans and rodents exposed to early-life adversity (ELA), with males often being more affected than females. Using the limited bedding (LB) paradigm as a rodent model of ELA, we found that male adolescent mice that had been exposed to LB exhibit significant deficits in contextual fear conditioning and synaptic connectivity in the hippocampus, which are not observed in females. This is linked to altered developmental refinement of connectivity, with LB severely impairing microglial-mediated synaptic pruning in the hippocampus of male and female pups on postnatal day 17 (P17), but not in adolescent P33 mice when levels of synaptic engulfment by microglia are substantially lower. Since the rodent hippocampus undergoes intense synaptic pruning during the second and third weeks of life, we investigated whether microglia are required for the synaptic and behavioral aberrations observed in adolescent LB mice. Indeed, transient ablation of microglia from P13-21 in normally developing mice caused sex-specific behavioral and synaptic abnormalities similar to those observed in adolescent LB mice. Furthermore, chemogenetic activation of microglia during the same period reversed the microglial-mediated phagocytic deficits at P17 and restored normal contextual fear conditioning and synaptic connectivity in adolescent LB male mice. Our data support an additional contribution of astrocytes in the sex-specific effects of LB, with increased expression of the membrane receptor MEGF10 and enhanced synaptic engulfment in hippocampal astrocytes of 17-day-old LB females, but not in LB male littermates. These findings suggest a potential compensatory mechanism that may explain the relative resilience of LB females. Collectively, our study highlights a novel role for glial cells in mediating sex-specific hippocampal deficits in a mouse model of ELA.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Basavaraju G Sanganahalli
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
| | - Zoe MacDowell Kaswan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Dana Kim
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, 100 College Street, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA.
| |
Collapse
|
6
|
Li H, Fang Y, Wang D, Shi B, Thompson GJ. Impaired brain glucose metabolism in glucagon-like peptide-1 receptor knockout mice. Nutr Diabetes 2024; 14:86. [PMID: 39389952 PMCID: PMC11466955 DOI: 10.1038/s41387-024-00343-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: 01/26/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Quantitative mapping of the brain's metabolism is a critical tool in studying and diagnosing many conditions, from obesity to neurodegenerative diseases. In particular, noninvasive approaches are urgently required. Recently, there have been promising drug development approaches for the treatment of disorders related to glucose metabolism in the brain and, therefore, against obesity-associated diseases. One of the most important drug targets to emerge has been the Glucagon-like peptide-1 (GLP-1) and its receptor (GLP-1R). GLP and GLP-1R play an important role in regulating blood sugar and maintaining energy homeostasis. However, the macroscopic effects on brain metabolism and function due to the presence of GLP-1R are unclear. METHODS To explore the physiological role of GLP-1R in mouse brain glucose metabolism, and its relationship to brain function, we used three methods. We used deuterium magnetic resonance spectroscopy (DMRS) to provide quantitative information about metabolic flux, fluorodeoxyglucose positron emission tomography (FDG-PET) to measure brain glucose metabolism, and resting state-functional MRI (rs-fMRI) to measure brain functional connectivity. We used these methods in both mice with complete GLP-1R knockout (GLP-1R KO) and wild-type C57BL/6N (WT) mice. RESULTS The metabolic rate of GLP-1R KO mice was significantly slower than that of WT mice (p = 0.0345, WT mice 0.02335 ± 0.057 mM/min, GLP-1R KO mice 0.01998 ± 0.07 mM/min). Quantification of the mean [18F]FDG signal in the whole brain also showed significantly reduced glucose uptake in GLP-1R KO mice versus control mice (p = 0.0314). Observing rs-fMRI, the functional brain connectivity in GLP-1R KO mice was significantly lower than that in the WT group (p = 0.0032 for gFCD, p = 0.0002 for whole-brain correlation, p < 0.0001 for ALFF). CONCLUSIONS GLP-1R KO mice exhibit impaired brain glucose metabolism to high doses of exogenous glucose, and they also have reduced functional connectivity. This suggests that the GLP-1R KO mouse model may serve as a model for correlated metabolic and functional connectivity loss.
Collapse
Affiliation(s)
- Hui Li
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Yujiao Fang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Da Wang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bowen Shi
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | | |
Collapse
|
7
|
Gong X, Tian T, Xu J, Qin S, Lin D. Psychophysiological Factors Moderate Amygdala-Prefrontal Connectivity Linked to Perceived Peer Victimization and Depressive Symptoms in Preadolescent Migrant Children. Depress Anxiety 2024; 2024:5596651. [PMID: 40226685 PMCID: PMC11919204 DOI: 10.1155/2024/5596651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 04/15/2025] Open
Abstract
Background: As a sense of an intense stressor, perceived peer victimization can cause adverse effects on mental health, like depressive symptoms. Yet, little is known about the neurobiological mechanisms underlying how perceived peer victimization causes and maintains depressive symptoms in preadolescence. Methods: Here we investigate the effects of peer victimization on amygdala subregional functional connectivity in 101 preadolescent migrant children, and their relations to depressive symptoms and potential protective factors of self-esteem and daily cortisol. Further control analyses were conducted to verify whether there are any specific effects in migrant children compared to 54 age-matched preadolescent children from nonmigrant background. Results: Children with higher perceived peer victimization exhibited greater intrinsic functional connectivity of the amygdala with the middle frontal gyrus extending into the superior frontal gyrus. Perceived peer victimization could account for an indirect association between amygdala hyperconnectivity and depressive symptoms. Moderated mediation analyses revealed that basolateral amygdala connectivity with the superior frontal gyrus acted as a neural marker linking peer victimization and greater risk for depressive symptoms only in preadolescent children with low self-esteem or low daily cortisol. Conclusions: These findings suggest that considering neurobiological vulnerability and psychophysiological factors may gain a nuanced understanding of the adverse effects of perceived peer victimization on depressive symptoms, a risk for internalizing pathology. This study could inform personalized intervention strategies to prevent or ameliorate depressive symptoms in this disadvantaged population.
Collapse
Affiliation(s)
- Xinyu Gong
- Institute of Developmental Psychology, Beijing Normal University, Beijing, China
| | - Ting Tian
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Jiahua Xu
- Psychiatry Research Center, Beijing Huilongguan Hospital, Peking University Huilongguan Clinical Medical School, Beijing, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Danhua Lin
- Institute of Developmental Psychology, Beijing Normal University, Beijing, China
| |
Collapse
|
8
|
Sanguino-Gómez J, Huijgens S, den Hartog M, Schenk IJM, Kluck W, Versluis TD, Krugers HJ. Neural correlates of learning and memory are altered by early-life stress. Neurobiol Learn Mem 2024; 213:107952. [PMID: 38906243 DOI: 10.1016/j.nlm.2024.107952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/12/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
The ability to learn and remember, which is fundamental for behavioral adaptation, is susceptible to stressful experiences during the early postnatal period, such as abnormal levels of maternal care. The exact mechanisms underlying these effects still remain elusive. This study examined whether early life stress (ELS) alters memory and brain activation patterns in male mice. Therefore, we examined the expression of the immediate early genes (IEGs) c-Fos and Arc in the dentate gyrus (DG) and basolateral amygdala (BLA) after training and memory retrieval in a fear conditioning task. Furthermore, we examined the potential of RU38486 (RU486), a glucocorticoid receptor antagonist, to mitigate ELS-induced memory deficits by blocking stress signalling during adolescence. Arc::dVenus reporter mice, which allow investigating experience-dependent expression of the immediate early gene Arc also at more remote time points, were exposed to ELS by housing dams and offspring with limited bedding and nesting material (LBN) between postnatal days (PND) 2-9 and trained in a fear conditioning task at adult age. We found that ELS reduced both fear acquisition and contextual memory retrieval. RU486 did not prevent these effects. ELS reduced the number of Arc::dVenus+ cells in DG and BLA after training, while the number of c-Fos+ cells were left unaffected. After memory retrieval, ELS decreased c-Fos+ cells in the ventral DG and BLA. ELS also altered the colocalization of c-Fos+ cells with Arc::dVenus+ cells in the ventral DG, possibly indicating impaired engram allocation in the ventral DG after memory retrieval. In conclusion, this study shows that ELS alters neuronal activation patterns after fear acquisition and retrieval, which may provide mechanistic insights into enduring impact of ELS on the processing of fear memories, possibly via changes in cell (co-) activation and engram cell allocation.
Collapse
Affiliation(s)
| | - Stefan Huijgens
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Maxine den Hartog
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Inim J M Schenk
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Wenya Kluck
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Tamara D Versluis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Ahmed S, Polis B, Kaffman A. Microglia: The Drunken Gardeners of Early Adversity. Biomolecules 2024; 14:964. [PMID: 39199352 PMCID: PMC11353196 DOI: 10.3390/biom14080964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Early life adversity (ELA) is a heterogeneous group of negative childhood experiences that can lead to abnormal brain development and more severe psychiatric, neurological, and medical conditions in adulthood. According to the immune hypothesis, ELA leads to an abnormal immune response characterized by high levels of inflammatory cytokines. This abnormal immune response contributes to more severe negative health outcomes and a refractory response to treatment in individuals with a history of ELA. Here, we examine this hypothesis in the context of recent rodent studies that focus on the impact of ELA on microglia, the resident immune cells in the brain. We review recent progress in our ability to mechanistically link molecular alterations in microglial function during a critical period of development with changes in synaptic connectivity, cognition, and stress reactivity later in life. We also examine recent research showing that ELA induces long-term alterations in microglial inflammatory response to "secondary hits" such as traumatic brain injury, substance use, and exposure to additional stress in adulthood. We conclude with a discussion on future directions and unresolved questions regarding the signals that modify microglial function and the clinical significance of rodent studies for humans.
