1
|
Ramsaran AI, Ventura S, Gallucci J, De Snoo ML, Josselyn SA, Frankland PW. A sensitive period for the development of episodic-like memory in mice. Curr Biol 2025; 35:2032-2048.e3. [PMID: 40215964 PMCID: PMC12055481 DOI: 10.1016/j.cub.2025.03.032] [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: 10/28/2024] [Revised: 02/26/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Episodic-like memory is a later-developing cognitive function supported by the hippocampus. In mice, the formation of extracellular perineuronal nets in subfield cornu ammonis (CA) 1 of the dorsal hippocampus controls the emergence of episodic-like memory during the fourth post-natal week. Whether the timing of episodic-like memory onset is hard-wired, or flexibly set by early-life experiences during a critical or sensitive period for hippocampal maturation, is unknown. Here, we show that the trajectories for episodic-like memory development vary for mice given different sets of experiences spanning the second and third post-natal weeks. Specifically, episodic-like memory precision developed later in mice that experienced early-life adversity, while it developed earlier in mice that experienced early-life enrichment. Moreover, we demonstrate that early-life experiences set the timing of episodic-like memory development by modulating the pace of perineuronal net formation in dorsal CA1, which is dependent on the brain-derived neurotrophic factor (BDNF)-tropomysin receptor kinase B (TrkB) signaling pathway. These results indicate that the hippocampus undergoes a sensitive period during which early-life experiences determine the timing for episodic-like memory development.
Collapse
Affiliation(s)
- Adam I Ramsaran
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Silvia Ventura
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Julia Gallucci
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada
| | - Mitchell L De Snoo
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sheena A Josselyn
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Paul W Frankland
- Neurosciences & Mental Health Program, The Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Child & Brain Development Program, Canadian Institute for Advanced Research, 661 University Ave., Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
2
|
Rong Y, Yan W, Gao Z, Yang Y, Xu C, Zhang C. NRXN3-NLGN1 complex influences the development of depression induced by maternal separation in rats. Brain Res 2025; 1858:149659. [PMID: 40286836 DOI: 10.1016/j.brainres.2025.149659] [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: 10/24/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Early-life stress (ELS) increases the risk of major depressive disorder in children and adolescents. However, the molecular and cellular mechanisms of major depressive disorder (MDD) induced by ELS are poorly understood. Here, we establish a stress model in rats in which maternal separation stress (MS) during the postnatal period increases susceptibility to restraint stress (RS) later in life. In terms of mechanism, MS causes long-lasting synaptic plasticity alterations in rats, which is accompanied by reduced branch and spine lengths in the hippocampus. We identified the role of the cell adhesion factor neurexin 3 (NRXN3) and its ligand neuroligin 1 (NLGN1) as mediators of these effects. NRXN3 and NLGN1 downregulation in the hippocampus occurred prior to the observed synaptic changes and depression-related behaviors. In conclusion, NRXN3 is involved in the development of depression induced by maternal separation, and the specific mechanism involves the NRXN3-NLGN1 complex, which can mediate synaptic plasticity and increase susceptibility todepression.
Collapse
Affiliation(s)
- Yuru Rong
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China; Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou 221000, China; Department of Psychiatry, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China; The Key Lab of Psychiatry, Xuzhou Medical University, Xuzhou 221000, China
| | - Wanyue Yan
- Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou 221000, China; Department of Psychiatry, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China; The Key Lab of Psychiatry, Xuzhou Medical University, Xuzhou 221000, China
| | - Zheng Gao
- Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou 221000, China; Department of Psychiatry, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China; The Key Lab of Psychiatry, Xuzhou Medical University, Xuzhou 221000, China
| | - Yujing Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China; Department of Psychiatry, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China; The Key Lab of Psychiatry, Xuzhou Medical University, Xuzhou 221000, China
| | - Chengyun Xu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| | - Caiyi Zhang
- Department of Psychiatry, The Affiliated Xuzhou Oriental Hospital of Xuzhou Medical University, Xuzhou 221000, China; Department of Psychiatry, Second Clinical College, Xuzhou Medical University, Xuzhou 221000, China; The Key Lab of Psychiatry, Xuzhou Medical University, Xuzhou 221000, China.
| |
Collapse
|
3
|
Vlaikou AM, Nussbaumer M, Iliou A, Papageorgiou MP, Komini C, Theodoridou D, Benaki D, Mikros E, Gikas E, Syrrou M, Filiou MD. Early Life Stress Induces Brain Mitochondrial Dynamics Changes and Sex-Specific Adverse Effects in Adulthood. J Neurosci Res 2025; 103:e70023. [PMID: 40195806 DOI: 10.1002/jnr.70023] [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: 04/04/2023] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 04/09/2025]
Abstract
Early life stress exposure exerts detrimental effects in adulthood and is a risk factor for psychiatric disorders. Studies addressing the molecular mechanisms of early life stress have primarily focused on hormones and stress circuits. However, little is known on how mitochondria and mitochondrial dynamics (i.e., the orchestration of mitochondrial fission, fusion, mitophagy, and biogenesis) modulate early life stress responses. Here, we used a maternal separation with early weaning (MSEW) paradigm to investigate the behavioral and molecular early life stress-elicited effects in male and female C57BL/6 mice in adulthood. We first applied a behavioral test battery to assess MSEW-driven, anxiety-related and stress-coping alterations. We then looked for MSEW-induced, mitochondria-centered changes in cingulate cortex, hippocampus and cerebellum, as well as in plasma by combining protein, mRNA, mitochondrial DNA copy number (mtDNAcn) and metabolomics analyses. We found that MSEW mice are more anxious, show decreased antioxidant capacity in the cingulate cortex and have higher mRNA levels of the fission regulator Fis1 and the mitophagy activator Pink1 in the hippocampus, indicating a shift towards mitochondrial degradation. Hippocampal mRNA level alterations of apoptotic markers further suggest an MSEW-driven activation of apoptosis accompanied by a dysregulation of purine catabolism in the cerebellum in MSEW mice. Sex-specific analysis revealed distinct MSEW-induced changes in male and female mice at the molecular level. Our work reveals a previously unexplored role of mitochondrial dynamics in regulating early life stress effects and highlights a mitochondria-centered dysregulation as a persistent outcome of early life stress in adulthood.
Collapse
Affiliation(s)
- Angeliki-Maria Vlaikou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Markus Nussbaumer
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Aikaterini Iliou
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Maria P Papageorgiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Chrysoula Komini
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Daniela Theodoridou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitra Benaki
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Emmanuel Mikros
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Evangelos Gikas
- Section of Analytical Chemistry, Department of Chemistry, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Maria Syrrou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Michaela D Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina, University of Ioannina, Ioannina, Greece
| |
Collapse
|
4
|
He ZC, Yu YJ, Wang T, Yin HR, Sun YX, Liu X, Xie XM, Wang HL, Su YA, Li JT, Si TM. Early-life stress of limited bedding/nesting material induced recognition memory loss and decreased hippocampal VGluT1 and nectin3 levels in aged male mice. Pharmacol Biochem Behav 2025; 249:173980. [PMID: 39987993 DOI: 10.1016/j.pbb.2025.173980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Exposure to early-life stress has been found to lead to enduring psychiatric symptoms, including cognitive impairments that persist into adulthood and even old age. In this study, we investigated the behavioral effects and molecular changes of a well-established animal model of early-life stress, the limited bedding and nesting (LBN) model, in aged male mice. After 16 months, stressed mice showed a marked impairment in novel and spatial object recognition tasks, but not in temporal order memory or spatial working memory in the Y-maze spontaneous alternation task. These cognitive deficits were accompanied by a reduction in VGluT1 expression and a lower VGluT1/VGAT ratio in the CA1 region of the hippocampus, as well as reduced nectin3 expression in the mouse hippocampus. No significant molecular alterations were observed in the medial prefrontal cortex. These data support the notion that early-life stress leads to cognitive impairments in aged male mice, and these effects may be associated with a dysregulated excitatory/inhibitory balance and reduced nectin3 levels in the hippocampus.
Collapse
Affiliation(s)
- Ze-Cong He
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Ya-Jie Yu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Hui-Rong Yin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China; The First Affiliated Hospital of Xinxiang Medical College, 453100 Xinxiang, Henan, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xiao-Meng Xie
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Hong-Li Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
5
|
Ferreira-Rodrigues M, Sousa IS, Baptista FI, Coelho-Santos V. Stress in utero: prenatal dexamethasone exposure causes greater structural gliovascular alterations in female offspring than in males. Front Neurosci 2025; 19:1539867. [PMID: 40196234 PMCID: PMC11973320 DOI: 10.3389/fnins.2025.1539867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
From early in life, experiences like prenatal stress profoundly affect long-term health and behavior. Fetal exposure to increased levels of glucocorticoids (GC), via maternal stress or through antenatal corticosteroid therapy (commonly used in women at risk of preterm birth), can disrupt brain development and raise the susceptibility to psychiatric disorders. Previous studies on prenatal exposure to synthetic GCs, such as dexamethasone (DEX), revealed impairments in neurogenesis and dendritic spine development. However, the impact of prenatal stress, specifically antenatal DEX exposure, on the gliovascular interface remains unclear. This interface, involving the relationship between astrocytes and blood vessels, is essential for healthy brain development. Astrocytic endfeet coverage and organization are crucial features of the gliovascular interface, and in this study, we evaluated these aspects through aquaporin-4 (AQ4) expression and organization along the lectin labelled-vasculature. At Postnatal Day 14, no differences in AQ4 expression were observed between males and females. However, prenatal stress induced by DEX exposure (50 μg/kg was administered subcutaneously to pregnant mice through gestational days 16, 17 and 18) significantly impacted this structure in females but not in males. Specifically, in female offspring prenatally exposed to DEX, AQ4 expression was significantly upregulated in the hippocampus, and its rearrangement was observed in the prefrontal cortex. A comparison of vascular density between male and female brains showed no significant sex differences in any analyzed regions, though male cerebellar vessel segments were shorter. Interestingly, prenatal stress caused morphological alterations in female brains, including increased vessel tortuosity, while no such changes were seen in males. In the hippocampus, prenatal DEX exposure reduced vessel segment length in males but did not affect females. In the cerebellum, DEX exposure increased vessel segment length in females. This study highlights sex-specific differences in the impact of prenatal stress on the gliovascular structure across various brain regions, suggesting AQ4 as a potential molecular target relevant to depressive-like behaviors in female offspring. Future studies are needed to correlate the gliovascular structural alterations found with functional disturbances and sex-specific mental health issues.
Collapse
Affiliation(s)
- Magda Ferreira-Rodrigues
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
| | - Inês Santos Sousa
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
| | - Filipa I. Baptista
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, University of Coimbra, Coimbra, Portugal
| | - Vanessa Coelho-Santos
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, Institute of Physiology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
6
|
Callaghan RM, Yang H, Moloney RD, Waeber C. Behavioural assessment of neuropsychiatric outcomes in rodent stroke models. J Cereb Blood Flow Metab 2025:271678X251317369. [PMID: 40110694 PMCID: PMC11926818 DOI: 10.1177/0271678x251317369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 03/22/2025]
Abstract
Stroke-associated mood disorders are less recognised than sensorimotor impairment, despite their high prevalence. Similarly, few experimental stroke studies assess non-sensorimotor functions. This study examined the prevalence and implementation of non-sensorimotor tests in three stroke-focused journals over the last twenty years. Of 965 experimental ischaemic stroke papers which used behavioural testing in rodents, 932 included sensorimotor testing, while 137 used non-sensorimotor tests (most commonly the Morris water maze, open field, Y-maze, and novel object recognition tests, but with a more diverse range of tests introduced in recent years). Cognition, anxiety and depression were assessed in 70%, 27% and 3% of these 137 papers. Non-sensorimotor deficits were typically observed after recovery of sensorimotor function. Potential confounding factors and challenges for data interpretation were identified in the most prevalent tests. More generally, experimental rigor (a priori power calculation, randomisation, blinding, and pre-defined inclusion/exclusion) improved over the years, but remained unsatisfactory with only 26% of studies providing some evidence of adequate statistical power. Furthermore, most studies focused on male animals, limiting external validity. This review confirms the disparity between sensorimotor and non-sensorimotor testing in experimental stroke but shows that the share of the studies including the latter is increasing. It is essential that research into the neuropsychiatric sequalae of stroke addresses methodological issues noted and continues to expand to improve patient outcomes post-stroke.
