1
|
Zhao X, Wang D, Chang M, He Z, Zeng Z, Ren M, Hu Y, Li Z. HPA-axis multilocus genetic interaction with stress life events in predicting changes in adolescent suicidal ideation. J Affect Disord 2025; 380:288-297. [PMID: 40139404 DOI: 10.1016/j.jad.2025.03.138] [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: 09/14/2024] [Revised: 01/15/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Research suggests that genetic variants linked to the hypothalamic-pituitary-adrenal (HPA) axis moderate the association between stressors and change in suicidal ideation, but examining gene-environment interactions with single polymorphisms limits power. This study investigates how interactions between stressful life events and HPA-axis polygenic risk predict changes in suicidal ideation. METHODS A sample of 760 adolescents (Mage = 14.15 ± 0.63 years; 54.6 % girls) was followed up over two years. The polygenic risk was assessed using a multilocus genetic profile score (MGPS) based on the additive effects of six HPA-axis candidate genes (SKS2, NR3C1, NR3C2, FKBP5). Participants were categorized into distinct trajectories of suicidal ideation based on time-varying changes in their ideation scores. Multivariate logistic regression analyses were conducted to identify predictors of trajectory membership. RESULTS (1) Five distinct trajectories of suicidal ideation were identified: resistance (52.7 %), persistence (13.1 %), delayed (11.4 %), remission (15.6 %), and relapsing (7.2 %). (2) The combination of HPA-axis MGPS and SLEs showed good predictive accuracy for suicidal ideation trajectories, as indicated by the area under the ROC curve. (3) The interaction between all six stressful life events and HPA-axis MGPS is more predictive of developing persistent trajectories of suicidal ideation. Meanwhile, HPA-axis MGPS interacted with factors such as interpersonal relationships, academic pressure, and loss to better predict the delayed and relapsing suicidal ideation compared to the resistance group. CONCLUSIONS This study suggests that genetic variants associated with the HPA axis exert a polygenic, additive effect on the relationship between stressful life events and longitudinal increases in suicidal ideation.
Collapse
Affiliation(s)
- Xian Zhao
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Dongfang Wang
- School of Psychology, Centre for Studies of Psychological Applications, Guangdong Key Laboratory of Mental Health and Cognitive Science, Ministry of Education Key Laboratory of Brain Cognition and Educational Science, South China Normal University, Guangzhou 510631, China
| | - Mengmeng Chang
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Zhen He
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Zihao Zeng
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Menghao Ren
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Yiqiu Hu
- School of Educational Science, Hunan Normal University, Changsha 410081, Hunan Province, China; China Research Center for Mental Health Education of Hunan Province, Hunan Normal University, Changsha 410081, Hunan Province, China.
| | - Zhihua Li
- Institute of Education, Hunan University of Science & Technology, Xiangtan 411201, Hunan Province, China.
| |
Collapse
|
2
|
Bi H, Jin J, Sun M, Chen M, Li X, Wang Y. Epigenetic changes caused by early life stress in the pathogenesis of depression. Eur J Pharmacol 2025; 999:177671. [PMID: 40288560 DOI: 10.1016/j.ejphar.2025.177671] [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: 12/13/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Major depressive disorder (MDD) is a severe psychiatric disorder with a complex and poorly understood pathogenesis. Epigenetics, a rapidly advancing field of biology, has been implicated in various psychiatric conditions, including schizophrenia, anxiety, substance addiction, and autism. Furthermore, substantial research indicates that epigenetic modifications play a crucial role in the etiology of depression. Early life stress (ELS) refers to adverse experiences occurring during prenatal development (e.g., maternal physical and mental health complications during pregnancy) and/or postnatal life (e.g., abuse, neglect, poverty, parental loss, family conflict, violence, and malnutrition). These early-life adversities can lead to epigenetic modifications, which, in turn, influence key biological processes and contribute to the pathogenesis of MDD. This review provides an overview of the regulatory mechanisms and functions of various epigenetic modifications, including non-coding RNAs, DNA methylation, and histone modifications. We then examine ELS-induced epigenetic alterations and their biological consequences, such as dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, neurogenesis, and neuroplasticity. Finally, we explore their potential implications for both the pathogenesis and treatment of MDD. We hypothesize that ELS-induced epigenetic changes may serve as biomarkers for MDD diagnosis and offer novel therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hongsheng Bi
- Department of Psychiatry, The First Hospital of China Medical University, China; The Third Hospital of Daqing, Psychiatric Ward No. 2, China
| | - Jingyan Jin
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingyuan Sun
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingrui Chen
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, China.