Collapse
Affiliation(s)
| | | | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA; (S.A.); (B.P.)
| |
Collapse
|
10
|
Cattaneo A, Begni V, Zonca V, Riva MA. Early life adversities, psychopathologies and novel pharmacological strategies. Pharmacol Ther 2024; 260:108686. [PMID: 38969307 DOI: 10.1016/j.pharmthera.2024.108686] [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: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Exposure to adversities during early life stages (early life adversities - ELA), ranging from pregnancy to adolescence, represents a major risk factor for the vulnerability to mental disorders. Hence, it is important to understand the molecular and functional underpinning of such relationship, in order to develop strategies aimed at reducing the psychopathologic burden associated with ELA, which may eventually lead to a significant improvement in clinical practice. In this review, we will initially recapitulate clinical and preclinical evidence supporting the link between ELA and psychopathology and we will primarily discuss the main biological mechanisms that have been described as potential mediators of the effects of ELA on the psychopathologic risk, including the role for genetic factors as well as sex differences. The knowledge emerging from these studies may be instrumental for the development of novel therapeutic strategies aimed not only at correcting the deficits that emerge from ELA exposure, but also in preventing the manifestation of a full-blown psychopathologic condition. With this respect, we will specifically focus on adolescence as a key time frame for disease onset as well as for early therapeutic intervention. We believe that incorporating clinical and preclinical research data in the context of early life adversities can be instrumental to elucidate the mechanisms contributing to the risk for psychopathology or that may promote resilience. This will ultimately allow the identification of 'at risk' individuals who may benefit from specific forms of interventions that, by interfering with disease trajectories, could result in more benign clinical outcomes.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Valentina Zonca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
11
|
Islam R, White JD, Arefin TM, Mehta S, Liu X, Polis B, Giuliano L, Ahmed S, Bowers C, Zhang J, Kaffman A. Early adversity causes sex-specific deficits in perforant pathway connectivity and contextual memory in adolescent mice. Biol Sex Differ 2024; 15:39. [PMID: 38715106 PMCID: PMC11075329 DOI: 10.1186/s13293-024-00616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Early life adversity impairs hippocampal development and function across diverse species. While initial evidence indicated potential variations between males and females, further research is required to validate these observations and better understand the underlying mechanisms contributing to these sex differences. Furthermore, most of the preclinical work in rodents was performed in adult males, with only few studies examining sex differences during adolescence when such differences appear more pronounced. To address these concerns, we investigated the impact of limited bedding (LB), a mouse model of early adversity, on hippocampal development in prepubescent and adolescent male and female mice. METHODS RNA sequencing, confocal microscopy, and electron microscopy were used to evaluate the impact of LB and sex on hippocampal development in prepubescent postnatal day 17 (P17) mice. Additional studies were conducted on adolescent mice aged P29-36, which included contextual fear conditioning, retrograde tracing, and ex vivo diffusion magnetic resonance imaging (dMRI). RESULTS More severe deficits in axonal innervation and myelination were found in the perforant pathway of prepubescent and adolescent LB males compared to LB female littermates. These sex differences were due to a failure of reelin-positive neurons located in the lateral entorhinal cortex (LEC) to innervate the dorsal hippocampus via the perforant pathway in males, but not LB females, and were strongly correlated with deficits in contextual fear conditioning. CONCLUSIONS LB impairs the capacity of reelin-positive cells located in the LEC to project and innervate the dorsal hippocampus in LB males but not female LB littermates. Given the critical role that these projections play in supporting normal hippocampal function, a failure to establish proper connectivity between the LEC and the dorsal hippocampus provides a compelling and novel mechanism to explain the more severe deficits in myelination and contextual freezing found in adolescent LB males.
Collapse
Affiliation(s)
- Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jordon D White
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Tanzil M Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
- Department of Biomedical Engineering, Center for Neurotechnology in Mental Health Research (CNMHR), The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sameet Mehta
- Yale Center for Genomic Analysis, P.O. Box 27386, West Haven, CT, 06516-7386, USA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, SHM IE26, New Haven, CT, 06510, USA
- Center for Cellular and Molecular Imaging, Electron Microscopy Core Facility, Yale University School of Medicine, New Haven, CT, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
| |
Collapse
|
12
|
Li H, Ye Q, Wang D, Shi B, Xu W, Zhang S, Han X, Zhang XY, Thompson GJ. Resting State Brain Networks under Inverse Agonist versus Complete Knockout of the Cannabinoid Receptor 1. ACS Chem Neurosci 2024; 15:1669-1683. [PMID: 38575140 PMCID: PMC11027912 DOI: 10.1021/acschemneuro.3c00804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
The cannabinoid receptor 1 (CB1) is famous as the target of Δ9-tetrahydrocannabinol (THC), which is the active ingredient of marijuana. Suppression of CB1 is frequently suggested as a drug target or gene therapy for many conditions (e.g., obesity, Parkinson's disease). However, brain networks affected by CB1 remain elusive, and unanticipated psychological effects in a clinical trial had dire consequences. To better understand the whole brain effects of CB1 suppression we performed in vivo imaging on mice under complete knockout of the gene for CB1 (cnr1-/-) and also under the CB1 inverse agonist rimonabant. We examined white matter structural changes and brain function (network activity and directional uniformity) in cnr1-/- mice. In cnr1-/- mice, white matter (in both sexes) and functional directional uniformity (in male mice) were altered across the brain but network activity was largely unaltered. Conversely, under rimonabant, functional directional uniformity was not altered but network activity was altered in cortical regions, primarily in networks known to be altered by THC (e.g., neocortex, hippocampal formation). However, rimonabant did not alter many brain regions found in both our cnr1-/- results and previous behavioral studies of cnr1-/- mice (e.g., thalamus, infralimbic area). This suggests that chronic loss of cnr1 is substantially different from short-term suppression, subtly rewiring the brain but largely maintaining the network activity. Our results help explain why pathological mutations in CB1 (e.g., chronic pain) do not always provide insight into the side effects of CB1 suppression (e.g., clinical depression), and thus urge more preclinical studies for any drugs that suppress CB1.
Collapse
Affiliation(s)
- Hui Li
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
| | - Qiong Ye
- High
Magnetic Field Laboratory, Hefei Institutes
of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Da Wang
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Bowen Shi
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Wenjing Xu
- Institute
of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
- Key
Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai 200433, China
| | - Shuning Zhang
- iHuman
Institute, ShanghaiTech University, Shanghai 201210, China
- School
of Life Science and Technology, ShanghaiTech
University, Shanghai 201210, China
| | - Xiaoyang Han
- Institute
of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
- Key
Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai 200433, China
| | - Xiao-Yong Zhang
- Institute
of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
- Key
Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai 200433, China
| | | |
Collapse
|
13
|
Ahmed S, Polis B, Jamwal S, Sanganahalli BG, Kaswan ZM, Islam R, Kim D, Bowers C, Giuliano L, Biederer T, Hyder F, Kaffman A. Transient Impairment in Microglial Function Causes Sex-Specific Deficits in Synaptic and Hippocampal Function in Mice Exposed to Early Adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580284. [PMID: 38405887 PMCID: PMC10888912 DOI: 10.1101/2024.02.14.580284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Abnormal development and function of the hippocampus are two of the most consistent findings in humans and rodents exposed to early life adversity, with males often being more affected than females. Using the limited bedding (LB) paradigm as a rodent model of early life adversity, we found that male adolescent mice that had been exposed to LB exhibit significant deficits in contextual fear conditioning and synaptic connectivity in the hippocampus, which are not observed in females. This is linked to altered developmental refinement of connectivity, with LB severely impairing microglial-mediated synaptic pruning in the hippocampus of male and female pups on postnatal day 17 (P17), but not in adolescent P33 mice when levels of synaptic engulfment by microglia are substantially lower. Since the hippocampus undergoes intense synaptic pruning during the second and third weeks of life, we investigated whether microglia are required for the synaptic and behavioral aberrations observed in adolescent LB mice. Indeed, transient ablation of microglia from P13-21, in normally developing mice caused sex-specific behavioral and synaptic abnormalities similar to those observed in adolescent LB mice. Furthermore, chemogenetic activation of microglia during the same period reversed the microglial-mediated phagocytic deficits at P17 and restored normal contextual fear conditioning and synaptic connectivity in adolescent LB male mice. Our data support an additional contribution of astrocytes in the sex-specific effects of LB, with increased expression of the membrane receptor MEGF10 and enhanced synaptic engulfment in hippocampal astrocytes of 17-day-old LB females, but not in LB male littermates. This finding suggests a potential compensatory mechanism that may explain the relative resilience of LB females. Collectively, these studies highlight a novel role for glial cells in mediating sex-specific hippocampal deficits in a mouse model of early-life adversity.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Basavaraju G. Sanganahalli
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Zoe MacDowell Kaswan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Dana Kim
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, 100 College Street, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| |
Collapse
|
14
|
Gee DG, Cohodes EM. Leveraging the developmental neuroscience of caregiving to promote resilience among youth exposed to adversity. Dev Psychopathol 2023; 35:2168-2185. [PMID: 37929292 PMCID: PMC10872788 DOI: 10.1017/s0954579423001128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Early adversity is a major risk factor for the emergence of psychopathology across development. Identifying mechanisms that support resilience, or favorable mental health outcomes despite exposure to adversity, is critical for informing clinical intervention and guiding policy to promote youth mental health. Here we propose that caregivers play a central role in fostering resilience among children exposed to adversity via caregiving influences on children's corticolimbic circuitry and emotional functioning. We first delineate the numerous ways that caregivers support youth emotional learning and regulation and describe how early attachment lays the foundation for optimal caregiver support of youth emotional functioning in a developmental stage-specific manner. Second, we outline neural mechanisms by which caregivers foster resilience-namely, by modulating offspring corticolimbic circuitry to support emotion regulation and buffer stress reactivity. Next, we highlight the importance of developmental timing and sensitive periods in understanding caregiving-related mechanisms of resilience. Finally, we discuss clinical implications of this line of research and how findings can be translated to guide policy that promotes the well-being of youth and families.
Collapse
|
15
|
Shah P, Kaneria A, Fleming G, Williams CRO, Sullivan RM, Lemon CH, Smiley J, Saito M, Wilson DA. Homeostatic NREM sleep and salience network function in adult mice exposed to ethanol during development. Front Neurosci 2023; 17:1267542. [PMID: 38033546 PMCID: PMC10682725 DOI: 10.3389/fnins.2023.1267542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Developmental exposure to ethanol is a leading cause of cognitive, emotional and behavioral problems, with fetal alcohol spectrum disorder (FASD) affecting more than 1:100 children. Recently, comorbid sleep deficits have been highlighted in these disorders, with sleep repair a potential therapeutic target. Animal models of FASD have shown non-REM (NREM) sleep fragmentation and slow-wave oscillation impairments that predict cognitive performance. Here we use a mouse model of perinatal ethanol exposure to explore whether reduced sleep pressure may contribute to impaired NREM sleep, and compare the function of a brain network reported to be impacted by insomnia-the Salience network-in developmental ethanol-exposed mice with sleep-deprived, saline controls. Mice were exposed to ethanol or saline on postnatal day 7 (P7) and allowed to mature to adulthood for testing. At P90, telemetered cortical recordings were made for assessment of NREM sleep in home cage before and after 4 h of sleep deprivation to assess basal NREM sleep and homeostatic NREM sleep response. To assess Salience network functional connectivity, mice were exposed to the 4 h sleep deprivation period or left alone, then immediately sacrificed for immunohistochemical analysis of c-Fos expression. The results show that developmental ethanol severely impairs both normal rebound NREM sleep and sleep deprivation induced increases in slow-wave activity, consistent with reduced sleep pressure. Furthermore, the Salience network connectome in rested, ethanol-exposed mice was most similar to that of sleep-deprived, saline control mice, suggesting a sleep deprivation-like state of Salience network function after developmental ethanol even without sleep deprivation.