Collapse
Affiliation(s)
- Robert M Callaghan
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| | - Huiyuan Yang
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| | - Rachel D Moloney
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
7
|
Birnie MT, Baram TZ. The evolving neurobiology of early-life stress. Neuron 2025:S0896-6273(25)00134-5. [PMID: 40101719 DOI: 10.1016/j.neuron.2025.02.016] [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: 12/10/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 03/20/2025]
Abstract
Because early-life stress is common and constitutes a strong risk factor for cognitive and mental health disorders, it has been the focus of a multitude of studies in humans and experimental models. Yet, we have an incomplete understanding of what is perceived as stressful by the developing brain, what aspects of stress influence brain maturation, what developmental ages are particularly vulnerable to stress, which molecules mediate the effects of stress on brain operations, and how transient stressful experiences can lead to enduring emotional and cognitive dysfunctions. Here, we discuss these themes, highlight the challenges and progress in resolving them, and propose new concepts and avenues for future research.
Collapse
Affiliation(s)
- Matthew T Birnie
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
8
|
Orso R, Viola TW, Heberle BA, Creutzberg KC, Lumertz FS, Grassi-Oliveira R. Sex-Specific Effects of Early-Life Stress Exposure on Memory Performance and the Medial Prefrontal Cortex Transcriptomic Pattern in Adolescent Mice. Mol Neurobiol 2025:10.1007/s12035-025-04803-x. [PMID: 40038196 DOI: 10.1007/s12035-025-04803-x] [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: 08/03/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Early life stress (ELS) is considered a risk factor for the development of cognitive and executive dysfunctions throughout development. The medial portion of the prefrontal cortex (mPFC) is directly implicated in short-term working memory. Furthermore, due to its late development compared to other brain regions, the mPFC is considered a vulnerable brain region to ELS exposure. Here, we investigated the effects of the ELS on PFC-dependent memory and mPFC transcriptomic profiles. From postnatal day (PND) 2 to PND 15, BALB/cJ mice were exposed to maternal separation (MS) for 3 h per day combined with limited bedding (ELS group) or left undisturbed (CT group). During the period of stress, maternal behavior was recorded pre-MS and post-MS. From PND 45 to PND 47, males and females were tested for working memory performance in the Y-maze and short-term recognition memory in the object in place task (OIP). Later, we assessed mRNA level alterations in the mPFC by RNA-seq. Here, we showed that ELS increases maternal care post-MS and the number of nest exits pre-MS and post-MS. Furthermore, males and females exposed to ELS exhibited impairments in the OIP, while only females performed worse in the Y-maze. With respect to the mPFC transcriptome, we identified 13 DEGs in the females, which were significantly influenced by chaperone-mediated protein folding processes, while 4 genes were altered in males. In conclusion, we showed that, compared with male sex, ELS alters maternal behavior and leads to more extensive impairments in memory function and transcriptomic alterations in females.
Collapse
Affiliation(s)
- Rodrigo Orso
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Thiago Wendt Viola
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, Brazil
| | | | | | | | - Rodrigo Grassi-Oliveira
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, Brazil.
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, A701-129, 8200, Aarhus, Denmark.
| |
Collapse
|
9
|
Sagae SC, Paz EDR, Zanardini B, Amaral AC, Bronczek GA, Koehler-Santos P, de Oliveira JR, Franci CR, Donadio MVF, Holman PJ, Raineki C. Alternate-day fasting differentially affects body composition, metabolic and immune response to fasting in male rats exposed to early-life adversity: Modulatory role of cafeteria diet. PLoS One 2025; 20:e0313103. [PMID: 40029907 PMCID: PMC11875342 DOI: 10.1371/journal.pone.0313103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 12/11/2024] [Indexed: 03/06/2025] Open
Abstract
The increased risk for obesity and metabolic disorders following early-life adversity is aggravated by poor diet (e.g., cafeteria diet). Alternate-day fasting (ADF) is a dietary regimen shown to improve immune and metabolic dysfunction related to obesity. Here, we evaluate if ADF can ameliorate the negative effects of early-life adversity and/or cafeteria diet on biological, immune and metabolic parameters. At weaning, animals reared under normal or adverse conditions (i.e., low bedding) were fed either standard chow or cafeteria diets ad libitum or subjected to an ADF regimen. In adulthood, we measured 24-hour fasted cholesterol, triglycerides, cytokines, oxidative stress markers, and body composition parameters including perigonadal, retroperitoneal, and brown fat pad weight. Animals exposed to early-life adversity respond differently to cafeteria diet and ADF. Adverse reared animals fed chow diet in the ADF regimen showed the largest reduction in body weight and perigonadal and retroperitoneal fat pad weight, the smallest increase in corticosterone levels, and the largest increase in TNF-α levels. However, the differential effects of the ADF regimen on body, perigonadal and retroperitoneal fat weight observed in adversely reared animals fed chow diet compared to controls were not present if the adversely reared animals were fed cafeteria diet in the ADF regimen. Furthermore, adversely reared animals fed cafeteria diet in the ADF regimen showed high IL-1β and IL-6 levels. Together, the data suggest that the altered vulnerability to metabolic and immune dysfunction following early-life adversity is not just due to the type of diet but also how the diet is consumed.
Collapse
Affiliation(s)
- Sara C. Sagae
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Edson D. R. Paz
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
- Departamento de Fisiologia Geral do Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Bárbara Zanardini
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Ana Claudia Amaral
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
| | - Gabriela A. Bronczek
- Centro de Ciências Biológicas e da Saúde, Universidade Estadual do Oeste do Paraná, Cascavel, Brazil
- Departmento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade de Campinas, Campinas, Brazil
| | - Patrícia Koehler-Santos
- Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Jarbas R. de Oliveira
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Celso R. Franci
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Márcio V. F. Donadio
- Laboratório de Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
- Departmento de Fisioterapia, Facultad de Medicina y Ciencias de la Salud, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Parker J. Holman
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
10
|
Tabbaa M, Gamez A, Dust A, Mataric M, Levitt P. Offspring genetic diversity regulates rearing experiences that predict differential susceptibility to Chd8 haploinsufficiency. RESEARCH SQUARE 2025:rs.3.rs-6058389. [PMID: 40092436 PMCID: PMC11908356 DOI: 10.21203/rs.3.rs-6058389/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Mouse models of human disease focus on determining the direct impact of genetic mutations on phenotypes related to clinical presentations. For example, loss of function mutations in the autism-associated CHD8 gene is highly penetrant for trait and behavioral abnormalities in children, but there is substantial clinical heterogeneity in the occurrence and extent of disruptions between individuals. Using a large genetic reference panel of mice, we recently showed that genetic background strongly regulates variability in trait disruptions caused by Chd8 haploinsufficiency. Here, we hypothesized that genetics could also impact the variability in response to early life experiences, thus contributing to differential susceptibility to neurodevelopmental disorders. To examine how genetic diversity impacts rearing experience, we systematically observed the behavior of genetically diverse offspring raised by genetically identical mothers. The results reveal strain differences in pup and maternal behaviors. Machine learning analysis reveals that early life litter experiences are strong predictors of sex-dependent postweaning social, anxiety-like, and cognitive trait disruptions due to Chd8 haploinsufficiency. The study suggests that offspring phenotypes in mutant models of disease are due to a combination of heritable and early experience factors, demonstrating the utility of incorporating genetic diversity in studies to model the mechanisms that underlie the heterogeneity of disrupted phenotypes in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Manal Tabbaa
- Division of Neurology, Department of Pediatrics and Developmental Neuroscience and Neurogenetics Program, Children's Hospital Los Angeles, The Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Alexis Gamez
- Division of Neurology, Department of Pediatrics and Developmental Neuroscience and Neurogenetics Program, Children's Hospital Los Angeles, The Saban Research Institute, Los Angeles, CA 90027, USA
| | - A'Di Dust
- Department of Computer Science, University of Southern California, Los Angeles, CA 90027, USA
| | - Maja Mataric
- Department of Computer Science, University of Southern California, Los Angeles, CA 90027, USA
| | - Pat Levitt
- Division of Neurology, Department of Pediatrics and Developmental Neuroscience and Neurogenetics Program, Children's Hospital Los Angeles, The Saban Research Institute, Los Angeles, CA 90027, USA
- Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
11
|
Mengelkoch S, Alley JC, Cole SW, Slavich GM. Transcriptional evidence of HPA axis dysregulation in adolescent females: Unique contributions of chronic early-life stressor exposure and maternal depression history. J Affect Disord 2025; 371:245-252. [PMID: 39532234 DOI: 10.1016/j.jad.2024.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/12/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Depression risk increases dramatically for adolescent females following the pubertal transition. Although chronic early-life stressor exposure and a maternal history of depression are established risk factors for depression onset in this population, we know little about the biological mechanisms underlying these associations. METHOD To investigate, we examined how chronic early-life stressor exposure and maternal depression history were associated with stress-related gene expression patterns, using a high-risk family design in 48 psychiatrically healthy adolescent females, 20 of whom had a mother with a lifetime history of depression. Lifetime chronic stressor exposure was assessed using the STRAIN and gene expression patterns were estimated using transcriptional profiling of whole blood. RESULTS Consistent with hypotheses, we found that adolescent females with greater chronic stressor exposure had higher NR3C1 expression levels compared to those with less chronic stressor exposure. Additionally, youth with a depressed mother had lower levels of FKBP5 expression compared to those without a depressed mother. Levels of FKBP5 expression, in turn, interacted with chronic stressor exposure to predict NR3C1 expression. Specifically, for those with low chronic stressor exposure, levels of FKBP5 and NR3C1 expression were strongly interrelated, whereas for those with high chronic stressor exposure, NR3C1 expression was high regardless of levels of FKBP5 expression. LIMITATIONS This study was correlational, the sample size was limited, and additional research is needed to elucidate the underlying mechanisms and predict who subsequently develops depression. CONCLUSIONS Notwithstanding these limitations, these data indicate that having low FKBP5 expression, alongside high NR3C1 expression, may be a potential preclinical marker of depression risk in adolescent females that warrants additional investigation.
Collapse
Affiliation(s)
- Summer Mengelkoch
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | - Jenna C Alley
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | - Steven W Cole
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| | - George M Slavich
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, Anacker C. Sex-specific and developmental effects of early life adversity on stress reactivity are rescued by postnatal knockdown of 5-HT 1A autoreceptors. Neuropsychopharmacology 2025; 50:507-518. [PMID: 39396089 PMCID: PMC11736140 DOI: 10.1038/s41386-024-01999-9] [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: 10/23/2023] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 10/14/2024]
Abstract
Early Life Adversity (ELA) predisposes to stress hypersensitivity in adulthood, but neurobiological mechanisms that protect from the enduring effects of ELA are poorly understood. Serotonin 1A (5HT1A) autoreceptors in the raphé nuclei regulate adult stress vulnerability, but whether 5HT1A could be targeted to prevent ELA effects on susceptibility to future stressors is unknown. Here, we exposed mice with postnatal knockdown of 5HT1A autoreceptors to the limited bedding and nesting model of ELA from postnatal day (P)3-10 and tested behavioral, neuroendocrine, neurogenic, and neuroinflammatory responses to an acute swim stress in male and female mice in adolescence (P35) and in adulthood (P56). In females, ELA decreased raphé 5HT neuron activity in adulthood and increased passive coping with the acute swim stress, corticosterone levels, neuronal activity, and corticotropin-releasing factor (CRF) levels in the paraventricular nucleus (PVN) of the hypothalamus. ELA also reduced neurogenesis in the ventral dentate gyrus (vDG) of the hippocampus, an important mediator of individual differences in stress susceptibility, and increased microglia activation in the PVN and vDG. These effects of ELA were specific to females and manifested predominantly in adulthood, but not earlier on in adolescence. Postnatal knockdown of 5HT1A autoreceptors prevented these effects of ELA on 5HT neuron activity, stress reactivity, neurogenesis, and neuroinflammation in adult female mice. Our findings demonstrate that ELA induces long-lasting and sex-specific impairments in the serotonin system, stress reactivity, and vDG function, and identify 5HT1A autoreceptors as potential targets to prevent these enduring effects of ELA.