| | - Yan Wang
- Center for Psychological Development, China Medical University, China.
| |
Collapse
|
3
|
Wang M, Wei J, Yan Y, Du Y, Fan H, Dou Y, Zhao L, Ni R, Yang X, Ma X. Altered Brain Functional Connectivity and Peripheral Transcriptomic Profiles in Major Depressive Disorder With Childhood Maltreatment. Depress Anxiety 2025; 2025:6059502. [PMID: 40225733 PMCID: PMC11976050 DOI: 10.1155/da/6059502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Childhood maltreatment (CM) is a significant risk factor for major depressive disorder (MDD), yet the underlying biological mechanisms remain unclear. This study aimed to investigate brain functional networks and peripheral transcriptomics in patients with MDD who have a history of CM. Methods: Functional imaging data were collected and network-based statistics were used to identify differences in functional networks among MDD patients with CM (MDD_CM, n = 78), MDD patients without CM (MDD_nCM, n = 61), and healthy controls (HC, n = 126). Additionally, blood transcriptional data were clustered into co-expression modules, and module differential connectivity analysis was utilized to assess variations in gene co-expression network modules among the groups. Results: The results revealed a significant difference in an inferior occipital gyrus-centered functional network among the three groups. Furthermore, eight gene co-expression modules differed among the groups and were enriched in multiple branches related to immune responses or metabolic processes. Notably, a module enriched in type I interferon-related signaling pathways demonstrated a significant correlation with the disrupted network in the MDD_nCM group. Moreover, multiple immune-related gene modules were found to be significantly correlated with sleep disturbances in MDD_CM patients. Conclusions: Dysregulation of an inferior occipital gyrus-centered functional network and immune-related transcriptomic alterations significantly associate with the pathophysiology of MDD_CM.
Collapse
Affiliation(s)
- Min Wang
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jinxue Wei
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yushun Yan
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yue Du
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Huanhuan Fan
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yikai Dou
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liansheng Zhao
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rongjun Ni
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiao Yang
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaohong Ma
- Mental Health Center and Institute of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Watson CB, Sharpley CF, Bitsika V, Evans I, Vessey K. A Systematic Review and Meta-Analysis of the Association Between Childhood Maltreatment and Adult Depression. Acta Psychiatr Scand 2025; 151:572-599. [PMID: 40025916 PMCID: PMC11962359 DOI: 10.1111/acps.13794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Childhood maltreatment (CM) and depression are serious global issues with high prevalence and lifelong impacts on physical and mental health. CM has been proposed as a modifiable risk factor for depression that, if prevented, may contribute to a reduction in the global incidence of depressive disorders. Despite this, there is a paucity of reviews examining the strength of the association between these variables. The aim of this systematic review and meta-analysis was to evaluate the empirical evidence and determine if CM is supported as a preventable risk factor for depression. METHODS A search was performed in July 2024 for all peer-reviewed journal articles written in English examining the relationship between CM and adult depression in the electronic databases EBSCOhost, Proquest, and Embase. Studies were included in this review if they measured maltreatment before 18 years of age as the independent variable and adult depression as the dependent variable. Studies were excluded if the outcome variable was grouped with comorbidity and if they did not report primary quantitative data. A total of 77 studies with 516,302 participants met the inclusion criteria for review. RESULTS A random-effects meta-analysis was used to generate a pooled odds ratio from 87 effect estimates and demonstrated that individuals with a history of any CM are 2.5 times more likely to experience adult depression (OR = 2.49 [95% CI: 2.25-2.76]). This increase in odds remained regardless of how the primary studies screened for depression. CONCLUSIONS These findings confirmed the strong association between the experience of CM and adult depression. High heterogeneity in the meta-analytic results also suggested that further research is required that applies consistent adjustments for comorbidities and confounding factors and examines the temporal relationship between the variables to establish causality.