Collapse
Affiliation(s)
- Prachi Shah
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Aayush Kaneria
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Gloria Fleming
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Colin R. O. Williams
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States
| | - Christian H. Lemon
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
| | - John Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- Department of Psychiatry, New York University Medical Center, New York, NY,United States
| | - Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- Department of Psychiatry, New York University Medical Center, New York, NY,United States
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY,United States
- School of Biological Sciences, University of Oklahoma, Norman, OK, United States
- Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
16
|
Zanfino G, Puzzo C, de Laurenzi V, Adriani W. Characterization of Behavioral Phenotypes in Heterozygous DAT Rat Based on Pedigree. Biomedicines 2023; 11:2565. [PMID: 37761006 PMCID: PMC10526166 DOI: 10.3390/biomedicines11092565] [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: 07/11/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Dopamine is an essential neurotransmitter whose key roles include movement control, pleasure and reward, attentional and cognitive skills, and regulation of the sleep/wake cycle. Reuptake is carried out by the dopamine transporter (DAT; DAT1 SLC6A3 gene). In order to study the effects of hyper-dopaminergia syndrome, the gene was silenced in rats. DAT-KO rats show stereotypical behavior, hyperactivity, a deficit in working memory, and an altered circadian cycle. In addition to KO rats, heterozygous (DAT-HET) rats show relative hypofunction of DAT; exact phenotypic effects are still unknown and may depend on whether the sire or the dam was KO. Our goal was to elucidate the potential importance of the parental origin of the healthy or silenced allele and its impact across generations, along with the potential variations in maternal care. We thus generated specular lines to study the effects of (grand) parental roles in inheriting the wild or mutated allele. MAT-HETs are the progeny of a KO sire and a WT dam; by breeding MAT-HET males and KO females, we obtained subjects with a DAT -/- epigenotype, named QULL, to reflect additional epigenetic DAT modulation when embryos develop within a hyper-dopaminergic KO uterus. We aimed to verify if any behavioral anomaly was introduced by a QULL (instead of KO) rat acting as a direct father or indirect maternal grandfather (or both). We thus followed epigenotypes obtained after three generations and observed actual effects on impaired maternal care of the offspring (based on pedigree). In particular, offspring of MAT-HET-dam × QULL-sire breeding showed a compulsive and obsessive phenotype. Although the experimental groups were all heterozygous, the impact of having a sire of epigenotype QULL (who developed in the uterus of a KO grand-dam) has emerged clearly. Along the generations, the effects of the DAT epigenotype on the obsessive/compulsive phenotype do vary as a function of the uterine impact on either allele in one's genealogical line.
Collapse
Affiliation(s)
- Gioia Zanfino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
| | - Concetto Puzzo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| | - Vincenzo de Laurenzi
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Walter Adriani
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (G.Z.); (C.P.)
- Faculty of Psychology, International Telematic University Uninettuno, 00186 Rome, Italy
| |
Collapse
|
17
|
Ip CK, Rezitis J, Qi Y, Bajaj N, Koller J, Farzi A, Shi YC, Tasan R, Zhang L, Herzog H. Critical role of lateral habenula circuits in the control of stress-induced palatable food consumption. Neuron 2023; 111:2583-2600.e6. [PMID: 37295418 DOI: 10.1016/j.neuron.2023.05.010] [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: 06/12/2022] [Revised: 12/15/2022] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Chronic stress fuels the consumption of palatable food and can enhance obesity development. While stress- and feeding-controlling pathways have been identified, how stress-induced feeding is orchestrated remains unknown. Here, we identify lateral habenula (LHb) Npy1r-expressing neurons as the critical node for promoting hedonic feeding under stress, since lack of Npy1r in these neurons alleviates the obesifying effects caused by combined stress and high fat feeding (HFDS) in mice. Mechanistically, this is due to a circuit originating from central amygdala NPY neurons, with the upregulation of NPY induced by HFDS initiating a dual inhibitory effect via Npy1r signaling onto LHb and lateral hypothalamus neurons, thereby reducing the homeostatic satiety effect through action on the downstream ventral tegmental area. Together, these results identify LHb-Npy1r neurons as a critical node to adapt the response to chronic stress by driving palatable food intake in an attempt to overcome the negative valence of stress.
Collapse
Affiliation(s)
- Chi Kin Ip
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Jemma Rezitis
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Yue Qi
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Nikita Bajaj
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Julia Koller
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Aitak Farzi
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Yan-Chuan Shi
- Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia; Neuroendocrinology Group, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Ramon Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Lei Zhang
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
18
|
Oh SJ, Lee N, Nam KR, Kang KJ, Han SJ, Choi JY. Effects of Escitalopram on the Functional Neural Circuits in an Animal Model of Adolescent Depression. Mol Imaging Biol 2023:10.1007/s11307-023-01825-6. [PMID: 37193806 DOI: 10.1007/s11307-023-01825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/18/2023]
Abstract
PURPOSE Although escitalopram is known to be an effective drug for adult depression, its disease-modifying efficacy on adolescents remains controversial. The present study aimed to evaluate the therapeutic effect of escitalopram on behavioral aspects as well as functional neural circuits by means of positron emission tomography. PROCEDURES To generate the animal models of depression, restraint stress was used during the peri-adolescent period (RS group). Thereafter, escitalopram was administered after the end of stress exposure (Tx group). We performed NeuroPET studies of glutamate, glutamate, GABA, and serotonin systems. RESULTS The Tx group showed no body weight change compared to the RS group. In the behavioral tests, the Tx group also displayed the similar time spent in open arms and immobility time to those for RS. In the PET studies, brain uptake values for the Tx group revealed no significant differences in terms of glucose, GABAA, and 5-HT1A receptor densities, but lower mGluR5 PET uptake compared to the RS group. In the immunohistochemistry, the Tx group showed the significant loss of neuronal cells in the hippocampus compared to the RS group. CONCLUSION The administration of escitalopram had no therapeutic effect on the adolescent depression.
Collapse
Affiliation(s)
- Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812
| | - Namhun Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea, 01812.
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, South Korea, 01812.
| |
Collapse
|
19
|
Khatibi VA, Salimi M, Rahdar M, Rezaei M, Nazari M, Dehghan S, Davoudi S, Raoufy MR, Mirnajafi-Zadeh J, Javan M, Hosseinmardi N, Behzadi G, Janahmadi M. Glycolysis inhibition partially resets epilepsy-induced alterations in the dorsal hippocampus-basolateral amygdala circuit involved in anxiety-like behavior. Sci Rep 2023; 13:6520. [PMID: 37085688 PMCID: PMC10119516 DOI: 10.1038/s41598-023-33710-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/18/2023] [Indexed: 04/23/2023] Open
Abstract
Pharmacoresistant temporal lobe epilepsy affects millions of people around the world with uncontrolled seizures and comorbidities, like anxiety, being the most problematic aspects calling for novel therapies. The intrahippocampal kainic acid model of temporal lobe epilepsy is an appropriate rodent model to evaluate the effects of novel interventions, including glycolysis inhibition, on epilepsy-induced alterations. Here, we investigated kainic acid-induced changes in the dorsal hippocampus (dHPC) and basolateral amygdala (BLA) circuit and the efficiency of a glycolysis inhibitor, 2-deoxy D-glucose (2-DG), in resetting such alterations using simultaneous local field potentials (LFP) recording and elevated zero-maze test. dHPC theta and gamma powers were lower in epileptic groups, both in the baseline and anxiogenic conditions. BLA theta power was higher in baseline condition while it was lower in anxiogenic condition in epileptic animals and 2-DG could reverse it. dHPC-BLA coherence was altered only in anxiogenic condition and 2-DG could reverse it only in gamma frequency. This coherence was significantly correlated with the time in which the animals exposed themselves to the anxiogenic condition. Further, theta-gamma phase-locking was lower in epileptic groups in the dHPC-BLA circuit and 2-DG could considerably increase it.
Collapse
Affiliation(s)
- Vahid Ahli Khatibi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Salimi
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Milad Nazari
- Department of Molecular Biology and Genetics, Aarhus University, Åarhus, Denmark
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center and Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Liu Q, Song X, Zhou X, Huang L, Zhang X, Wang L, Zhu S, Lan C, Yang W, Zhao W. Regional superficial amygdala resting-state functional connectivity in adults infers childhood maltreatment severity. PSYCHORADIOLOGY 2023; 3:kkad004. [PMID: 38666120 PMCID: PMC11003424 DOI: 10.1093/psyrad/kkad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2024]
Abstract
Background Childhood maltreatment (CM) is a potential risk factor for some neuropsychiatric disorders in adulthood (e.g. depression and anxiety) and alters trajectories of brain development. Accumulating evidence suggests that functional connectivity of the limbic system, especially the amygdala, is highly associated with childhood maltreatment, although not all studies have found this. These inconsistent results may be due to differential alterations of amygdala resting-state functional connectivity (rsFC) following childhood maltreatment. Objective Our aim was to investigate the relationship between the rsFC of amygdala subregions and CM severity, as well as to develop a stable rsFC-based model for inferring the severity of CM. Methods In this study, we employed the Childhood Trauma Questionnaire (CTQ) to assess CM severity in each individual. We explored the relationship between the rsFC of amygdala subregions (i.e. centromedial -CMA, basolateral -BLA, superficial-SFA amygdala) and CM experience in a discovery dataset of n = 110 healthy Chinese participants by linear multiple regression analysis. Subsequent dimensional and categorical approach were performed to elucidate the relationship between rsFCs and CM severity and CM subtypes, respectively. A support vector regression model was then conducted to validate the associations between rsFCs and total CTQ scores. Moreover, we also verified the model into another independent replication dataset (n = 38). Results Our findings suggested that childhood maltreatment was negatively associated with rsFC between the right superficial amygdala and perigenual anterior cingulate cortex (pgACC)/postcentral gyrus (PCG) but not the other two amygdala subregions. Moreover, SFA-pgACC coupling was more associated with physical neglect whereas the SFA-PCG was more related to emotional neglect. In addition, supervised machine learning confirmed that using these two rsFCs as predictors could stably estimate continuous maltreatment severity in both discovery and replication datasets. Conclusion The current study supports that the rsFCs of superficial amygdala are related to childhood maltreatment and which may be a potential biomarker for the effects of childhood maltreatment-related psychiatric disorders (i.e. depression and anxiety).