Collapse
Affiliation(s)
- Rushell Dixon
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Lauren Malave
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Rory Thompson
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Serena Wu
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Yifei Li
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Noah Sadik
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA
| | - Christoph Anacker
- Division of Systems Neuroscience, Department of Psychiatry, Columbia University, and Research Foundation for Mental Hygiene, Inc. (RFMH), New York State Psychiatric Institute (NYSPI), New York, NY, 10032, USA.
- Columbia University Institute for Developmental Sciences, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), Department of Psychiatry, Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA.
- Columbia University Stem Cell Initiative (CSCI), Columbia University Irving Medical Center (CUIMC), New York, NY, 10032, USA.
| |
Collapse
|
13
|
Coleman EM, White M, Antonoudiou P, Weiss GL, Scarpa G, Stone B, Maguire J. Early life stress influences epilepsy outcomes in mice. Epilepsy Behav 2025; 163:110217. [PMID: 39689578 PMCID: PMC11830541 DOI: 10.1016/j.yebeh.2024.110217] [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: 09/18/2024] [Revised: 10/21/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Stress is a common seizure trigger that has been implicated in worsening epilepsy outcomes, which encompasses psychiatric and cognitive comorbidities and sudden unexpected death in epilepsy (SUDEP) risk. The neuroendocrine response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and HPA axis dysfunction worsens epilepsy outcomes, increasing seizure burden, behavioral comorbidities, and risk for SUDEP in mice. Early life stress (ELS) reprograms the HPA axis into adulthood, impacting both the basal and stress-induced activity. Thus, we propose that ELS may influence epilepsy outcomes by influencing the function of the HPA axis. To test this hypothesis, we utilized the maternal separation paradigm and examined the impact on seizure susceptibility. We show that ELS exerts a sex dependent effect on seizure susceptibility in response to acute administration of the chemoconvulsant, kainic acid, which is associated with an altered relationship between seizure activity and HPA axis function. To further examine the impact of ELS on epilepsy outcomes, we utilized the intrahippocampal kainic acid model of chronic epilepsy in mice previously exposed to maternal separation. We find that the relationship between corticosterone levels and the extent of epileptiform activity is altered in mice subjected to ELS. We demonstrate that ELS impacts behavioral outcomes associated with chronic epilepsy in a sex-dependent manner, with females being more affected. We also observe reduced mortality (presumed SUDEP) in female mice subjected to ELS, consistent with previous findings suggesting a role for HPA axis dysfunction in SUDEP risk. These data demonstrate for the first time that ELS influences epilepsy outcomes and suggest that previous life experiences may impact the trajectory of epilepsy.
Collapse
Affiliation(s)
- Emanuel M Coleman
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Maya White
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | | | - Grant L Weiss
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Garrett Scarpa
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Bradly Stone
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA
| | - Jamie Maguire
- Tufts University School of Medicine, Neuroscience Department, Boston, MA, USA.
| |
Collapse
|
14
|
Ma YN, Zhang CC, Sun YX, Liu X, Li XX, Wang H, Wang T, Wang XD, Su YA, Li JT, Si TM. Dorsal CA1 NECTIN3 Reduction Mediates Early-Life Stress-Induced Object Recognition Memory Deficits in Adolescent Female Mice. Neurosci Bull 2025; 41:243-260. [PMID: 39395912 PMCID: PMC11794733 DOI: 10.1007/s12264-024-01305-z] [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: 01/10/2024] [Accepted: 04/28/2024] [Indexed: 10/14/2024] Open
Abstract
Early-life stress (ES) leads to cognitive dysfunction in female adolescents, but the underlying neural mechanisms remain elusive. Recent evidence suggests that the cell adhesion molecules NECTIN1 and NECTIN3 play a role in cognition and ES-related cognitive deficits in male rodents. In this study, we aimed to investigate whether and how nectins contribute to ES-induced cognitive dysfunction in female adolescents. Applying the well-established limited bedding and nesting material paradigm, we found that ES impairs recognition memory, suppresses prefrontal NECTIN1 and hippocampal NECTIN3 expression, and upregulates corticotropin-releasing hormone (Crh) and its receptor 1 (Crhr1) mRNA levels in the hippocampus of adolescent female mice. Genetic experiments revealed that the reduction of dorsal CA1 (dCA1) NECTIN3 mediates ES-induced object recognition memory deficits, as knocking down dCA1 NECTIN3 impaired animals' performance in the novel object recognition task, while overexpression of dCA1 NECTIN3 successfully reversed the ES-induced deficits. Notably, prefrontal NECTIN1 knockdown did not result in significant cognitive impairments. Furthermore, acute systemic administration of antalarmin, a CRHR1 antagonist, upregulated hippocampal NECTIN3 levels and rescued object and spatial memory deficits in stressed mice. Our findings underscore the critical role of dCA1 NECTIN3 in mediating ES-induced object recognition memory deficits in adolescent female mice, highlighting it as a potential therapeutic target for stress-related psychiatric disorders in women.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, 100191, China.
| |
Collapse
|
15
|
Kim YJ, Lee YA. Social group size alters social behavior and dopaminergic and serotonergic systems. Soc Neurosci 2025; 20:1-15. [PMID: 39863521 DOI: 10.1080/17470919.2025.2454257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/12/2024] [Indexed: 01/27/2025]
Abstract
Social behavior is affected by social structure type, but how neural function changes with social type remains unclear. We investigated whether social group size affects social behaviors based on dopamine (DA) and serotonin (5-HT) systems. Four-week-old male mice were housed under different social group sizes: one, two, four, and eight mice per cage (1mpc, 2mpc, 4mpc, 8mpc, respectively). After 4 weeks, social preference, social interaction, and forced swim tests were performed to test sociability and anxiety, respectively, followed by analysis of DA and 5-HT and their metabolites (3,4-dihydroxyphenylacetic acid [DOPAC], 3-methoxytyramine [3-MT], norepinephrine, and 5-hydroxyindoleacetic acid [5-HIAA]). Social interactions and anxious behavior decreased with increased social group size. DA, 3-MT, and 5-HT levels decreased with increasing social group size, whereas DOPAC and 5-HIAA levels increased in the extended mesocorticolimbic system, including the dorsal striatum. Moreover, the increased social group size resulted in increased DOPAC/DA and 5-HIAA/5-HT ratios, accompanied by a decrease in the 3-MT/DA ratio within the extended mesocorticolimbic system. Linear regression analysis also revealed that social group size affects DA and 5-HT turnover. These suggest that social group size may influence behavior and monoamine levels, potentially contributing to DA- and 5-HT-related psychiatric disorders.
Collapse
Affiliation(s)
- Ye-Jin Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, Republic of Korea
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), Daegu, Republic of Korea
| | - Young-A Lee
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, Republic of Korea
| |
Collapse
|
16
|
Ciano Albanese N, Poggini S, Reccagni A, Barezzi C, Salciccia C, Poleggi A, Branchi I. Adolescent social isolation induces sex-specific behavioral and neural alterations. Psychoneuroendocrinology 2025; 172:107264. [PMID: 39721084 DOI: 10.1016/j.psyneuen.2024.107264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/11/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Adversities associated with isolation during adolescence, including the lack of appropriate emotional and social experiences, can jeopardize the individual development leading to the onset of mental illnesses such as major depressive disorder. Girls have higher rates of depression compared to boys; however, the relative contribution of biological and cultural factors to such a gender-dependent difference remains unclear. To identify the role of biological factors in this distinct susceptibility, we exposed adolescent C57BL/6 male and female mice (n = 12-14) to social isolation and we evaluated their behavioral responses, investigating both emotional and cognitive competencies during adolescence. In addition, we assessed alterations in immune activation and brain plasticity by measuring the expression levels of several pro- and anti-inflammatory cytokines and neural plasticity markers. Finally, we investigated the hormonal stress response by evaluating the hypothalamic-pituitary-adrenal (HPA) axis reactivity. Our findings indicate that adolescent social isolation leads to the development of depressive- and anxiety-like traits, although it impacts distinct behavioral domains with varying degrees in males and females. Both genders exhibited deficiencies in the emotional realm, displaying enhanced anhedonia and anxiety compared to their respective control groups. Moreover, males exhibited cognitive impairments linked to an altered HPA axis activity. Remarkably, social isolation influenced immune activation in both sexes, resulting in decreased pro-inflammatory markers. Overall, these results affirm the significant impact of social experiences during adolescence on neurodevelopment. Notably, our study reveals that both males and females are equally susceptible to the effects of isolation, although sex-specific differences become apparent in the alterations observed at both behavioral and physiological levels.
Collapse
Affiliation(s)
- Naomi Ciano Albanese
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Silvia Poggini
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy.
| | - Alice Reccagni
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy; PhD program in Clinical-experimental Neuroscience and Psychiatry, Sapienza University of Rome, Piazzale Aldo Moro, 5, Rome 00185, Italy
| | - Caterina Barezzi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Clara Salciccia
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Anna Poleggi
- Department of Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| |
Collapse
|
17
|
Orso R, Creutzberg KC, Begni V, Petrillo G, Cattaneo A, Riva MA. Emotional dysregulation following prenatal stress is associated with altered prefrontal cortex responsiveness to an acute challenge in adolescence. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111162. [PMID: 39383932 DOI: 10.1016/j.pnpbp.2024.111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Exposure to prenatal stress (PNS) has the potential to elicit multiple neurobiological alterations and increase the susceptibility to psychiatric disorders. Moreover, gestational stress may sensitize the brain toward an altered response to subsequent challenges. Here, we investigated the effects of PNS in rats and assessed whether these animals exhibit an altered brain responsiveness to an acute stress (AS) during adolescence. From gestational day 14 until delivery, Sprague Dawley dams were exposed to PNS or left undisturbed. During adolescence (PND38 to PND41), offspring were tested in the social interaction and splash test. At PND44 half of the animals were exposed to 5 min of forced swim stress. Males and Females exposed to PNS showed reduced sociability and increased anhedonic-like behavior. At the molecular level, exposure of adolescent rats to AS produced increased activation of the amygdala and ventral and dorsal hippocampus. Regarding the prefrontal cortex (PFC), we observed a pronounced activation in PNS males exposed to AS. Cell-type specific transcriptional analyses revealed a significant imbalance in the activation of PFC excitatory and inhibitory neurons in PNS males and females exposed to AS. Furthermore, stressed males exhibited disrupted HPA-axis function, while females showed impairments in the modulation of antioxidant genes. Our study shows that PNS induces emotional dysregulation and alters the responsiveness of the PFC to an acute stressor. Moreover, the disruption of excitatory and inhibitory balance during adolescence could influence the ability to respond to challenging events that may contribute to precipitate a full-blown pathologic condition.
Collapse
Affiliation(s)
- Rodrigo Orso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | | | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Giulia Petrillo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
18
|
Birmann PT, Sinott A, Zugno GP, Rodrigues RR, Conceição FR, Sousa FSS, Collares T, Seixas FK, Savegnago L. The antidepressant effect of Komagataella pastoris KM 71 H in maternal separation mice model mediated by the microbiota-gut-brain axis. Behav Brain Res 2025; 476:115287. [PMID: 39393682 DOI: 10.1016/j.bbr.2024.115287] [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: 06/06/2024] [Revised: 09/16/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND The intestinal microbiota plays a fundamental role in maintaining host health, especially during childhood, a critical period for its establishment. Early life stress can lead to shifts in gut microbiota composition, thus increasing the risk of major depressive disorder (MDD) in adulthood. The supplementation with probiotics restores intestinal permeability and the health of gut microbial communities, therefore being potential study targets for the treatment of MDD. In this sense, the yeast Komagataella pastoris was reported as a promising probiotic with antidepressant effect. METHODS Hence, the present study aims to investigate this effect in mice submitted to maternal separation (MS) 3 h per day from PND2 to PND14. Adult mice and mothers were treated with K. pastoris KM71H (8 log UFC.g-1/per animal, i.g.) or PBS (500 µl, i.g.) for 14 days. After behavioral tests, the animals were euthanized, followed by hippocampi and intestines removal for biochemical analysis. RESULTS On behavioral tests, K. pastoris KM71H treatment reduced the immobility time in TST of adult mice and increased the grooming activity in splash test of adult mice and mothers induced by MS. The probiotic treatment restored plasma corticosterone levels and glucocorticoid receptor expression in hippocampi, alongside nitrate/nitrite levels and superoxide dismutase activity in intestine, in addition to reducing reactive species levels in both structures. Moreover, it also normalized the fecal pH and water content of feces. CONCLUSION Thus, we conclude that K. pastoris KM71H is a promising therapeutic strategy for the treatment of MDD.
Collapse
Affiliation(s)
- Paloma T Birmann
- Neurobiotechnology Research Group, Postgraduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Airton Sinott
- Neurobiotechnology Research Group, Postgraduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Giuliana P Zugno
- Neurobiotechnology Research Group, Postgraduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Rafael R Rodrigues
- Applied Immunology Laboratory, Postgraduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabricio R Conceição
- Applied Immunology Laboratory, Postgraduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fernanda S S Sousa
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Postgraduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Tiago Collares
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Postgraduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabiana K Seixas
- Molecular and Cellular Oncology Research Group and Functional Genomics Laboratory, Postgraduate Program in Biotechnology, Technological Development Center, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Lucielli Savegnago
- Neurobiotechnology Research Group, Postgraduate Program in Biotechnology, Technologic Development Center, Federal University of Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
19
|
Bordes J, Bajaj T, Miranda L, van Doeselaar L, Brix LM, Narayan S, Yang H, Mitra S, Kovarova V, Springer M, Kleigrewe K, Müller-Myhsok B, Gassen NC, Schmidt MV. Sex-specific fear acquisition following early life stress is linked to amygdala and hippocampal purine and glutamate metabolism. Commun Biol 2024; 7:1684. [PMID: 39702524 DOI: 10.1038/s42003-024-07396-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
Early life stress (ELS) can negatively impact health, increasing the risk of stress-related disorders, such as post-traumatic stress disorder (PTSD). Importantly, PTSD disproportionately affects women, emphasizing the critical need to explore how sex differences influence the genetic and metabolic neurobiological pathways underlying trauma-related behaviors. This study uses the limited bedding and nesting (LBN) paradigm to model ELS and investigate its sex-specific effects on fear memory formation. Employing innovative unsupervised behavioral classification, the current study reveals distinct behavioral patterns associated with fear acquisition and retrieval in male and female mice following ELS. Females exposed to LBN display heightened active fear responses, contrasting with males. Furthermore, the study examined the crucial link between behavioral regulation and cellular metabolism in key brain regions involved in fear and stress processing. Sex-specific and stress-dependent alterations were observed in purine, pyrimidine, and glutamate metabolism within the basolateral amygdala, the dorsal hippocampus, and the ventral hippocampus. These findings provide crucial insights into the complex interplay between metabolic pathways, the neurobiological underpinnings of fear memory, and stress responses. Importantly, they emphasize the significance of considering sex-specific metabolic alterations when investigating stress-related disorders, opening potential avenues for the development of targeted interventions.
Collapse
Affiliation(s)
- Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Thomas Bajaj
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127, Bonn, Germany
| | - Lucas Miranda
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Lotte van Doeselaar
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Lea Maria Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Huanqing Yang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Shiladitya Mitra
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Veronika Kovarova
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Margherita Springer
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Bertram Müller-Myhsok
- Research Group Statistical Genetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Nils C Gassen
- Neurohomeostasis Research Group, Department of Psychiatry and Psychotherapy, Bonn Clinical Center, University of Bonn, 53127, Bonn, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
20
|
Hardy KA, Rybolt S, Patel B, Dye R, Rosen MJ. Characterizing Behavioral Effects of Early-Life Stress in an Animal Model of Auditory Processing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626725. [PMID: 39677688 PMCID: PMC11642929 DOI: 10.1101/2024.12.03.626725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Animal models provide significant insight into the development of typical and disordered sensory processing. Such models have been established to take advantage of physical and behavioral characteristics of specific species. For example, the Mongolian gerbil is a well-established model for auditory processing, with a hearing range similar in frequency to that of humans and an easily accessible cochlea. Recently, early-life stress (ELS) has been shown to affect sensory processing in auditory, visual, and somatosensory neural regions. To understand the functional impact of ELS, it is necessary to evaluate the susceptibility of sensory perceptual abilities to this early perturbation. Yet measuring sensory perception - e.g., using operant conditioning - often concurrently involves animal behavioral elements such as attention, memory, learning, and emotion. All of these elements are well-known to be impacted by ELS, and may affect behavioral measurements in ways that could be misconstrued as sensory deficits. Thus, it is critical to characterize which behavioral elements are affected by ELS in any sensory model. Here we induced ELS during a developmental time window for maturation of the auditory cortex in Mongolian gerbils. We conducted behavioral measures in juveniles, a developmental age when ELS is known to impair the auditory pathway. ELS had no effect on overall activity but reduced anxiety-related behavior, impaired recognition memory, and improved spatial memory, with some sex-specific effects. These effects may influence the ability of gerbils to learn and retain operant training, particularly if anxiety-provoking reinforcement is used.
Collapse
|
21
|
Gore IR, Brown CJ, Waters RC, Gould E. Social and nonsocial environmental loss have differential effects on ventral hippocampus-dependent behavior and inhibitory synaptic markers in adult male mice. Learn Mem 2024; 31:a053968. [PMID: 39681456 DOI: 10.1101/lm.053968.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/01/2024] [Indexed: 12/18/2024]
Abstract
In humans, psychological loss, whether social or nonsocial, can lead to clinical depression, anxiety disorders, and social memory impairments. Researchers have modeled combined social and nonsocial loss in rodents by transitioning them from social, enriched environments (EE) to individual housing, affecting behaviors related to avoidance, stress coping, and cognitive function. However, it remains unclear if these effects are driven by social or nonsocial loss. We examined the effects of nonsocial loss by housing adult male mice in EE before moving them to standard cages, where they were pair-housed, and compared this to mice experiencing complete social loss. Continuous EE reduced social investigation time while leaving social memory intact, also decreasing avoidance behavior. Nonsocial loss restored social investigation and avoidance behavior to control levels, while social loss impaired social memory and increased avoidance. In rodents, social memory and avoidance require ventral hippocampus (vHIP) neuronal oscillations, which involve parvalbumin-positive (PV+) inhibitory interneurons. We found decreased vHIP PV intensity in the social loss group, with no differences in the nonsocial loss group. Most PV+ cells are surrounded by perineuronal nets (PNNs) concentrating GABAA receptors in their lattice-like holes. Social loss decreased GABAA-δ expression, a subunit associated with extrasynaptic receptors, across PNN+ soma and in PNN holes, while nonsocial loss reduced gephyrin in these regions. These findings suggest social and nonsocial losses differentially affect vHIP function and behavior, with social loss having a more pronounced impact through mechanisms involving PV+ interneurons, PNN structure, and neurotransmitter receptor expression.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08542, USA
| | - Casey J Brown
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08542, USA
| | - Renée C Waters
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08542, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08542, USA
| |
Collapse
|
22
|
Jiang C, Ruiz-Sanchez I, Mei C, Pittenger C. Circuit mechanisms underlying sexually dimorphic outcomes of early life stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625736. [PMID: 39651173 PMCID: PMC11623607 DOI: 10.1101/2024.11.27.625736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Stress during early life influences brain development and can affect social, motor, and emotional processes. We describe a striking sex difference in the effects of early life stress (ELS), which produces anhedonia and anxiety-like behaviors in female adolescent mice, as reported previously, but repetitive behavioral pathology and social deficits in male adolescent mice. Notably, this parallels sex differences seen in the prevalence of psychiatric symptoms: depression and anxiety disorders are more common in girls and women, whereas neurodevelopmental disorders like autism spectrum disorder and Tourette syndrome are markedly more common in boys and men. We characterized the effects of ELS on the medial prefrontal cortex (mPFC) and on its projections to the dorsal striatum (dStr) and lateral septum (LS). ELS males, but not females, developed hyperactivity in the cortico-striatal circuit and hypoactivity in the cortico-septal circuit. Chemogenetic manipulation of cortico-striatal projection neurons modulates repetitive behavioral pathology and social behaviors in stressed males, and anhedonia in stressed females. Activation of cortico-septal projection neurons rescues social deficits in stressed males. We conclude that early life stress produces sexually dimorphic behavioral effects, with potential relevance to human psychiatric symptoms, through its differential effects on cortico-striatal and cortico-septal circuits.
Collapse
|
23
|
Ma YN, Yang CJ, Zhang CC, Sun YX, Yao XD, Liu X, Li XX, Wang HL, Wang H, Wang T, Wang XD, Zhang C, Su YA, Li JT, Si TM. Prefrontal parvalbumin interneurons mediate CRHR1-dependent early-life stress-induced cognitive deficits in adolescent male mice. Mol Psychiatry 2024:10.1038/s41380-024-02845-6. [PMID: 39578519 DOI: 10.1038/s41380-024-02845-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
Cognitive impairment, a core symptom of psychiatric disorders, is frequently observed in adolescents exposed to early-life stress (ES). However, the underlying neural mechanisms are unclear, and therapeutic efficacy is limited. Targeting parvalbumin-expressing interneurons (PVIs) in the medial prefrontal cortex (mPFC), we report that ES reduces mPFC PVI activity, which causally mediated ES-induced cognitive deficits in adolescent male mice through chemogenetic and optogenetic experiments. To understand the possible causes of PVI activity reduction following ES, we then demonstrated that ES upregulated corticotropin-releasing hormone (CRH) receptor 1 [CRHR1, mainly expressed in pyramidal neurons (PNs)] and reduced activity of local pyramidal neurons (PNs) and their excitatory inputs to PVIs. The subsequent genetic manipulation experiments (CRHR1 knockout, CRH overexpression, and chemogenetics) highlight that ES-induced PVI activity reduction may result from CRHR1 upregulation and PN activity downregulation and that PVIs play indispensable roles in CRHR1- or PN-mediated cognitive deficits induced by ES. These results suggest that ES-induced cognitive deficits could be attributed to the prefrontal CRHR1-PN-PVI pathway. Finally, treatment with antalarmin (a CRHR1 antagonist) and environmental enrichment successfully restored the PVI activity and cognitive deficits induced by ES. These findings reveal the neurobiological mechanisms underlying ES-induced cognitive deficits in adolescent male mice and highlight the therapeutic potentials of PVIs in stress-related cognitive deficits in adolescent individuals.