Collapse
Affiliation(s)
- Qi Liu
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xinwei Song
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xinqi Zhou
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu 610066, China
| | - Linghong Huang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xiaodong Zhang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lan Wang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Siyu Zhu
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Chunmei Lan
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenxu Yang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Weihua Zhao
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Institute of Electronic and Information Engineering of UESTC in Guangdong, Dongguan 523808, China
| |
Collapse
|
21
|
Komanchuk J, Letourneau N, Duffett-Leger L, Cameron JL. History of "Serve and Return" and a Synthesis of the Literature on its Impacts on Children's Health and Development. Issues Ment Health Nurs 2023; 44:406-417. [PMID: 37015096 DOI: 10.1080/01612840.2023.2192794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
Parent/caregiver sensitivity and responsiveness are important for children's health and development. The "serve and return" metaphor was created to help providers and caregivers understand the importance of sensitive and responsive early caregiving. In this review, we explain the concept of "serve and return", outline historical and theoretical principles that culminated in this metaphor, highlight parent and child constructs associated with "serve and return" interactions, and synthesize literature on sensitive and responsive caregiving and children's health and developmental outcomes. Nurses and other healthcare professionals in public policy, clinical, community, education, and research roles need knowledge of "serve and return" interactions.
Collapse
Affiliation(s)
- Jelena Komanchuk
- Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada
| | | | | | - Judy L Cameron
- Faculty of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Holz NE, Berhe O, Sacu S, Schwarz E, Tesarz J, Heim CM, Tost H. Early Social Adversity, Altered Brain Functional Connectivity, and Mental Health. Biol Psychiatry 2023; 93:430-441. [PMID: 36581495 DOI: 10.1016/j.biopsych.2022.10.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022]
Abstract
Early adverse environmental exposures during brain development are widespread risk factors for the onset of severe mental disorders and strong and consistent predictors of stress-related mental and physical illness and reduced life expectancy. Current evidence suggests that early negative experiences alter plasticity processes during developmentally sensitive time windows and affect the regular functional interaction of cortical and subcortical neural networks. This, in turn, may promote a maladapted development with negative consequences on the mental and physical health of exposed individuals. In this review, we discuss the role of functional magnetic resonance imaging-based functional connectivity phenotypes as potential biomarker candidates for the consequences of early environmental exposures-including but not limited to-childhood maltreatment. We take an expanded concept of developmentally relevant adverse experiences from infancy over childhood to adolescence as our starting point and focus our review of functional connectivity studies on a selected subset of functional magnetic resonance imaging-based phenotypes, including connectivity in the limbic and within the frontoparietal as well as default mode networks, for which we believe there is sufficient converging evidence for a more detailed discussion in a developmental context. Furthermore, we address specific methodological challenges and current knowledge gaps that complicate the interpretation of early stress effects on functional connectivity and deserve particular attention in future studies. Finally, we highlight the forthcoming prospects and challenges of this research area with regard to establishing functional connectivity measures as validated biomarkers for brain developmental processes and individual risk stratification and as target phenotypes for mechanism-based interventions.
Collapse
Affiliation(s)
- Nathalie E Holz
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands; Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig Holstein, Kiel University, Kiel, Germany; Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Oksana Berhe
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Seda Sacu
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jonas Tesarz
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Christine M Heim
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Psychology, Berlin, Germany; College of Health and Human Development, The Pennsylvania State University, University Park, Pennsylvania
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
23
|
A Novel Early Life Stress Model Affects Brain Development and Behavior in Mice. Int J Mol Sci 2023; 24:ijms24054688. [PMID: 36902120 PMCID: PMC10002977 DOI: 10.3390/ijms24054688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Early life stress (ELS) in developing children has been linked to physical and psychological sequelae in adulthood. In the present study, we investigated the effects of ELS on brain and behavioral development by establishing a novel ELS model that combined the maternal separation paradigm and mesh platform condition. We found that the novel ELS model caused anxiety- and depression-like behaviors and induced social deficits and memory impairment in the offspring of mice. In particular, the novel ELS model induced more enhanced depression-like behavior and memory impairment than the maternal separation model, which is the established ELS model. Furthermore, the novel ELS caused upregulation of arginine vasopressin expression and downregulation of GABAergic interneuron markers, such as parvalbumin (PV), vasoactive intestinal peptide, and calbindin-D28k (CaBP-28k), in the brains of the mice. Finally, the offspring in the novel ELS model showed a decreased number of cortical PV-, CaBP-28k-positive cells and an increased number of cortical ionized calcium-binding adaptors-positive cells in their brains compared to mice in the established ELS model. Collectively, these results indicated that the novel ELS model induced more negative effects on brain and behavioral development than the established ELS model.
Collapse
|
24
|
Stieger B, Wesseler Y, Kaiser S, Sachser N, Richter SH. Behavioral lateralization of mice varying in serotonin transporter genotype. Front Behav Neurosci 2023; 16:1095567. [PMID: 36710954 PMCID: PMC9875089 DOI: 10.3389/fnbeh.2022.1095567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
In humans, non-right-handedness is associated with a higher incidence of psychiatric disorders. Since serotonin seems to be involved in both, the development of psychiatric disorders and lateralization, the present study focuses on the effect of the serotonin transporter (5-HTT) gene on behavioral lateralization. For this, we used the 5-HTT knockout mouse model, a well-established animal model for the study of human depression and anxiety disorders. For female mice from all three 5-HTT genotypes (wild type, heterozygous, and homozygous knockout), we repeatedly observed the direction and strength of lateralization of the following four behaviors: grid climbing (GC), food-reaching in an artificial test situation (FRT), self-grooming (SG), and barrier crossing (BC), with the FRT being the standard test for assessing behavioral lateralization in mice. We found no association between behavioral lateralization and 5-HTT genotype. However, in accordance with previous findings, the strength and temporal consistency of lateralization differed between the four behaviors observed. In conclusion, since the 5-HTT genotype did not affect behavioral lateralization in mice, more research on other factors connected with behavioral lateralization and the development of symptoms of psychiatric disorders, such as environmental influences, is needed.
Collapse
Affiliation(s)
- Binia Stieger
- Department of Behavioural Biology, University of Münster, Münster, Germany,DFG Research Training Group EvoPAD, University of Münster, Münster, Germany,*Correspondence: Binia Stieger,
| | - Yvonne Wesseler
- Department of Behavioural Biology, University of Münster, Münster, Germany
| | - Sylvia Kaiser
- Department of Behavioural Biology, University of Münster, Münster, Germany,DFG Research Training Group EvoPAD, University of Münster, Münster, Germany
| | - Norbert Sachser
- Department of Behavioural Biology, University of Münster, Münster, Germany,DFG Research Training Group EvoPAD, University of Münster, Münster, Germany
| | - S. Helene Richter
- Department of Behavioural Biology, University of Münster, Münster, Germany,DFG Research Training Group EvoPAD, University of Münster, Münster, Germany
| |
Collapse
|
25
|
Dayananda KK, Ahmed S, Wang D, Polis B, Islam R, Kaffman A. Early life stress impairs synaptic pruning in the developing hippocampus. Brain Behav Immun 2023; 107:16-31. [PMID: 36174883 PMCID: PMC10497209 DOI: 10.1016/j.bbi.2022.09.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023] Open
Abstract
Early life adversity impairs normal hippocampal function and connectivity in various mammalian species, including humans and rodents. According to the 'cumulative model' the number of early adversities can be summed up to determine the risk for developing psychopathology later in life. In contrast, the 'dimensional model' argues that 'Deprivation' and 'Threat' impact different developmental processes that should not be added in determining clinical outcomes. Here we examine these predictions in male and female mice exposed to a single adversity - limited bedding (LB) - versus mice exposed to multiple adversities - unpredictable postnatal stress (UPS) - focusing on microglia-mediated synaptic pruning in the developing hippocampus. Exposure to both LB and UPS reduced the ramification of microglia, impaired their ability to phagocytose synaptic material in vivo and ex vivo, and decreased expression of TREM2. Abnormal phagocytic activity was associated with increased spine density in CA1 pyramidal neurons that was seen in 17-day-old groups and persisted in peri-pubescent 29-day-old LB and UPS mice. Exposure to LB caused more severe impairment in microglial ramification and synaptic engulfment compared to UPS, outcomes that were accompanied by a UPS-specific increase in the expression of several genes implicated in synaptic pruning. We propose that despite being a single stressor, LB represents a more severe form of early deprivation, and that appropriate levels of hippocampal stimulation during the second and third weeks of life are necessary to support normal microglial ramification and synaptic pruning. Further, impaired synaptic pruning during this critical period of hippocampal development contributes to the abnormal hippocampal function and connectivity seen in UPS and LB later in life.
Collapse
Affiliation(s)
- Kiran K Dayananda
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Daniel Wang
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| |
Collapse
|
26
|
Demaili A, Portugalov A, Dudai M, Maroun M, Akirav I, Braun K, Bock J. Epigenetic (re)programming of gene expression changes of CB1R and FAAH in the medial prefrontal cortex in response to early life and adolescence stress exposure. Front Cell Neurosci 2023; 17:1129946. [PMID: 36909279 PMCID: PMC9992175 DOI: 10.3389/fncel.2023.1129946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Environmental factors, including stress, that are experienced during early life (ELS) or adolescence are potential risk factors for the development of behavioral and mental disorders later in life. The endocannabinoid system plays a major role in the regulation of stress responses and emotional behavior, thereby acting as a mediator of stress vulnerability and resilience. Among the critical factors, which determine the magnitude and direction of long-term consequences of stress exposure is age, i.e., the maturity of brain circuits during stress exposure. Thus, the present study addressed the hypotheses that ELS and adolescent stress differentially affect the expression of regulatory elements of the endocannabinoid system, cannabinoid receptor 1 (CB1R) and fatty acid amide hydrolase (FAAH) in the medial prefrontal cortex (mPFC) of adult female rats. We also tested the hypothesis that the proposed gene expression changes are epigenetically modulated via altered DNA-methylation. The specific aims were to investigate if (i) ELS and adolescent stress as single stressors induce changes in CB1R and FAAH expression (ii) ELS exposure influences the effect of adolescent stress on CB1R and FAAH expression, and (iii) if the proposed gene expression changes are paralleled by changes of DNA methylation. The following experimental groups were investigated: (1) non-stressed controls (CON), (2) ELS exposure (ELS), (3) adolescent stress exposure (forced swimming; FS), (4) ELS + FS exposure. We found an up-regulation of CB1R expression in both single-stressor groups and a reduction back to control levels in the ELS + FS group. An up-regulation of FAAH expression was found only in the FS group. The data indicate that ELS, i.e., stress during a very immature stage of brain development, exerts a buffering programming effect on gene expression changes induced by adolescent stress. The detected gene expression changes were accompanied by altered DNA methylation patterns in the promoter region of these genes, specifically, a negative correlation of mean CB1R DNA methylation with gene expression was found. Our results also indicate that ELS induces a long-term "(re)programming" effect, characterized by CpG-site specific changes within the promoter regions of the two genes that influence gene expression changes in response to FS at adolescence.