Collapse
Affiliation(s)
- Yu-Nu Ma
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Chao-Juan Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Chen-Chen Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ya-Xin Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xing-Duo Yao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao Liu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xue-Xin Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Hong-Li Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Han Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Ting Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
| |
Collapse
|
24
|
Ramsaran AI, Ventura S, Gallucci J, De Snoo ML, Josselyn SA, Frankland PW. A sensitive period for the development of episodic-like memory in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622296. [PMID: 39574753 PMCID: PMC11580884 DOI: 10.1101/2024.11.06.622296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Episodic-like memory is a later-developing cognitive function supported by the hippocampus. In mice, the formation of extracellular perineuronal nets in subfield CA1 of the dorsal hippocampus controls the emergence of episodic-like memory during the fourth postnatal week (Ramsaran et al., 2023). Whether the timing of episodic-like memory onset is hard-wired, or flexibly set by early-life experiences during a critical or sensitive period for hippocampal maturation, is unknown. Here, we show that the trajectories for episodic-like memory development vary for mice given different sets of experiences spanning the second and third postnatal weeks. Specifically, episodic-like memory precision developed later in mice that experienced early-life adversity, while it developed earlier in mice that experienced early-life enrichment. Moreover, we demonstrate that early-life experiences set the timing of episodic-like memory development by modulating the pace of perineuronal net formation in dorsal CA1. These results indicate that the hippocampus undergoes a sensitive period during which early-life experiences determine the timing for episodic-like memory development.
Collapse
Affiliation(s)
- Adam I Ramsaran
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Silvia Ventura
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Julia Gallucci
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mitchell L De Snoo
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
| | - Sheena A Josselyn
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Paul W Frankland
- Neurosciences & Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Child & Brain Development Program, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Rekapalli AK, Roman IC, Brenhouse HC, Cody CR. An adverse rearing environment alters maternal responsiveness to infant ultrasonic vocalizations. Int J Dev Neurosci 2024; 84:797-803. [PMID: 39003605 DOI: 10.1002/jdn.10367] [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: 03/16/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024] Open
Abstract
Rodent pups use a variety of ultrasonic vocalizations (USVs) to facilitate maternal care. Importantly, infant USV repertoires are dependent on both the age and early life experiences of the pups. We have shown that an adverse rearing environment modeled with the maternal separation (MS) paradigm alters caregiving behavior but little is known about how pup USVs differentially elicit maternal attention. In the present study, maternal approach towards a vocalizing pup over a non-vocalizing pup was tested in a Y-maze apparatus at two developmental time points over the course of MS. At postnatal day (P)10, MS dams engaged in longer interaction times with vocalizing pups compared to non-vocalizing pup, and this effect was strongest in male pups. As expected at P20, dams did not show a preference for either the vocalizing or non-vocalizing pups regardless of rearing environment; however, MS dams spent a greater amount of time in the center of the apparatus as compared to control dams, which can be interpreted as a measure of uncertainty or indecision. These effects of MS on dam USV sensitivity are important considering the sex specific effects of MS exposure across all developmental stages. Our novel findings support the hypothesis that sex-specific pup-dam interactions may drive later life outcomes following adversity.
Collapse
Affiliation(s)
| | - Isabel C Roman
- Psychology Department, Northeastern University, Boston, Massachusetts, USA
| | | | - Caitlyn R Cody
- Psychology Department, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
26
|
da Costa Rodrigues K, da Conceição Oliveira M, Dos Santos BF, de Campos Domingues NL, Fronza MG, Savegnago L, Wilhelm EA, Luchese C. Mechanisms involved in the antidepressant-like action of orally administered 5-((4-methoxyphenyl)thio)benzo[c][1,2,5]thiadiazole (MTDZ) in male and female mice. Psychopharmacology (Berl) 2024; 241:2385-2402. [PMID: 39008059 DOI: 10.1007/s00213-024-06647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
RATIONALE The compound 5-((4-methoxyphenyl)thio)benzo[c][1,2,5]thiadiazole (MTDZ) has recently been shown to inhibit in vitro acetylcholinesterase activity, reduce cognitive damage, and improve neuropsychic behavior in mice, making it a promising molecule to treat depression. OBJECTIVES This study investigated the antidepressant-like action of MTDZ in mice and its potential mechanisms of action. RESULTS Molecular docking assays were performed and suggested a potential inhibition of monoamine oxidase A (MAO-A) by MTDZ. The toxicity study revealed that MTDZ displayed no signs of toxicity, changes in oxidative parameters, or alterations to biochemistry markers, even at a high dose of 300 mg/kg. In behavioral tests, MTDZ administration reduced immobility behavior during the forced swim test (FST) without adjusting the climbing parameter, suggesting it has an antidepressant effect. The antidepressant-like action of MTDZ was negated with the administration of 5-HT1A, 5-HT1A/1B, and 5-HT3 receptor antagonists, implying the involvement of serotonergic pathways. Moreover, the antidepressant-like action of MTDZ was linked to the NO system, as L-arginine pretreatment inhibited its activity. The ex vivo assays indicated that MTDZ normalized ATPase activity, potentially linking this behavior to its antidepressant-like action. MTDZ treatment restricted MAO-A activity in the cerebral cortices and hippocampi of mice, proposing a selective inhibition of MAO-A associated with the antidepressant-like effect of the compound. CONCLUSIONS These findings suggest that MTDZ may serve as a promising antidepressant agent due to its selective inhibition of MAO-A and the involvement of serotonergic and NO pathways.
Collapse
Affiliation(s)
- Karline da Costa Rodrigues
- Research Laboratory in Biochemical Pharmacology (LaFarBio), Neurobiotechnology Research Group (GPN), Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Meliza da Conceição Oliveira
- Research Laboratory in Biochemical Pharmacology (LaFarBio), Neurobiotechnology Research Group (GPN), Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Beatriz Fuzinato Dos Santos
- Laboratory of Organic Catalysis and Biocatalysis, Federal University of Grande Dourados, Dourados, MS, Brazil
| | | | - Mariana Gallio Fronza
- Graduate Program in Biotechnology (GPN), Technological Development Center, Federal University of Pelotas (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Lucielli Savegnago
- Graduate Program in Biotechnology (GPN), Technological Development Center, Federal University of Pelotas (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Ethel Antunes Wilhelm
- Research Laboratory in Biochemical Pharmacology (LaFarBio), Neurobiotechnology Research Group (GPN), Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Cristiane Luchese
- Research Laboratory in Biochemical Pharmacology (LaFarBio), Neurobiotechnology Research Group (GPN), Center for Chemical, Pharmaceutical and Food Sciences, Federal University of Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
27
|
Demaili A, Portugalov A, Maroun M, Akirav I, Braun K, Bock J. Early life stress induces decreased expression of CB1R and FAAH and epigenetic changes in the medial prefrontal cortex of male rats. Front Cell Neurosci 2024; 18:1474992. [PMID: 39503008 PMCID: PMC11534599 DOI: 10.3389/fncel.2024.1474992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Several studies in both animal models and in humans have provided substantial evidence that early life stress (ELS) induces long-term changes in behavior and brain function, making it a significant risk factor in the aetiology of various mental disorders, including anxiety and depression. In this study, we tested the hypothesis that ELS in male rats (i) leads to increased anxiety and depressive-like symptoms; and (ii) that these behavioral changes are associated with functional alterations in the endocannabinoid system of the medial prefrontal cortex (mPFC). We further assessed whether the predicted changes in the gene expression of two key components of the endocannabinoid system, cannabinoid receptor 1 (CB1R) and the fatty acid amide hydrolase (FAAH), are regulated by epigenetic mechanisms. Behavioral profiling revealed that the proportion of behaviorally affected animals was increased in ELS exposed male rats compared to control animals, specifically showing symptoms of anhedonia and impaired social behavior. On the molecular level we observed a decrease in CB1R and FAAH mRNA expression in the mPFC of adult ELS exposed animals. These gene expression changes were accompanied by reduced global histone 3 acetylation in the mPFC, while no significant changes in DNA methylation and no significant changes of histone-acetylation at the promoter regions of the analyzed genes were detected. Taken together, our data provide evidence that ELS induces a long-term reduction of CB1R and FAAH expression in the mPFC of adult male rats, which may partially contribute to the ELS-induced changes in adult socio-emotional behavior.
Collapse
Affiliation(s)
- Arijana Demaili
- Department of Zoology/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 (IBBR), University of Haifa, Haifa, Israel
| | - Mouna Maroun
- The Integrated Brain and Behavior Research Center (IBBR), 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 (IBBR), University of Haifa, Haifa, Israel
| | - Katharina Braun
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Jörg Bock
- Department of Zoology/Developmental Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- PG Epigenetics and Structural Plasticity, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
28
|
Zhang AY, Elias E, Manners MT. Sex-dependent astrocyte reactivity: Unveiling chronic stress-induced morphological changes across multiple brain regions. Neurobiol Dis 2024; 200:106610. [PMID: 39032799 PMCID: PMC11500746 DOI: 10.1016/j.nbd.2024.106610] [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: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
Chronic stress is a major precursor to various neuropsychiatric disorders and is linked with increased inflammation in the brain. However, the bidirectional association between inflammation and chronic stress has yet to be fully understood. Astrocytes are one of the key inflammatory regulators in the brain, and the morphological change in reactive astrocytes serves as an important indicator of inflammation. In this study, we evaluated the sex-specific astrocyte response to chronic stress or systemic inflammation in key brain regions associated with mood disorders. We conducted the unpredictable chronic mild stress (UCMS) paradigm to model chronic stress, or lipopolysaccharide (LPS) injection to model systemic inflammation. To evaluate stress-induced morphological changes in astrocyte complexity, we measured GFAP fluorescent intensity for astrocyte expression, branch bifurcation by quantifying branch points and terminal points, branch arborization by conducting Sholl analysis, and calculated the ramification index. Our analysis indicated that chronic stress-induced morphological changes in astrocytes in all brain regions investigated. The effects of chronic stress were region and sex specific. Notably, females had greater stress or inflammation-induced astrocyte activation in the hypothalamus (HYPO), CA1, CA3, and amygdala (AMY) than males. These findings indicate that chronic stress induces astrocyte activation that may drive sex and region-specific effects in females, potentially contributing to sex-dependent mechanisms of disease.
Collapse
Affiliation(s)
- Ariel Y Zhang
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| | - Elias Elias
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| | - Melissa T Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| |
Collapse
|
29
|
Coleman EM, White M, Antonoudiou P, Weiss GL, Scarpa G, Stone B, Maguire J. Early life stress influences epilepsy outcomes in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612052. [PMID: 39314367 PMCID: PMC11419006 DOI: 10.1101/2024.09.09.612052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Stress is a common seizure trigger that has been implicated in worsening epilepsy outcomes. The neuroendocrine response to stress is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and HPA axis dysfunction worsens epilepsy outcomes, increasing seizure burden, behavioral comorbidities, and risk for sudden unexpected death in epilepsy (SUDEP) in mice. Early life stress (ELS) reprograms the HPA axis into adulthood, impacting both the basal and stress-induced activity. Thus, we propose that ELS may influence epilepsy outcomes by influencing the function of the HPA axis. To test this hypothesis, we utilized the maternal separation paradigm and examined the impact on seizure susceptibility. We show that ELS exerts a sex dependent effect on seizure susceptibility in response to acute administration of the chemoconvulsant, kainic acid, which is associated with an altered relationship between seizure activity and HPA axis function. To further examine the impact of ELS on epilepsy outcomes, we utilized the intrahippocampal kainic acid model of chronic epilepsy in mice previously exposed to maternal separation. We find that the relationship between corticosterone levels and the extent of epileptiform activity is altered in mice subjected to ELS. We demonstrate that ELS impacts behavioral outcomes associated with chronic epilepsy in a sex-dependent manner, with females being more affected. We also observe reduced mortality (presumed SUDEP) in female mice subjected to ELS, consistent with previous findings suggesting a role for HPA axis dysfunction in SUDEP risk. These data demonstrate for the first time that ELS influences epilepsy outcomes and suggest that previous life experiences may impact the trajectory of epilepsy.