Collapse
Affiliation(s)
- Arijana Demaili
- Department of Zoology and Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Anna Portugalov
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa, Israel.,The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| | - Michal Dudai
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa, Israel.,The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| | - Mouna Maroun
- The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel.,Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Irit Akirav
- Department of Psychology, School of Psychological Sciences, University of Haifa, Haifa, Israel.,The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| | - Katharina Braun
- Department of Zoology and Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Center for Brain and Behavioral Science, Magdeburg, Germany
| | - Jörg Bock
- Department of Zoology and Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Center for Brain and Behavioral Science, Magdeburg, Germany.,Project Group (PG) Epigenetics and Structural Plasticity, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
27
|
Haikonen J, Englund J, Amarilla SP, Kharybina Z, Shintyapina A, Kegler K, Garcia MS, Atanasova T, Taira T, Hartung H, Lauri SE. Aberrant cortical projections to amygdala GABAergic neurons contribute to developmental circuit dysfunction following early life stress. iScience 2022; 26:105724. [PMID: 36582824 PMCID: PMC9792886 DOI: 10.1016/j.isci.2022.105724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Early life stress (ELS) results in enduring dysfunction of the corticolimbic circuitry, underlying emotional and social behavior. However, the neurobiological mechanisms involved remain elusive. Here, we have combined viral tracing and electrophysiological techniques to study the effects of maternal separation (MS) on frontolimbic connectivity and function in young (P14-21) rats. We report that aberrant prefrontal inputs to basolateral amygdala (BLA) GABAergic interneurons transiently increase the strength of feed-forward inhibition in the BLA, which raises LTP induction threshold in MS treated male rats. The enhanced GABAergic activity after MS exposure associates with lower functional synchronization within prefrontal-amygdala networks in vivo. Intriguingly, no differences in these parameters were detected in females, which were also resistant to MS dependent changes in anxiety-like behaviors. Impaired plasticity and synchronization during the sensitive period of circuit refinement may contribute to long-lasting functional changes in the prefrontal-amygdaloid circuitry that predispose to neuropsychiatric conditions later on in life.
Collapse
Affiliation(s)
- Joni Haikonen
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Jonas Englund
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Shyrley Paola Amarilla
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Zoia Kharybina
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Alexandra Shintyapina
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kristel Kegler
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Marta Saez Garcia
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Tsvetomira Atanasova
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Henrike Hartung
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Sari E. Lauri
- HiLife Neuroscience Center, University of Helsinki, Helsinki, Finland,Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland,Corresponding author
| |
Collapse
|
28
|
Gee DG. Neurodevelopmental mechanisms linking early experiences and mental health: Translating science to promote well-being among youth. AMERICAN PSYCHOLOGIST 2022; 77:1033-1045. [PMID: 36595400 PMCID: PMC9875304 DOI: 10.1037/amp0001107] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Early experiences can have profound and lasting effects on mental health. Delineating neurodevelopmental pathways related to risk and resilience following adversity exposure is critical for promoting well-being and targeting interventions. A rapidly growing cross-species literature has facilitated advances in identifying neural and behavioral mechanisms linking early experiences with mental health, highlighting a central role of corticolimbic circuitry involved in learning and emotion regulation. Building upon knowledge of corticolimbic development related to stress and buffering factors, we describe the importance of the developmental timing and experiential elements of adversity in mental health outcomes. Finally, we discuss opportunities to translate knowledge of the developing brain and early experiences to optimize interventions for youth with psychopathology and to inform policy that promotes healthy development at the societal level. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
|
29
|
Caetano I, Ferreira S, Coelho A, Amorim L, Castanho TC, Portugal-Nunes C, Soares JM, Gonçalves N, Sousa R, Reis J, Lima C, Marques P, Moreira PS, Rodrigues AJ, Santos NC, Morgado P, Magalhães R, Picó-Pérez M, Cabral J, Sousa N. Perceived stress modulates the activity between the amygdala and the cortex. Mol Psychiatry 2022; 27:4939-4947. [PMID: 36117211 DOI: 10.1038/s41380-022-01780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 01/14/2023]
Abstract
The significant link between stress and psychiatric disorders has prompted research on stress's impact on the brain. Interestingly, previous studies on healthy subjects have demonstrated an association between perceived stress and amygdala volume, although the mechanisms by which perceived stress can affect brain function remain unknown. To better understand what this association entails at a functional level, herein, we explore the association of perceived stress, measured by the PSS10 questionnaire, with disseminated functional connectivity between brain areas. Using resting-state fMRI from 252 healthy subjects spanning a broad age range, we performed both a seed-based amygdala connectivity analysis (static connectivity, with spatial resolution but no temporal definition) and a whole-brain data-driven approach to detect altered patterns of phase interactions between brain areas (dynamic connectivity with spatiotemporal information). Results show that increased perceived stress is directly associated with increased amygdala connectivity with frontal cortical regions, which is driven by a reduced occurrence of an activity pattern where the signals in the amygdala and the hippocampus evolve in opposite directions with respect to the rest of the brain. Overall, these results not only reinforce the pathological effect of in-phase synchronicity between subcortical and cortical brain areas but also demonstrate the protective effect of counterbalanced (i.e., phase-shifted) activity between brain subsystems, which are otherwise missed with correlation-based functional connectivity analysis.
Collapse
Affiliation(s)
- Inês Caetano
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Sónia Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Ana Coelho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Liliana Amorim
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal.,Association P5 Digital Medical Center (ACMP5), 4710-057, Braga, Portugal
| | - Teresa Costa Castanho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal.,Association P5 Digital Medical Center (ACMP5), 4710-057, Braga, Portugal
| | - Carlos Portugal-Nunes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal.,CECAV-Veterinary and Animal Science Research Centre, Quinta de Prados, 5000-801, Vila Real, Portugal
| | - José Miguel Soares
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Nuno Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Rui Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal.,Departamento de Psiquiatria e Saúde Mental, Centro Hospitalar Tondela-Viseu, 3500-228, Viseu, Portugal
| | - Joana Reis
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Catarina Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Paulo Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Pedro Silva Moreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Nadine Correia Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Pedro Morgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Ricardo Magalhães
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Maria Picó-Pérez
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Joana Cabral
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal. .,ICVS/3B's, PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal. .,Clinical Academic Center-Braga (2CA), 4710-243, Braga, Portugal. .,Association P5 Digital Medical Center (ACMP5), 4710-057, Braga, Portugal.
| |
Collapse
|
30
|
Schnider M, Jenni R, Ramain J, Camporesi S, Golay P, Alameda L, Conus P, Do KQ, Steullet P. Time of exposure to social defeat stress during childhood and adolescence and redox dysregulation on long-lasting behavioral changes, a translational study. Transl Psychiatry 2022; 12:413. [PMID: 36163247 PMCID: PMC9512907 DOI: 10.1038/s41398-022-02183-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/09/2022] Open
Abstract
Traumatic events during childhood/early adolescence can cause long-lasting physiological and behavioral changes with increasing risk for psychiatric conditions including psychosis. Genetic factors and trauma (and their type, degree of repetition, time of occurrence) are believed to influence how traumatic experiences affect an individual. Here, we compared long-lasting behavioral effects of repeated social defeat stress (SD) applied during either peripuberty or late adolescence in adult male WT and Gclm-KO mice, a model of redox dysregulation relevant to schizophrenia. As SD disrupts redox homeostasis and causes oxidative stress, we hypothesized that KO mice would be particularly vulnerable to such stress. We first found that peripubertal and late adolescent SD led to different behavioral outcomes. Peripubertal SD induced anxiety-like behavior in anxiogenic environments, potentiated startle reflex, and increased sensitivity to the NMDA-receptor antagonist, MK-801. In contrast, late adolescent SD led to increased exploration in novel environments. Second, the long-lasting impact of peripubertal but not late adolescent SD differed in KO and WT mice. Peripubertal SD increased anxiety-like behavior in anxiogenic environments and MK-801-sensitivity mostly in KO mice, while it increased startle reflex in WT mice. These suggest that a redox dysregulation during peripuberty interacts with SD to remodel the trajectory of brain maturation, but does not play a significant role during later SD. As peripubertal SD induced persisting anxiety- and fear-related behaviors in male mice, we then investigated anxiety in a cohort of 89 early psychosis male patients for whom we had information about past abuse and clinical assessment during the first year of psychosis. We found that a first exposure to physical/sexual abuse (analogous to SD) before age 12, but not after, was associated with higher anxiety at 6-12 months after psychosis onset. This supports that childhood/peripuberty is a vulnerable period during which physical/sexual abuse in males has wide and long-lasting consequences.
Collapse
Affiliation(s)
- Mirko Schnider
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Sara Camporesi
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Luis Alameda
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), 1008, Prilly-Lausanne, Switzerland.
| |
Collapse
|
31
|
Uliana DL, Zhu X, Gomes FV, Grace AA. Using animal models for the studies of schizophrenia and depression: The value of translational models for treatment and prevention. Front Behav Neurosci 2022; 16:935320. [PMID: 36090659 PMCID: PMC9449416 DOI: 10.3389/fnbeh.2022.935320] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Animal models of psychiatric disorders have been highly effective in advancing the field, identifying circuits related to pathophysiology, and identifying novel therapeutic targets. In this review, we show how animal models, particularly those based on development, have provided essential information regarding circuits involved in disorders, disease progression, and novel targets for intervention and potentially prevention. Nonetheless, in recent years there has been a pushback, largely driven by the US National Institute of Mental Health (NIMH), to shift away from animal models and instead focus on circuits in normal subjects. This has been driven primarily from a lack of discovery of new effective therapeutic targets, and the failure of targets based on preclinical research to show efficacy. We discuss why animal models of complex disorders, when strongly cross-validated by clinical research, are essential to understand disease etiology as well as pathophysiology, and direct new drug discovery. Issues related to shortcomings in clinical trial design that confound translation from animal models as well as the failure to take patient pharmacological history into account are proposed to be a source of the failure of what are likely effective compounds from showing promise in clinical trials.