Collapse
|
30
|
Dixon R, Malave L, Thompson R, Wu S, Li Y, Sadik N, Anacker C. Sex-specific and Developmental Effects of Early Life Adversity on Stress Reactivity are Rescued by Postnatal Knockdown of 5-HT 1A Autoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576344. [PMID: 38328253 PMCID: PMC10849559 DOI: 10.1101/2024.01.22.576344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Early Life Adversity (ELA) predisposes to stress hypersensitivity in adulthood, but neurobiological mechanisms that protect from the enduring effects of ELA are poorly understood. Serotonin 1A (5HT 1A ) autoreceptors in the raphé nuclei regulate adult stress vulnerability, but whether 5HT 1A could be targeted to prevent ELA effects on susceptibility to future stressors is unknown. Here, we exposed mice with postnatal knockdown of 5HT 1A autoreceptors to the limited bedding and nesting model of ELA from postnatal day (P)3-10 and tested behavioral, neuroendocrine, neurogenic, and neuroinflammatory responses to an acute swim stress in male and female mice in adolescence (P35) and in adulthood (P56). In females, ELA decreased raphé 5HT neuron activity in adulthood and increased passive coping with the acute swim stress, corticosterone levels, neuronal activity, and corticotropin-releasing factor (CRF) levels in the paraventricular nucleus (PVN) of the hypothalamus. ELA also reduced neurogenesis in the ventral dentate gyrus (vDG) of the hippocampus, an important mediator of individual differences in stress susceptibility, and increased microglia activation in the PVN and vDG. These effects of ELA were specific to females and manifested predominantly in adulthood, but not earlier on in adolescence. Postnatal knockdown of 5HT 1A autoreceptors prevented these effects of ELA on 5HT neuron activity, stress reactivity, neurogenesis, and neuroinflammation in adult female mice. Our findings demonstrate that ELA induces long-lasting and sex-specific impairments in the serotonin system, stress reactivity, and vDG function, and identify 5HT 1A autoreceptors as potential targets to prevent these enduring effects of ELA.
Collapse
|
31
|
Speranza L, Filiz KD, Lippiello P, Ferraro MG, Pascarella S, Miniaci MC, Volpicelli F. Enduring Neurobiological Consequences of Early-Life Stress: Insights from Rodent Behavioral Paradigms. Biomedicines 2024; 12:1978. [PMID: 39335492 PMCID: PMC11429222 DOI: 10.3390/biomedicines12091978] [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: 06/17/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Stress profoundly affects physical and mental health, particularly when experienced early in life. Early-life stress (ELS) encompasses adverse childhood experiences such as abuse, neglect, violence, or chronic poverty. These stressors can induce long-lasting changes in brain structure and function, impacting areas involved in emotion regulation, cognition, and stress response. Consequently, individuals exposed to high levels of ELS are at an increased risk for mental health disorders like depression, anxiety, and post-traumatic stress disorders, as well as physical health issues, including metabolic disorders, cardiovascular disease, and cancer. This review explores the biological and psychological consequences of early-life adversity paradigms in rodents, such as maternal separation or deprivation and limited bedding or nesting. The study of these experimental models have revealed that the organism's response to ELS is complex, involving genetic and epigenetic mechanisms, and is associated with the dysregulation of physiological systems like the nervous, neuroendocrine, and immune systems, in a sex-dependent fashion. Understanding the impact of ELS is crucial for developing effective interventions and preventive strategies in humans exposed to stressful or traumatic experiences in childhood.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Kardelen Dalim Filiz
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Pellegrino Lippiello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Silvia Pascarella
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| |
Collapse
|
32
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
33
|
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
|
34
|
Copenhaver AE, LeGates TA. Sex-Specific Mechanisms Underlie Long-Term Potentiation at Hippocampus→Medium Spiny Neuron Synapses in the Medial Shell of the Nucleus Accumbens. J Neurosci 2024; 44:e0100242024. [PMID: 38806250 PMCID: PMC11223474 DOI: 10.1523/jneurosci.0100-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp→NAc synapses is rewarding, and mice can establish learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigated sex differences in the mechanisms underlying Hipp→NAc LTP using whole-cell electrophysiology and pharmacology. We observed similarities in basal synaptic strength between males and females and found that LTP occurs postsynaptically with similar magnitudes in both sexes. However, key sex differences emerged as LTP in males required NMDA receptors (NMDAR), whereas LTP in females utilized an NMDAR-independent mechanism involving L-type voltage-gated Ca2+ channels (VGCCs) and estrogen receptor α (ERα). We also uncovered sex-similar features as LTP in both sexes depended on CaMKII activity and occurred independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders.
Collapse
Affiliation(s)
- Ashley E Copenhaver
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
| | - Tara A LeGates
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
35
|
Geertsema J, Kratochvil M, González-Domínguez R, Lefèvre-Arbogast S, Low D, Du Preez A, Lee H, Urpi-Sarda M, Sánchez-Pla A, Aigner L, Samieri C, Andres-Lacueva C, Manach C, Thuret S, Lucassen P, Korosi A. Coffee polyphenols ameliorate early-life stress-induced cognitive deficits in male mice. Neurobiol Stress 2024; 31:100641. [PMID: 38827176 PMCID: PMC11140806 DOI: 10.1016/j.ynstr.2024.100641] [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: 03/21/2024] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
Stress exposure during the sensitive period of early development has been shown to program the brain and increases the risk to develop cognitive deficits later in life. We have shown earlier that early-life stress (ES) leads to cognitive decline at an adult age, associated with changes in adult hippocampal neurogenesis and neuroinflammation. In particular, ES has been shown to affect neurogenesis rate and the survival of newborn cells later in life as well as microglia, modulating their response to immune or metabolic challenges later in life. Both of these processes possibly contribute to the ES-induced cognitive deficits. Emerging evidence by us and others indicates that early nutritional interventions can protect against these ES-induced effects through nutritional programming. Based on human metabolomics studies, we identified various coffee-related metabolites to be part of a protective molecular signature against cognitive decline in humans. Caffeic and chlorogenic acids are coffee-polyphenols and have been described to have potent anti-oxidant and anti-inflammatory actions. Therefore, we here aimed to test whether supplementing caffeic and chlorogenic acids to the early diet could also protect against ES-induced cognitive deficits. We induced ES via the limited nesting and bedding paradigm in mice from postnatal(P) day 2-9. On P2, mice received a diet to which 0.02% chlorogenic acid (5-O-caffeoylquinic acid) + 0.02% caffeic acid (3',4'-dihydroxycinnamic acid) were added, or a control diet up until P42. At 4 months of age, all mice were subjected to a behavioral test battery and their brains were stained for markers for microglia and neurogenesis. We found that coffee polyphenols supplemented early in life protected against ES-induced cognitive deficits, potentially this is mediated by the survival of neurons or microglia, but possibly other mechanisms not studied here are mediating the effects. This study provides additional support for the potential of early nutritional interventions and highlights polyphenols as nutrients that can protect against cognitive decline, in particular for vulnerable populations exposed to ES.
Collapse
Affiliation(s)
- J. Geertsema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - M. Kratochvil
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - R. González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - S. Lefèvre-Arbogast
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - D.Y. Low
- Université Clermont Auvergne, INRAE, UNH, F-63000, Clermont Ferrand, France
| | - A. Du Preez
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - H. Lee
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - M. Urpi-Sarda
- Biomarkers and Nutrimetabolomics Laboratory, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - A. Sánchez-Pla
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029, Madrid, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, 08028, Barcelona, Spain
| | - L. Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, 5020, Austria
| | - C. Samieri
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR 1219, F-33000, Bordeaux, France
| | - C. Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - C. Manach
- Université Clermont Auvergne, INRAE, UNH, F-63000, Clermont Ferrand, France
| | - S. Thuret
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 9NU, UK
| | - P.J. Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - A. Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Song N, Liu Z, Gao Y, Lu S, Yang S, Yuan C. NAc-DBS corrects depression-like behaviors in CUMS mouse model via disinhibition of DA neurons in the VTA. Mol Psychiatry 2024; 29:1550-1566. [PMID: 38361128 DOI: 10.1038/s41380-024-02476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
Major depressive disorder (MDD) is characterized by diverse debilitating symptoms that include loss of motivation and anhedonia. If multiple medications, psychotherapy, and electroconvulsive therapy fail in some patients with MDD, their condition is then termed treatment-resistant depression (TRD). MDD can be associated with abnormalities in the reward-system-dopaminergic mesolimbic pathway, in which the nucleus accumbens (NAc) and ventral tegmental area (VTA) play major roles. Deep brain stimulation (DBS) applied to the NAc alleviates the depressive symptoms of MDD. However, the mechanism underlying the effects of this DBS has remained elusive. In this study, using the chronic unpredictable mild stress (CUMS) mouse model, we investigated the behavioral and neurobiological effects of NAc-DBS on the multidimensional depression-like phenotypes induced by CUMS by integrating behavioral, in vivo microdialysis coupled with high-performance liquid chromatography-electrochemical detector (HPLC-ECD), calcium imaging, pharmacological, and genetic manipulation methods in freely moving mice. We found that long-term and repeated, but not single, NAc-DBS induced robust antidepressant responses in CUMS mice. Moreover, even a single trial NAc-DBS led to the elevation of the γ-aminobutyric acid (GABA) neurotransmitter, accompanied by the increase in dopamine (DA) neuron activity in the VTA. Both the inhibition of the GABAA receptor activity and knockdown of the GABAA-α1 gene in VTA-GABA neurons blocked the antidepressant effect of NAc-DBS in CUMS mice. Our results showed that NAc-DBS could disinhibit VTA-DA neurons by regulating the level of GABA and the activity of VTA-GABA in the VTA and could finally correct the depression-like behaviors in the CUMS mouse model.
Collapse
Affiliation(s)
- Nan Song
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Zhenhong Liu
- Institute for Brain Disorders, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Yan Gao
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shanshan Lu
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Shenglian Yang
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Chao Yuan
- Center of Cognition and Brain Science, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| |
Collapse
|
37
|
Yusupov N, Roeh S, Sotillos Elliott L, Chang S, Loganathan S, Urbina-Treviño L, Fröhlich AS, Sauer S, Ködel M, Matosin N, Czamara D, Deussing JM, Binder EB. DNA methylation patterns of FKBP5 regulatory regions in brain and blood of humanized mice and humans. Mol Psychiatry 2024; 29:1510-1520. [PMID: 38317011 PMCID: PMC11189813 DOI: 10.1038/s41380-024-02430-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/19/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024]
Abstract
Humanized mouse models can be used to explore human gene regulatory elements (REs), which frequently lie in non-coding and less conserved genomic regions. Epigenetic modifications of gene REs, also in the context of gene x environment interactions, have not yet been explored in humanized mouse models. We applied high-accuracy measurement of DNA methylation (DNAm) via targeted bisulfite sequencing (HAM-TBS) to investigate DNAm in three tissues/brain regions (blood, prefrontal cortex and hippocampus) of mice carrying the human FK506-binding protein 5 (FKBP5) gene, an important candidate gene associated with stress-related psychiatric disorders. We explored DNAm in three functional intronic glucocorticoid-responsive elements (at introns 2, 5, and 7) of FKBP5 at baseline, in cases of differing genotype (rs1360780 single nucleotide polymorphism), and following application of the synthetic glucocorticoid dexamethasone. We compared DNAm patterns in the humanized mouse (N = 58) to those in human peripheral blood (N = 447 and N = 89) and human postmortem brain prefrontal cortex (N = 86). Overall, DNAm patterns in the humanized mouse model seem to recapitulate DNAm patterns observed in human tissue. At baseline, this was to a higher extent in brain tissue. The animal model also recapitulated effects of dexamethasone on DNAm, especially in peripheral blood and to a lesser extent effects of genotype on DNAm. The humanized mouse model could thus assist in reverse translation of human findings in psychiatry that involve genetic and epigenetic regulation in non-coding elements.
Collapse
Affiliation(s)
- Natan Yusupov
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Simone Roeh
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Laura Sotillos Elliott
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Simon Chang
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Srivaishnavi Loganathan
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Anna S Fröhlich
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Susann Sauer
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Maik Ködel
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Darina Czamara
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
38
|
Kooiker CL, Birnie MT, Floriou-Servou A, Ding Q, Thiagarajan N, Hardy M, Baram TZ. Paraventricular Thalamus Neuronal Ensembles Encode Early-life Adversity and Mediate the Consequent Sex-dependent Disruptions of Adult Reward Behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591547. [PMID: 38746198 PMCID: PMC11092514 DOI: 10.1101/2024.04.28.591547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Early-life adversity increases risk for mental illnesses including depression and substance use disorders, disorders characterized by dysregulated reward behaviors. However, the mechanisms by which transient ELA enduringly impacts reward circuitries are not well understood. In mice, ELA leads to anhedonia-like behaviors in males and augmented motivation for palatable food and sex-reward cues in females. Here, the use of genetic tagging demonstrated robust, preferential, and sex-specific activation of the paraventricular nucleus of the thalamus (PVT) during ELA and a potentiated reactivation of these PVT neurons during a reward task in adult ELA mice. Chemogenetic manipulation of specific ensembles of PVT neurons engaged during ELA identified a role for the posterior PVT in ELA-induced aberrantly augmented reward behaviors in females. In contrast, anterior PVT neurons activated during ELA were required for the anhedonia-like behaviors in males. Thus, the PVT encodes adverse experiences early-in life, prior to the emergence of the hippocampal memory system, and contributes critically to the lasting, sex-modulated impacts of ELA on reward behaviors.
Collapse
Affiliation(s)
- Cassandra L. Kooiker
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - Amalia Floriou-Servou
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Qinxin Ding
- School of Biological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Neeraj Thiagarajan
- School of Biological Sciences, University of California-Irvine, Irvine, CA, USA
| | - Mason Hardy
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z. Baram
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
39
|
Gencturk S, Unal G. Rodent tests of depression and anxiety: Construct validity and translational relevance. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2024; 24:191-224. [PMID: 38413466 PMCID: PMC11039509 DOI: 10.3758/s13415-024-01171-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/03/2024] [Indexed: 02/29/2024]
Abstract
Behavioral testing constitutes the primary method to measure the emotional states of nonhuman animals in preclinical research. Emerging as the characteristic tool of the behaviorist school of psychology, behavioral testing of animals, particularly rodents, is employed to understand the complex cognitive and affective symptoms of neuropsychiatric disorders. Following the symptom-based diagnosis model of the DSM, rodent models and tests of depression and anxiety focus on behavioral patterns that resemble the superficial symptoms of these disorders. While these practices provided researchers with a platform to screen novel antidepressant and anxiolytic drug candidates, their construct validity-involving relevant underlying mechanisms-has been questioned. In this review, we present the laboratory procedures used to assess depressive- and anxiety-like behaviors in rats and mice. These include constructs that rely on stress-triggered responses, such as behavioral despair, and those that emerge with nonaversive training, such as cognitive bias. We describe the specific behavioral tests that are used to assess these constructs and discuss the criticisms on their theoretical background. We review specific concerns about the construct validity and translational relevance of individual behavioral tests, outline the limitations of the traditional, symptom-based interpretation, and introduce novel, ethologically relevant frameworks that emphasize simple behavioral patterns. Finally, we explore behavioral monitoring and morphological analysis methods that can be integrated into behavioral testing and discuss how they can enhance the construct validity of these tests.
Collapse
Affiliation(s)
- Sinem Gencturk
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey.
| |
Collapse
|
40
|
Zheng JY, Li XX, Liu X, Zhang CC, Sun YX, Ma YN, Wang HL, Su YA, Si TM, Li JT. Fluoxetine reverses early-life stress-induced depressive-like behaviors and region-specific alterations of monoamine transporters in female mice. Pharmacol Biochem Behav 2024; 237:173722. [PMID: 38336220 DOI: 10.1016/j.pbb.2024.173722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
The sex difference that females are more vulnerable to depression than males has been recently replicated in an animal model of early-life stress (ES) called the limited bedding and nesting material (LBN) paradigm. Adopting this animal model, we have previously examined the effects of ES on monoamine transporter (MATs) expression in stress-related regions in adult female mice, and the reversal effects of a novel multimodal antidepressant, vortioxetine. In this study, replacing vortioxetine with a classical antidepressant, fluoxetine, we aimed to replicate the ES effects in adult female mice and to elucidate the commonality and differences between fluoxetine and vortioxetine. We found that systemic 30-day treatment with fluoxetine successfully reversed ES-induced depression-like behaviors (especially sucrose preference) in adult female mice. At the molecular level, we largely replicated the ES effects, such as reduced serotonin transporter (SERT) expression in the amygdala and increased norepinephrine transporter (NET) expression in the medial prefrontal cortex (mPFC) and hippocampus. Similar reversal effects of fluoxetine and vortioxetine were observed, including SERT in the amygdala and NET in the mPFC, whereas different reversal effects were observed for NET in the hippocampus and vesicular monoamine transporters expression in the nucleus accumbens. Overall, these results demonstrate the validity of the LBN paradigm to induce depression-like behaviors in female mice, highlight the involvement of region-specific MATs in ES-induced depression-like behaviors, and provide insights for further investigation of neurobiological mechanisms, treatment, and prevention associated with depression in women.
Collapse
Affiliation(s)
- Jia-Ya Zheng
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Xue-Xin Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Xiao Liu
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Chen-Chen Zhang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Ya-Xin Sun
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Yu-Nu Ma
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Hong-Li Wang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Yun-Ai Su
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China.
| | - Ji-Tao Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing, China.
| |
Collapse
|
41
|
Noel ES, Chen A, Peña YA, Honeycutt JA. Early life adversity drives sex-dependent changes in 5-mC DNA methylation of parvalbumin cells in the prefrontal cortex in rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578313. [PMID: 38352518 PMCID: PMC10862911 DOI: 10.1101/2024.01.31.578313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Early life adversity (ELA) can result in increased risk for developing affective disorders, such as anxiety or depression, later in life, with women showing increased risk. Interactions between an individual's genes and their environment play key roles in producing, as well as mitigating, later life neuropathology. Our current understanding of the underlying epigenomic drivers of ELA associated anxiety and depression are limited, and this stems in part from the complexity of underlying biochemical processes associated with how early experiences shapes later life behavior. Epigenetic alterations, or experience-driven modifications to DNA, can be leveraged to understand the interplay between genes and the environment. The present study characterized DNA methylation patterning, assessed via evaluation of 5-methylcytosine (5-mC), following ELA in a Sprague Dawley rat model of ELA induced by early caregiver deprivation. This study utilized maternal separation to investigate sex- and age-specific outcomes of ELA on epigenetic patterning in parvalbumin (PV)-containing interneurons in the prefrontal cortex (PFC), a subpopulation of inhibitory neurons which are associated with ELA and affective dysfunction. While global analysis of 5-mC methylation and CpG site specific pyrosequencing of the PV promoter, Pvalb, showed no obvious effects of ELA, when analyses were restricted to assessing 5-mC intensity in colocalized PV cells, there were significant sex and age dependent effects. We found that ELA leads sex-specific changes in PV cell counts, and that cell counts can be predicted by 5-mC intensity, with males and females showing distinct patterns of methylation and PV outcomes. ELA also produced sex-specific effects in corticosterone reactivity, with juvenile females showing a blunted stress hormone response compared to controls. Overall, ELA led to a sex-specific developmental shift in PV profile, which is comparable to profiles that are seen at a later developmental timepoint, and this shift may be mediated in part by epigenomic alterations driven by altered DNA methylation.
Collapse
Affiliation(s)
- Emma S Noel
- Program in Biochemistry, Brunswick, ME 04011 USA
| | - Alissa Chen
- Program in Neuroscience, Brunswick, ME 04011 USA
| | | | - Jennifer A Honeycutt
- Program in Neuroscience, Brunswick, ME 04011 USA
- Department of Psychology Bowdoin College, Brunswick, ME 04011 USA
| |
Collapse
|
42
|
Lee SH, Jung EM. Adverse effects of early-life stress: focus on the rodent neuroendocrine system. Neural Regen Res 2024; 19:336-341. [PMID: 37488887 PMCID: PMC10503627 DOI: 10.4103/1673-5374.377587] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/28/2023] [Accepted: 04/24/2023] [Indexed: 07/26/2023] Open
Abstract
Early-life stress is associated with a high prevalence of mental illnesses such as post-traumatic stress disorders, attention-deficit/hyperactivity disorder, schizophrenia, and anxiety or depressive behavior, which constitute major public health problems. In the early stages of brain development after birth, events such as synaptogenesis, neuron maturation, and glial differentiation occur in a highly orchestrated manner, and external stress can cause adverse long-term effects throughout life. Our body utilizes multifaceted mechanisms, including neuroendocrine and neurotransmitter signaling pathways, to appropriately process external stress. Newborn individuals first exposed to early-life stress deploy neurogenesis as a stress-defense mechanism; however, in adulthood, early-life stress induces apoptosis of mature neurons, activation of immune responses, and reduction of neurotrophic factors, leading to anxiety, depression, and cognitive and memory dysfunction. This process involves the hypothalamus-pituitary-adrenal axis and neurotransmitters secreted by the central nervous system, including norepinephrine, dopamine, and serotonin. The rodent early-life stress model is generally used to experimentally assess the effects of stress during neurodevelopment. This paper reviews the use of the early-life stress model and stress response mechanisms of the body and discusses the experimental results regarding how early-life stress mediates stress-related pathways at a high vulnerability of psychiatric disorder in adulthood.
Collapse
Affiliation(s)
- Seung Hyun Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Eui-Man Jung
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
43
|
Breton JM, Cort Z, Demaestri C, Critz M, Nevins S, Downend K, Ofray D, Romeo RD, Bath KG. Early life adversity reduces affiliative behavior with a stressed cagemate and leads to sex-specific alterations in corticosterone responses in adult mice. Horm Behav 2024; 158:105464. [PMID: 38070354 PMCID: PMC10872397 DOI: 10.1016/j.yhbeh.2023.105464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Experiencing early life adversity (ELA) alters stress physiology and increases the risk for developing psychiatric disorders. The social environment can influence dynamics of stress responding and buffer and/or transfer stress across individuals. Yet, the impact of ELA on sensitivity to the stress of others and social behavior following stress is unknown. Here, to test the impact of ELA on social and physiological responses to stress, circulating blood corticosterone (CORT) and social behaviors were assessed in adult male and female mice reared under limited bedding and nesting (LBN) or control conditions. To induce stress, one cagemate of a pair-housed cage underwent a footshock paradigm and was then returned to their unshocked partner. CORT was measured in both groups of mice 20 or 90 min after stress exposure, and social behaviors were recorded and analyzed. ELA rearing influenced the CORT response to stress in a sex-specific manner. In males, both control and ELA-reared mice exhibited similar stress transfer to unshocked cagemates and similar CORT dynamics. In contrast, ELA females showed a heightened stress transfer to unshocked cagemates, and sustained elevation of CORT relative to controls, indicating enhanced stress contagion and a failure to terminate the stress response. Behaviorally, ELA females displayed decreased allogrooming and increased investigative behaviors, while ELA males showed reduced huddling. Together, these findings demonstrate that ELA influenced HPA axis dynamics, social stress contagion and social behavior. Further research is needed to unravel the underlying mechanisms and long-term consequences of ELA on stress systems and their impact on behavioral outcomes.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Columbia University, Department of Psychiatry, New York, NY, USA; New York State Psychiatric Institute, Division of Developmental Neuroscience, New York, NY, 10032, USA
| | - Zoey Cort
- Barnard College of Columbia University, Department of Neuroscience and Behavior, New York, NY, USA
| | - Camila Demaestri
- Columbia University, Department of Psychiatry, New York, NY, USA
| | - Madalyn Critz
- Columbia University, Department of Psychiatry, New York, NY, USA; New York State Psychiatric Institute, Division of Developmental Neuroscience, New York, NY, 10032, USA
| | - Samuel Nevins
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, Providence, RI, USA
| | - Kendall Downend
- Barnard College of Columbia University, Department of Neuroscience and Behavior, New York, NY, USA
| | - Dayshalis Ofray
- Columbia University, Department of Psychiatry, New York, NY, USA; New York State Psychiatric Institute, Division of Developmental Neuroscience, New York, NY, 10032, USA
| | - Russell D Romeo
- Barnard College of Columbia University, Department of Neuroscience and Behavior, New York, NY, USA
| | - Kevin G Bath
- Columbia University, Department of Psychiatry, New York, NY, USA; New York State Psychiatric Institute, Division of Developmental Neuroscience, New York, NY, 10032, USA.