Collapse
Affiliation(s)
- Daniela L. Uliana
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiyu Zhu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A. Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
32
|
Petković A, Chaudhury D. Encore: Behavioural animal models of stress, depression and mood disorders. Front Behav Neurosci 2022; 16:931964. [PMID: 36004305 PMCID: PMC9395206 DOI: 10.3389/fnbeh.2022.931964] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Animal studies over the past two decades have led to extensive advances in our understanding of pathogenesis of depressive and mood disorders. Among these, rodent behavioural models proved to be of highest informative value. Here, we present a comprehensive overview of the most popular behavioural models with respect to physiological, circuit, and molecular biological correlates. Behavioural stress paradigms and behavioural tests are assessed in terms of outcomes, strengths, weaknesses, and translational value, especially in the domain of pharmacological studies.
Collapse
Affiliation(s)
| | - Dipesh Chaudhury
- Laboratory of Neural Systems and Behaviour, Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
33
|
Waters RC, Gould E. Early Life Adversity and Neuropsychiatric Disease: Differential Outcomes and Translational Relevance of Rodent Models. Front Syst Neurosci 2022; 16:860847. [PMID: 35813268 PMCID: PMC9259886 DOI: 10.3389/fnsys.2022.860847] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/19/2022] [Indexed: 12/02/2022] Open
Abstract
It is now well-established that early life adversity (ELA) predisposes individuals to develop several neuropsychiatric conditions, including anxiety disorders, and major depressive disorder. However, ELA is a very broad term, encompassing multiple types of negative childhood experiences, including physical, sexual and emotional abuse, physical and emotional neglect, as well as trauma associated with chronic illness, family separation, natural disasters, accidents, and witnessing a violent crime. Emerging literature suggests that in humans, different types of adverse experiences are more or less likely to produce susceptibilities to certain conditions that involve affective dysfunction. To investigate the driving mechanisms underlying the connection between experience and subsequent disease, neuroscientists have developed several rodent models of ELA, including pain exposure, maternal deprivation, and limited resources. These studies have also shown that different types of ELA paradigms produce different but somewhat overlapping behavioral phenotypes. In this review, we first investigate the types of ELA that may be driving different neuropsychiatric outcomes and brain changes in humans. We next evaluate whether rodent models of ELA can provide translationally relevant information regarding links between specific types of experience and changes in neural circuits underlying dysfunction.
Collapse
Affiliation(s)
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
| |
Collapse
|
34
|
Ivanova D, Li X, Liu Y, McIntyre C, Fernandes C, Lass G, Kong L, O’Byrne KT. Role of Posterodorsal Medial Amygdala Urocortin-3 in Pubertal Timing in Female Mice. Front Endocrinol (Lausanne) 2022; 13:893029. [PMID: 35655799 PMCID: PMC9152449 DOI: 10.3389/fendo.2022.893029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 12/04/2022] Open
Abstract
Post-traumatic stress disorder impedes pubertal development and disrupts pulsatile LH secretion in humans and rodents. The posterodorsal sub-nucleus of the medial amygdala (MePD) is an upstream modulator of the hypothalamic gonadotropin-releasing hormone (GnRH) pulse generator, pubertal timing, as well as emotional processing and anxiety. Psychosocial stress exposure alters neuronal activity within the MePD increasing the expression of Urocortin3 (Ucn3) and its receptor corticotropin-releasing factor type-2 receptor (CRFR2) while enhancing the inhibitory output from the MePD to key hypothalamic reproductive centres. We test the hypothesis that psychosocial stress, processed by the MePD, is relayed to the hypothalamic GnRH pulse generator to delay puberty in female mice. We exposed C57Bl6/J female mice to the predator odor, 2,4,5-Trimethylthiazole (TMT), during pubertal transition and examined the effect on pubertal timing, pre-pubertal LH pulses and anxiety-like behaviour. Subsequently, we virally infected Ucn3-cre-tdTomato female mice with stimulatory DREADDs targeting MePD Ucn3 neurons and determined the effect on pubertal timing and pre-pubertal LH pulse frequency. Exposure to TMT during pubertal development delayed puberty, suppressed pre-pubertal LH pulsatility and enhanced anxiety-like behaviour, while activation of MePD Ucn3 neurons reduced LH pulse frequency and delayed puberty. Early psychosocial stress exposure decreases GnRH pulse generator frequency delaying puberty while inducing anxiety-behaviour in female mice, an effect potentially involving Ucn3 neurons in the MePD.
Collapse
Affiliation(s)
- Deyana Ivanova
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - XiaoFeng Li
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Yali Liu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Caitlin McIntyre
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Geffen Lass
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Lingsi Kong
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Kevin T. O’Byrne
- Department of Women and Children’s Health, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
35
|
Ouyang M, Zhang Q, Shu J, Wang Z, Fan J, Yu K, Lei L, Li Y, Wang Q. Capsaicin Ameliorates the Loosening of Mitochondria-Associated Endoplasmic Reticulum Membranes and Improves Cognitive Function in Rats With Chronic Cerebral Hypoperfusion. Front Cell Neurosci 2022; 16:822702. [PMID: 35370565 PMCID: PMC8968035 DOI: 10.3389/fncel.2022.822702] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/10/2022] [Indexed: 12/17/2022] Open
Abstract
Based on accumulating evidence, vascular factors contribute to cognitive decline and dementia. Mitochondrial dysfunction is the core pathophysiological mechanism. Mitochondria-associated endoplasmic reticulum membranes (MAMs) are subcellular structures that physically and biologically connect mitochondria with the endoplasmic reticulum (ER) and regulate multiple functions ranging from calcium transfer to mitochondrial dynamics and bioenergetics. MAMs dysfunction has been speculated to be a key factor contributing to the pathogenesis of cognitive disorders and a new therapeutic target. However, the alteration of MAMs in vascular cognitive impairment remains to be revealed. Capsaicin, a specific agonist known to activated the transient receptor potential vanilloid type 1 (TRPV1), is involved in hippocampal synaptic plasticity and memory, but the detailed mechanism is still unclear. In this study, chronic cerebral hypoperfusion (CCH) model rats were created by bilateral common carotid artery occlusion (BCCAO), which is a widely used model to study vascular dementia. We observed that CCH rats showed obvious cognitive deficits, and ER-mitochondria contacts were loosener with lower expression of mitofusin2 (MFN2), a key protein connecting MAMs, in the hippocampal CA1 region, compared to the sham group. After capsaicin treatment for 12 weeks, we found that cognitive deficits induced by CCH were significantly alleviated and loosened ER-mitochondrial interactions were obviously improved. In conclusion, the findings of this study highlight that MAMs may contribute to the pathogenesis of cognitive impairment induced by CCH, and our new evidence that capsaicin improves cognitive function highlights a novel opportunity for drug discovery.
Collapse
Affiliation(s)
- Mengqi Ouyang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Qi Zhang
- Department of Pharmacology, Gaoping District People’s Hospital of Nanchong, Nanchong, China
| | - Jiahui Shu
- Department of Pharmacology, Yichang Yiling Hospital, Yichang, China
| | - Zhiqiang Wang
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Jin Fan
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Lei Lei
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
| | - Yuxia Li
- Department of Neurology, Chengdu BOE Hospital, Chengdu, China
| | - Qingsong Wang
- Department of Neurology, The General Hospital of Western Theater Command, Chengdu, China
- *Correspondence: Qingsong Wang,
| |
Collapse
|
36
|
Manno FAM, Kumar R, An Z, Khan MS, Su J, Liu J, Wu EX, He J, Feng Y, Lau C. Structural and Functional Hippocampal Correlations in Environmental Enrichment During the Adolescent to Adulthood Transition in Mice. Front Syst Neurosci 2022; 15:807297. [PMID: 35242015 PMCID: PMC8886042 DOI: 10.3389/fnsys.2021.807297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/14/2021] [Indexed: 01/13/2023] Open
Abstract
Environmental enrichment is known to induce neuronal changes; however, the underlying structural and functional factors involved are not fully known and remain an active area of study. To investigate these factors, we assessed enriched environment (EE) and standard environment (SE) control mice over 30 days using structural and functional MRI methods. Naïve adult male mice (n = 30, ≈20 g, C57BL/B6J, postnatal day 60 initial scan) were divided into SE and EE groups and scanned before and after 30 days. Structural analyses included volumetry based on manual segmentation as well as diffusion tensor imaging (DTI). Functional analyses included seed-based analysis (SBA), independent component analysis (ICA), the amplitude of low-frequency fluctuation (ALFF), and fractional ALFF (fALFF). Structural results indicated that environmental enrichment led to an increase in the volumes of cornu ammonis 1 (CA1) and dentate gyrus. Structural results indicated changes in radial diffusivity and mean diffusivity in the visual cortex and secondary somatosensory cortex after EE. Furthermore, SBA and ICA indicated an increase in resting-state functional MRI (rsfMRI) functional connectivity in the hippocampus. Using parallel structural and functional analyses, we have demonstrated coexistent structural and functional changes in the hippocampal subdivision CA1. Future research should map alterations temporally during environmental enrichment to investigate the initiation of these structural and functional changes.
Collapse
Affiliation(s)
- Francis A M Manno
- Center for Imaging Science, Department of Biomedical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States.,Department of Physics, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Rachit Kumar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Medical Scientist Training Program, University of Pennsylvania, Philadelphia, PA, United States
| | - Ziqi An
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| | - Muhammad Shehzad Khan
- Department of Physics, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Junfeng Su
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Jiaming Liu
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| | - Ed X Wu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yanqiu Feng
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| | - Condon Lau
- Department of Physics, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
37
|
An exploration of dimensions of early adversity and the development of functional brain network connectivity during adolescence: Implications for trajectories of internalizing symptoms. Dev Psychopathol 2022; 34:557-571. [PMID: 35094729 DOI: 10.1017/s0954579421001814] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Different dimensions of adversity may affect mental health through distinct neurobiological mechanisms, though current supporting evidence consists largely of cross-sectional associations between threat or deprivation and fronto-limbic circuitry. In this exploratory three-wave longitudinal study spanning ages 9-19 years, we examined the associations between experiences of unpredictability, threat, and deprivation with the development of functional connectivity within and between three brain networks implicated in psychopathology: the salience (SAL), default mode (DMN), and fronto-parietal (FPN) networks, and tested whether network trajectories moderated associations between adversity and changes in internalizing symptoms. Connectivity decreased with age on average; these changes differed by dimension of adversity. Whereas family-level deprivation was associated with lower initial levels and more stability across most networks, unpredictability was associated with stability only in SAL connectivity, and threat was associated with stability in FPN and DMN-SAL connectivity. In youth exposed to higher levels of any adversity, lower initial levels and more stability in connectivity were related to smaller increases in internalizing symptoms. Our findings suggest that whereas deprivation is associated with widespread neurodevelopmental differences in cognitive and emotion processing networks, unpredictability is related selectively to salience detection circuitry. Studies with wider developmental windows should examine whether these neurodevelopmental alterations are adaptive or serve to maintain internalizing symptoms.
Collapse
|
38
|
Waters RC, Worth HM, Vasquez B, Gould E. Inhibition of adult neurogenesis reduces avoidance behavior in male, but not female, mice subjected to early life adversity. Neurobiol Stress 2022; 17:100436. [PMID: 35146080 PMCID: PMC8819473 DOI: 10.1016/j.ynstr.2022.100436] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Early life adversity (ELA) increases the risk of developing neuropsychiatric illnesses such as anxiety disorders. However, the mechanisms connecting these negative early life experiences to illness later in life remain unclear. In rodents, plasticity mechanisms, specifically adult neurogenesis in the ventral hippocampus, have been shown to be altered by ELA and important for buffering against detrimental stress-induced outcomes. The current study sought to explore whether adult neurogenesis contributes to ELA-induced changes in avoidance behavior. Using the GFAP-TK transgenic model, which allows for the inhibition of adult neurogenesis, and CD1 littermate controls, we subjected mice to an ELA paradigm of maternal separation and early weaning (MSEW) or control rearing. We found that mice with intact adult neurogenesis showed no behavioral changes in response to MSEW. After reducing adult neurogenesis, however, male mice previously subjected to MSEW had an unexpected decrease in avoidance behavior. This finding was not observed in female mice, suggesting that a sex difference exists in the role of adult-born neurons in buffering against ELA-induced changes in behavior. Taken together with the existing literature on ELA and avoidance behavior, this work suggests that strain differences exist in susceptibility to ELA and that adult-born neurons may play a role in regulating adaptive behavior.
Collapse
|
39
|
Zhu Y, Wang MJ, Crawford KM, Ramírez-Tapia JC, Lussier AA, Davis KA, de Leeuw C, Takesian AE, Hensch TK, Smoller JW, Dunn EC. Sensitive period-regulating genetic pathways and exposure to adversity shape risk for depression. Neuropsychopharmacology 2022; 47:497-506. [PMID: 34689167 PMCID: PMC8674315 DOI: 10.1038/s41386-021-01172-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 07/23/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023]
Abstract
Animal and human studies have documented the existence of developmental windows (or sensitive periods) when experience can have lasting effects on brain structure or function, behavior, and disease. Although sensitive periods for depression likely arise through a complex interplay of genes and experience, this possibility has not yet been explored in humans. We examined the effect of genetic pathways regulating sensitive periods, alone and in interaction with common childhood adversities, on depression risk. Guided by a translational approach, we: (1) performed association analyses of three gene sets (60 genes) shown in animal studies to regulate sensitive periods using summary data from a genome-wide association study of depression (n = 807,553); (2) evaluated the developmental expression patterns of these genes using data from BrainSpan (n = 31), a transcriptional atlas of postmortem brain samples; and (3) tested gene-by-development interplay (dGxE) by analyzing the combined effect of common variants in sensitive period genes and time-varying exposure to two types of childhood adversity within a population-based birth cohort (n = 6254). The gene set regulating sensitive period opening associated with increased depression risk. Notably, 6 of the 15 genes in this set showed developmentally regulated gene-level expression. We also identified a statistical interaction between caregiver physical or emotional abuse during ages 1-5 years and genetic risk for depression conferred by the opening genes. Genes involved in regulating sensitive periods are differentially expressed across the life course and may be implicated in depression vulnerability. Our findings about gene-by-development interplay motivate further research in large, more diverse samples to further unravel the complexity of depression etiology through a sensitive period lens.
Collapse
Affiliation(s)
- Yiwen Zhu
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Min-Jung Wang
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Alexandre A Lussier
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Kathryn A Davis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Christiaan de Leeuw
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anne E Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear and Department of Otorhinolaryngology and Head/Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Takao K Hensch
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jordan W Smoller
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Erin C Dunn
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Center on the Developing Child, Cambridge, MA, USA.
| |
Collapse
|
40
|
OUP accepted manuscript. Cereb Cortex 2022; 32:4619-4639. [DOI: 10.1093/cercor/bhab506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
|
41
|
Dion A, Muñoz PT, Franklin TB. Epigenetic mechanisms impacted by chronic stress across the rodent lifespan. Neurobiol Stress 2022; 17:100434. [PMID: 35198660 PMCID: PMC8841894 DOI: 10.1016/j.ynstr.2022.100434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 01/27/2023] Open
Abstract
Exposures to stress at all stages of development can lead to long-term behavioural effects, in part through changes in the epigenome. This review describes rodent research suggesting that stress in prenatal, postnatal, adolescent and adult stages leads to long-term changes in epigenetic regulation in the brain which have causal impacts on rodent behaviour. We focus on stress-induced epigenetic changes that have been linked to behavioural deficits including poor learning and memory, and increased anxiety-like and depressive-like behaviours. Interestingly, aspects of these stress-induced behavioural changes can be transmitted to offspring across several generations, a phenomenon that has been proposed to result via epigenetic mechanisms in the germline. Here, we also discuss evidence for the differential impact of stress on the epigenome in males and females, conscious of the fact that the majority of published studies have only investigated males. This has led to a limited picture of the epigenetic impact of stress, highlighting the need for future studies to investigate females as well as males.
Collapse
|
42
|
Gee DG. Early Adversity and Development: Parsing Heterogeneity and Identifying Pathways of Risk and Resilience. Am J Psychiatry 2021; 178:998-1013. [PMID: 34734741 DOI: 10.1176/appi.ajp.2021.21090944] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adversity early in life is common and is a major risk factor for the onset of psychopathology. Delineating the neurodevelopmental pathways by which early adversity affects mental health is critical for early risk identification and targeted treatment approaches. A rapidly growing cross-species literature has facilitated advances in identifying the mechanisms linking adversity with psychopathology, specific dimensions of adversity and timing-related factors that differentially relate to outcomes, and protective factors that buffer against the effects of adversity. Yet, vast complexity and heterogeneity in early environments and neurodevelopmental trajectories contribute to the challenges of understanding risk and resilience in the context of early adversity. In this overview, the author highlights progress in four major areas-mechanisms, heterogeneity, developmental timing, and protective factors; synthesizes key challenges; and provides recommendations for future research that can facilitate progress in the field. Translation across species and ongoing refinement of conceptual models have strong potential to inform prevention and intervention strategies that can reduce the immense burden of psychopathology associated with early adversity.
Collapse
Affiliation(s)
- Dylan G Gee
- Department of Psychology, Yale University, New Haven, Conn
| |
Collapse
|
43
|
Lengvenyte A, Olié E, Strumila R, Navickas A, Gonzalez Pinto A, Courtet P. Immediate and short-term efficacy of suicide-targeted interventions in suicidal individuals: A systematic review. World J Biol Psychiatry 2021; 22:670-685. [PMID: 33783294 DOI: 10.1080/15622975.2021.1907712] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To address the extreme suicide risk period following a suicidal crisis, we aimed to assess the current evidence for specific healthcare system-based interventions on suicide-related outcomes within one-week or one-month in individuals with current suicidal ideation (SI) or a recent suicide attempt (SA). METHODS We performed a database (Medline, Academic Search Complete, PsycARTICLES, the Cochrane library, PubMed) and manual reference search for randomised controlled trials, published between March 2000 and March 2020. Antisuicidal efficacy was defined as SI, SA, or a closely related concept. Quality was assessed with the Cochrane Risk of Bias 2 tool for randomised trials. RESULTS Out of 34 trials, five reported ketamine or esketamine superiority over placebo in reducing SI in depressed subjects within one week, while five studies had negative findings. Single trials reported positive results for one-month antisuicidal efficacy of buprenorphine, paroxetine, a crisis response plan, and assertive case management. Most trials were underpowered and had moderate-to-high risk of bias. CONCLUSIONS Preliminary mixed evidence suggests the possible utility of several pharmacological (ketamine, esketamine paroxetine, and buprenorphine) and non-pharmacological (a crisis response plan, and assertive case management) interventions. Only the immediate efficacy of ketamine was supported by multiple studies, and replication is needed.
Collapse
Affiliation(s)
- Aiste Lengvenyte
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,Faculty of Medicine, Institute of Clinical Medicine, Psychiatric Clinic, Vilnius University, Vilnius, Lithuania
| | - Emilie Olié
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,French Association of Biological Psychiatry and Neuropsychopharmacology
| | - Robertas Strumila
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,Faculty of Medicine, Institute of Clinical Medicine, Psychiatric Clinic, Vilnius University, Vilnius, Lithuania
| | - Alvydas Navickas
- Faculty of Medicine, Institute of Clinical Medicine, Psychiatric Clinic, Vilnius University, Vilnius, Lithuania
| | - Ana Gonzalez Pinto
- CIBERSAM; Bioaraba, Research Group on Severe Mental Illness; Osakidetza, Araba University Hospital, Psychiatry Service; Faculty of Medicine, Department of Neurosciences, University of the Basque Country UPV / EHU, Vitoria-Gasteiz, Spain.,Spanish Society of Biological Psychiatry
| | - Philippe Courtet
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, Montpellier, France.,IGF, Univ. Montpellier, CNRS, INSERM, Montpellier, France.,French Association of Biological Psychiatry and Neuropsychopharmacology
| |
Collapse
|
44
|
Wendel KM, Short AK, Noarbe BP, Haddad E, Palma AM, Yassa MA, Baram TZ, Obenaus A. Early life adversity in male mice sculpts reward circuits. Neurobiol Stress 2021; 15:100409. [PMID: 34746338 PMCID: PMC8554344 DOI: 10.1016/j.ynstr.2021.100409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/24/2021] [Accepted: 10/12/2021] [Indexed: 01/08/2023] Open
Abstract
Early life adversity (ELA) comprises a wide variety of negative experiences during early life and has been linked to cognitive impairments, reduced experiences of pleasure (anhedonia), and other long-term consequences implying that ELA impacts the reward circuitry. In this study, we focused on the projections from the dorsal raphe (DR) to the ventral tegmental area (VTA) and on to the nucleus accumbens (NAcc), an important pathway within the reward circuit. We hypothesized that ELA alters connectivity within the DR-VTA-NAcc pathway, associated with deficient reward seeking behaviors in adulthood. We used the limited bedding and nesting model to induce ELA in mice and measured reward-related behaviors in adulthood using the three-chamber social interaction and sucrose preference tests. High resolution ex vivo diffusion tensor imaging (DTI) was acquired and processed for regional DTI metrics, including tractography to assess circuit organization. We found brain-wide changes in radial diffusivity (RD) and altered connectivity of the reward circuit in the ELA group. DR-VTA-NAcc circuit tractography and axial diffusivity (AD) along this tract exhibited dispersed organization where AD was increased in the VTA segment. Behaviorally, ELA elicited a social anhedonia-like phenotype in adulthood with decreased direct social approach and time spent with peers in the three-chamber task, and no overt differences in sucrose preference. Our findings suggest that reward circuits, assessed using DTI, are altered following ELA and that these changes may reflect enduring reward deficits.