| |
Collapse
|
44
|
Copenhaver AE, LeGates TA. Sex-specific mechanisms underlie long-term potentiation at hippocampus-nucleus accumbens synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575709. [PMID: 38293132 PMCID: PMC10827060 DOI: 10.1101/2024.01.15.575709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary in order to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp-NAc synapses is rewarding, and that mice can make learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigate sex differences in the mechanisms underlying Hipp-NAc LTP using whole-cell electrophysiology and pharmacology. We found that males and females display similar magnitudes of Hipp-NAc LTP which occurs postsynaptically. However, LTP in females requires L-type voltage-gated Ca 2+ channels (VGCC) for postsynaptic Ca 2+ influx, while males rely on NMDA receptors (NMDAR). Additionally, females require estrogen receptor α (ERα) activity for LTP while males do not. These differential mechanisms converge as LTP in both sexes depends on CAMKII activity and occurs independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral excitatory pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders. SIGNIFICANCE STATEMENT Strengthening of Hipp-NAc synapses drives reward-related behaviors. Male and female mice have similar magnitudes of long-term potentiation (LTP) and both sexes have a predicted postsynaptic locus of plasticity. Despite these similarities, we illustrate here that sex-specific molecular mechanisms are used to elicit LTP. Given the bidirectional relationship between Hipp-NAc synaptic strength in mediating reward-related behaviors, the use of distinct molecular mechanisms may explain sex differences observed in stress susceptibility or response to rewarding stimuli. Discovery and characterization of convergent sex differences provides mechanistic insight into the sex-specific function of Hipp-NAc circuitry and has widespread implications for circuits mediating learning and reward-related behavior.
Collapse
|
45
|
Holloway AL, Lerner TN. Hidden variables in stress neurobiology research. Trends Neurosci 2024; 47:9-17. [PMID: 37985263 PMCID: PMC10842876 DOI: 10.1016/j.tins.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Among the central goals of stress neurobiology research is to understand the mechanisms by which stressors change neural circuit function to precipitate or exacerbate psychiatric symptoms. Yet despite decades of effort, psychiatric medications that target the biological substrates of the stress response are largely lacking. We propose that the clinical advancement of stress response-based therapeutics for psychiatric disorders may be hindered by 'hidden variables' in stress research, including considerations of behavioral study design (stressors and outcome measures), individual variability, sex differences, and the interaction of the body's stress hormone system with endogenous circadian and ultradian rhythms. We highlight key issues and suggest ways forward in stress neurobiology research that may improve the ability to assess stress mechanisms and translate preclinical findings.
Collapse
Affiliation(s)
- Ashley L Holloway
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA
| | - Talia N Lerner
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Northwestern University Interdepartmental Neuroscience Program (NUIN), Evanston, IL, USA.
| |
Collapse
|
46
|
Gorthy AS, Balleste AF, Placeres-Uray F, Atkins CM. Chronic Stress in Early Development and Effects on Traumatic Brain Injury Outcome. ADVANCES IN NEUROBIOLOGY 2024; 42:179-204. [PMID: 39432043 PMCID: PMC11556197 DOI: 10.1007/978-3-031-69832-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
In recent years, significant advances have been made in the study of mild traumatic brain injury (mTBI). Complete recovery from mTBI normally requires days to weeks, yet a subset of the population suffers from symptoms for weeks to months after injury. The risk factors for these prolonged symptoms have not yet been fully understood. In this chapter, we address one proposed risk factor, early life stress (ELS) and its influence on mTBI recovery. To study the effects of ELS on mTBI recovery, accepted animal models of ELS, including maternal separation, limited bedding and nesting, and chronic unpredictable stress, have been implemented. Combining these ELS models with standardized mTBI models, such as fluid percussion injury or controlled cortical impact, has allowed for a deeper understanding of the neuronal, hormonal, and cognitive changes that occur after mTBI following ELS. These preclinical findings are being used to understand how adverse childhood experiences may predispose a subset of individuals to poorer recovery after mTBI.
Collapse
Affiliation(s)
- Aditi S Gorthy
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alyssa F Balleste
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fabiola Placeres-Uray
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
47
|
Martinez GJ, Appleton M, Kipp ZA, Loria AS, Min B, Hinds TD. Glucocorticoids, their uses, sexual dimorphisms, and diseases: new concepts, mechanisms, and discoveries. Physiol Rev 2024; 104:473-532. [PMID: 37732829 PMCID: PMC11281820 DOI: 10.1152/physrev.00021.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/07/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023] Open
Abstract
The normal stress response in humans is governed by the hypothalamic-pituitary-adrenal (HPA) axis through heightened mechanisms during stress, raising blood levels of the glucocorticoid hormone cortisol. Glucocorticoids are quintessential compounds that balance the proper functioning of numerous systems in the mammalian body. They are also generated synthetically and are the preeminent therapy for inflammatory diseases. They act by binding to the nuclear receptor transcription factor glucocorticoid receptor (GR), which has two main isoforms (GRα and GRβ). Our classical understanding of glucocorticoid signaling is from the GRα isoform, which binds the hormone, whereas GRβ has no known ligands. With glucocorticoids being involved in many physiological and cellular processes, even small disruptions in their release via the HPA axis, or changes in GR isoform expression, can have dire ramifications on health. Long-term chronic glucocorticoid therapy can lead to a glucocorticoid-resistant state, and we deliberate how this impacts disease treatment. Chronic glucocorticoid treatment can lead to noticeable side effects such as weight gain, adiposity, diabetes, and others that we discuss in detail. There are sexually dimorphic responses to glucocorticoids, and women tend to have a more hyperresponsive HPA axis than men. This review summarizes our understanding of glucocorticoids and critically analyzes the GR isoforms and their beneficial and deleterious mechanisms and the sexual differences that cause a dichotomy in responses. We also discuss the future of glucocorticoid therapy and propose a new concept of dual GR isoform agonist and postulate why activating both isoforms may prevent glucocorticoid resistance.
Collapse
Affiliation(s)
- Genesee J Martinez
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Malik Appleton
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Zachary A Kipp
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Terry D Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Barnstable Brown Diabetes Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
48
|
Yang EJ, Frolinger T, Iqbal U, Estill M, Shen L, Trageser KJ, Pasinetti GM. The role of the Toll like receptor 4 signaling in sex-specific persistency of depression-like behavior in response to chronic stress. Brain Behav Immun 2024; 115:169-178. [PMID: 37838079 PMCID: PMC11146676 DOI: 10.1016/j.bbi.2023.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/16/2023] Open
Abstract
Chronic stress is a major risk factor for Major Depressive Disorder (MDD), and it has been shown to impact the immune system and cause microglia activation in the medial prefrontal cortex (mPFC) involved in the pathogenesis of depression. The aim of this study is to further investigate cellular and molecular mechanisms underlying persistent depression behavior in sex specific manner, which is observed clinically. Here, we report that both male and female mice exhibited depression-like behavior following exposure to chronic stress. However, only female mice showed persistent depression-like behavior, which was associated with microglia activation in mPFC, characterized by distinctive alterations in the phenotype of microglia. Given these findings, to further investigate the underlying molecular mechanisms associated with persistent depression-like behavior and microglia activation in female mice, we used translating-ribosome affinity purification (TRAP). We find that Toll like receptor 4 (TLR4) signaling is casually related to persistent depression-like behavior in female mice. This is supported by the evidence that the fact that genetic ablation of TLR4 expression in microglia significantly reduced the persistent depression-like behavior to baseline levels in female mice. This study tentatively supports the hypothesis that the TLR4 signaling in microglia may be responsible for the sex differences in persistent depression-like behavior in female.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Tal Frolinger
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Umar Iqbal
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Molly Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kyle J Trageser
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Giulio M Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York 10468, United States.
| |
Collapse
|
49
|
Gersamia AG, Pochigaeva KI, Less YE, Akzhigitov RG, Guekht AB, Gulyaeva NV. [Gender characteristics of depressive disorders: clinical, psychological, neurobiological and translational aspects]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:7-16. [PMID: 38529858 DOI: 10.17116/jnevro20241240317] [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] [Indexed: 03/27/2024]
Abstract
Various studies have indicated that the prevalence of depression is almost twice as high among women as among men. A major factor associated with the development of depression and other affective disorders are adverse and psychologically traumatic life events that contribute to changes in the neuroendocrine system, altering the capacity to adapt to stress. These changes are involved in the pathogenesis of mental disorders, along with genetic and other factors, and are to a significant degree regulated by gender dependent mechanisms. While women have a high prevalence of depressive disorders, men show a higher rate of alcohol and substance abuse. These differences in the epidemiology are most likely explained by different predisposition to mental disorders in men and women and a diversity of biological consequences to adverse life events. Taking this into account, there is a need for a critical review of currently used approaches to modeling depressive disorders in preclinical studies, including the use of animals of both sexes. Adaptation of experimental models and protocols taking into account gender characteristics of neuroendocrine changes in response to stress, as well as structural-morphological, electrophysiological, molecular, genetic and epigenetic features, will significantly increase the translational validity of experimental work.
Collapse
Affiliation(s)
- A G Gersamia
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - K I Pochigaeva
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - Yu E Less
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - R G Akzhigitov
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | - A B Guekht
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - N V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology of RAS, Moscow, Russia
| |
Collapse
|
50
|
Gyles TM, Nestler EJ, Parise EM. Advancing preclinical chronic stress models to promote therapeutic discovery for human stress disorders. Neuropsychopharmacology 2024; 49:215-226. [PMID: 37349475 PMCID: PMC10700361 DOI: 10.1038/s41386-023-01625-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023]
Abstract
There is an urgent need to develop more effective treatments for stress-related illnesses, which include depression, post-traumatic stress disorder, and anxiety. We view animal models as playing an essential role in this effort, but to date, such approaches have generally not succeeded in developing therapeutics with new mechanisms of action. This is partly due to the complexity of the brain and its disorders, but also to inherent difficulties in modeling human disorders in rodents and to the incorrect use of animal models: namely, trying to recapitulate a human syndrome in a rodent which is likely not possible as opposed to using animals to understand underlying mechanisms and evaluating potential therapeutic paths. Recent transcriptomic research has established the ability of several different chronic stress procedures in rodents to recapitulate large portions of the molecular pathology seen in postmortem brain tissue of individuals with depression. These findings provide crucial validation for the clear relevance of rodent stress models to better understand the pathophysiology of human stress disorders and help guide therapeutic discovery. In this review, we first discuss the current limitations of preclinical chronic stress models as well as traditional behavioral phenotyping approaches. We then explore opportunities to dramatically enhance the translational use of rodent stress models through the application of new experimental technologies. The goal of this review is to promote the synthesis of these novel approaches in rodents with human cell-based approaches and ultimately with early-phase proof-of-concept studies in humans to develop more effective treatments for human stress disorders.
Collapse
Affiliation(s)
- Trevonn M Gyles
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|