Collapse
Affiliation(s)
- Kara M. Wendel
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Annabel K. Short
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Brenda P. Noarbe
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Elizabeth Haddad
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Anton M. Palma
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Michael A. Yassa
- Department of Neurobiology and Behavior, University of California, Irvine School of Biological Sciences, Irvine, CA, USA
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Andre Obenaus
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, USA
| |
Collapse
|
45
|
Guadagno A, Belliveau C, Mechawar N, Walker CD. Effects of Early Life Stress on the Developing Basolateral Amygdala-Prefrontal Cortex Circuit: The Emerging Role of Local Inhibition and Perineuronal Nets. Front Hum Neurosci 2021; 15:669120. [PMID: 34512291 PMCID: PMC8426628 DOI: 10.3389/fnhum.2021.669120] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/29/2021] [Indexed: 01/10/2023] Open
Abstract
The links between early life stress (ELS) and the emergence of psychopathology such as increased anxiety and depression are now well established, although the specific neurobiological and developmental mechanisms that translate ELS into poor health outcomes are still unclear. The consequences of ELS are complex because they depend on the form and severity of early stress, duration, and age of exposure as well as co-occurrence with other forms of physical or psychological trauma. The long term effects of ELS on the corticolimbic circuit underlying emotional and social behavior are particularly salient because ELS occurs during critical developmental periods in the establishment of this circuit, its local balance of inhibition:excitation and its connections with other neuronal pathways. Using examples drawn from the human and rodent literature, we review some of the consequences of ELS on the development of the corticolimbic circuit and how it might impact fear regulation in a sex- and hemispheric-dependent manner in both humans and rodents. We explore the effects of ELS on local inhibitory neurons and the formation of perineuronal nets (PNNs) that terminate critical periods of plasticity and promote the formation of stable local networks. Overall, the bulk of ELS studies report transient and/or long lasting alterations in both glutamatergic circuits and local inhibitory interneurons (INs) and their associated PNNs. Since the activity of INs plays a key role in the maturation of cortical regions and the formation of local field potentials, alterations in these INs triggered by ELS might critically participate in the development of psychiatric disorders in adulthood, including impaired fear extinction and anxiety behavior.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Claudia Belliveau
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
46
|
Kreiker M, Perez K, Brown KL. The effects of early weaning on Pavlovian fear conditioning in young rats. Dev Psychobiol 2021; 63:e22133. [PMID: 34423435 DOI: 10.1002/dev.22133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 11/09/2022]
Abstract
Early life stress enhances memory for Pavlovian fear conditioning. Stress enhancements on fear conditioning following early weaning, however, have yet to be studied in periweaning rats. Early weaning is a relevant animal model for human early life trauma, and Pavlovian fear conditioning is useful for identifying links between stress-induced developmental changes and behavior. We hypothesized that early weaning-on postnatal day (P)15-would lead to higher levels of conditional freezing relative to rats weaned later in life. Periweaning rats were trained with a discrete conditional stimulus (CS) and a shock unconditional stimulus (US), and tested 1 or 15 days later. Enhanced retention was observed in early weaned rats receiving forward paired CS-US training in Experiment 1, though this did not replicate in the second experiment. Despite overall enhancements in early weaned rats in Experiment 1, infantile amnesia effects were not overcome in young rats tested 15 days after training. Enhanced freezing levels in early weaned rats were not observed in subjects receiving unpaired CS, US training, and sensitivity to the US was not different due to age at weaning. Potential mechanisms underlying weaning-related enhancements and considerations for future studies including the role of social transmission of fear information are discussed.
Collapse
Affiliation(s)
- Malaz Kreiker
- Department of Psychology and Neuroscience, Drake University, Des Moines, Iowa, USA
| | - Katelyn Perez
- Department of Psychology and Neuroscience, Drake University, Des Moines, Iowa, USA
| | - Kevin L Brown
- Department of Psychology and Neuroscience, Drake University, Des Moines, Iowa, USA
| |
Collapse
|
47
|
Macro- and Microscale Stress-Associated Alterations in Brain Structure: Translational Link With Depression. Biol Psychiatry 2021; 90:118-127. [PMID: 34001371 DOI: 10.1016/j.biopsych.2021.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023]
Abstract
Major depressive disorder (MDD) is a stress-related disorder associated with many cytoarchitectural and neurochemical changes. However, the majority of these changes cannot be reliably detected in the living brain. The examination of animal stress models and postmortem human brain tissue has significantly contributed to our understanding of the pathophysiology of MDD. Ronald Duman's work in humans and in rodent models was critical to the investigation of the contribution of synaptic deficits to MDD and chronic stress pathology, their role in the development and expression of depressive-like behavior, and reversal by novel drugs. Here, we review evidence from magnetic resonance imaging in humans and animals that suggests that corticolimbic alterations are associated with depression symptomatology. We also discuss evidence of cytoarchitectural alterations affecting neurons, astroglia, and synapses in MDD and highlight how similar changes are described in rodent chronic stress models and are linked to the emotion-related behavioral deficits. Finally, we report on the latest approaches developed to measure the synaptic and astroglial alterations in vivo, using positron emission tomography, and how it can inform on the contribution of MDD-associated cytoarchitectural alterations to the symptomatology and the treatment of stress-related disorders.
Collapse
|
48
|
Cross SJ, Leslie FM. Combined nicotine and ethanol age-dependently alter neural and behavioral responses in male rats. Behav Pharmacol 2021; 32:321-334. [PMID: 33660662 PMCID: PMC8119310 DOI: 10.1097/fbp.0000000000000622] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Use of alcohol (EtOH) and nicotine (Nic) typically begins during adolescence. Smoking and drinking often occur together and lead to a higher consumption of alcohol. Although we have shown that Nic+EtOH is reinforcing in self-administration tests in adolescent male rats, whether Nic+EtOH affects other behaviors or neuronal activity in an age-dependent manner is unknown. To address this, adolescent and adult male rats were given intravenous injections of Nic (30 µg/kg)+EtOH (4 mg/kg) and evaluated for locomotor and anxiety-like behaviors. Regional neuronal activity, assessed by cFos mRNA expression, was measured and used to evaluate functional connectivity in limbic regions associated with anxiety and motivation. Nic+EtOH increased locomotor activity and was anxiolytic in adolescents, but not adults. The posterior ventral tegmental area (pVTA), a critical regulator of drug reward, was selectively activated by Nic+EtOH in adults, while activity in its target region, the NAc-shell, was decreased. Drug-induced alterations in functional connectivity were more extensive in adults than adolescents and may act to inhibit behavioral responses to Nic+EtOH that are seen in adolescence. Overall, our findings suggest that brief, low-dose exposure to Nic+EtOH produces marked, age-dependent changes in brain and behavior and that there may be an ongoing maturation of the pVTA during adolescence that allows increased sensitivity to Nic+EtOH's reinforcing, hyperlocomotor, and anxiolytic effects. Furthermore, this work provides a potential mechanism for high rates of co-use of nicotine and alcohol by teenagers: this drug combination is anxiolytic and recruits functional networks that are unique from protective, inhibitory networks recruited in the mature and adult brain.
Collapse
Affiliation(s)
- Sarah J Cross
- Department of Anatomy and Neurobiology, School of Medicine
| | - Frances M Leslie
- Department of Anatomy and Neurobiology, School of Medicine
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, California, USA
| |
Collapse
|
49
|
McKibben LA, Dwivedi Y. Early-life stress induces genome-wide sex-dependent miRNA expression and correlation across limbic brain areas in rats. Epigenomics 2021; 13:1031-1056. [PMID: 34008410 PMCID: PMC8244583 DOI: 10.2217/epi-2021-0037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aims: The aim of this study was to assess regional- and sex-dependent changes in miRNA expression resulting from early-life stress (ELS). Materials and methods: Small RNA sequencing was used to determine sex-dependent changes in miRNAs after maternal separation, a rodent model of ELS, across the prefrontal cortex, amygdala and hippocampus. Results: Maternal separation induced anhedonia and altered miRNA expression in a sex-dependent manner, particularly in the prefrontal cortex and hippocampus. Gene ontology revealed that these miRNAs target genes with brain-specific biological functions. Conclusion: Using a network approach to explore miRNA signaling across the brain after ELS, regional differences were highlighted as key to studying the brain’s stress response, which indicates that sex is critical for understanding miRNA-mediated ELS-induced behavior.
Collapse
Affiliation(s)
- Lauren A McKibben
- Department of Psychiatry & Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Yogesh Dwivedi
- Department of Psychiatry & Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
50
|
Manns M, Basbasse YE, Freund N, Ocklenburg S. Paw preferences in mice and rats: Meta-analysis. Neurosci Biobehav Rev 2021; 127:593-606. [PMID: 34004244 DOI: 10.1016/j.neubiorev.2021.05.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Mice and rats are among the most common animal model species in both basic and clinical neuroscience. Despite their ubiquity as model species, many clinically relevant brain-behaviour relationships in rodents are not well understood. In particular, data on hemispheric asymmetries, an important organizational principle in the vertebrate brain, are conflicting as existing studies are often statistically underpowered due to small sample sizes. Paw preference is one of the most frequently investigated forms of hemispheric asymmetries on the behavioural level. Here, we used meta-analysis to statistically integrate findings on paw preferences in rats and mice. For both species, results indicate significant hemispheric asymmetries on the individual level. In mice, 81 % of animals showed a preference for either the left or the right paw, while 84 % of rats showed this preference. However, contrary to what has been reported in humans, population level asymmetries were not observed. These results are particularly significant as they point out that paying attention to potential individual hemispheric differences is important in both basic and clinical neuroscience.
Collapse
Affiliation(s)
- Martina Manns
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Germany.
| | - Yasmin El Basbasse
- Institute of Cognitive Neuroscience, Department Biopsychology, Faculty of Psychology, Ruhr University Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Germany
| | - Sebastian Ocklenburg
- Institute of Cognitive Neuroscience, Department Biopsychology, Faculty of Psychology, Ruhr University Bochum, Germany
| |
Collapse
|