Collapse
Affiliation(s)
- Christopher B. Watson
- Brain Behaviour Research GroupUniversity of New EnglandArmidaleNew South WalesAustralia
| | | | - Vicki Bitsika
- Brain Behaviour Research GroupUniversity of New EnglandArmidaleNew South WalesAustralia
| | - Ian Evans
- Brain Behaviour Research GroupUniversity of New EnglandArmidaleNew South WalesAustralia
| | - Kirstan Vessey
- Brain Behaviour Research GroupUniversity of New EnglandArmidaleNew South WalesAustralia
| |
Collapse
|
5
|
Salles J, Lin R, Turecki G. Small Nucleolar RNAs and the Brain: Growing Evidence Supporting Their Role in Psychiatric Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100415. [PMID: 39867567 PMCID: PMC11758842 DOI: 10.1016/j.bpsgos.2024.100415] [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: 08/20/2024] [Revised: 10/20/2024] [Accepted: 10/28/2024] [Indexed: 01/28/2025] Open
Abstract
Noncoding RNAs comprise most of the transcriptome and represent an emerging area of research. Among them, small nucleolar RNAs (snoRNAs) have emerged as a promising target because they have been associated with the development and evolution of several diseases, including psychiatric disorders. snoRNAs are expressed in the brain, with some showing brain-specific expression that indicates specific roles in brain development, function, and dysfunction. However, the role of snoRNAs in conditions that affect the brain needs further investigation to be better understood. This scoping review summarizes existing literature on studies that have investigated snoRNAs in psychiatry and offers insight into potential pathophysiological mechanisms to be further investigated in future research.
Collapse
Affiliation(s)
- Juliette Salles
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Rixing Lin
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Jamil S, Raza ML, Moradikor N, Haghipanah M. Early life stress and brain development: Neurobiological and behavioral effects of chronic stress. PROGRESS IN BRAIN RESEARCH 2025; 291:49-79. [PMID: 40222792 DOI: 10.1016/bs.pbr.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Early life stress is the term used to describe a variety of traumatic events that a person may have as a kid, such as being subjected to domestic or public violence, being neglected, experiencing parental conflict, being abused physically, emotionally and sexually. These events have the potential to seriously impair the brains normal growth and development, which could have long term psychological and physiological repercussions. Early life stress (ELS) has profound and enduring effects on brain development, contributing to long-term neurological and behavioral changes. Neurologically, ELS can reduce hippocampal volume, impairing memory and emotional regulation, while also sensitizing the amygdala, leading to exaggerated fear and anxiety responses. Additionally, ELS can disrupt the development of the prefrontal cortex (PFC), affecting decision-making, planning, and impulse control. It also alters neurotransmitter systems, such as serotonin and dopamine, influencing mood and motivation, and can trigger chronic neuroinflammation, increasing the risk of neurodegenerative diseases. Behaviorally, ELS heightens the risk of anxiety, depression, and impulsivity, and can contribute to conditions like ADHD and substance abuse Social and emotional difficulties, such as challenges in relationships and empathy, often arise, along with cognitive impairments in learning and memory. Furthermore, ELS increases stress responsiveness, making individuals more vulnerable to future stress. However, these effects can be mitigated by supportive environments and targeted interventions.
Collapse
Affiliation(s)
- Subia Jamil
- Department of Pharmacology, Faculty of Pharmacy, Jinnah University for Women, Karachi, Pakistan.
| | - Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| | - Motahareh Haghipanah
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| |
Collapse
|
7
|
Elam KK, Su J, Qin WA, Lemery-Chalfant K. Polygenic risk for epigenetic aging and adverse life experiences interact to predict growth in adolescent depression in a racially/ethnically diverse sample. Front Psychiatry 2024; 15:1499395. [PMID: 39758447 PMCID: PMC11695374 DOI: 10.3389/fpsyt.2024.1499395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Research has yet to examine the interplay between indices of environmental risk and resilience processes and genetic predisposition for epigenetic aging in predicting early adolescent depressive symptoms. In the current study we examine whether adverse life events and parental acceptance moderate polygenic predisposition for GrimAge epigenetic aging in predicting trajectories of depressive symptoms across early adolescence. Method Using data from the Adolescent Brain Development Study (ABCD, N = 11,875), we created polygenic scores for GrimAge, and examined whether exposure to adverse life events and parental acceptance moderated the relation between genetic risk and depressive symptom trajectories from age 10/11 to 12/13 using growth mixture modelling. We examined models separately in European American (EA), African American (AA), and Latinx (LX) subgroups of ABCD. Results In the EA and AA subgroups, adverse life events moderated polygenic scores for GrimAge such that there was increased likelihood of membership in a higher vs. lower depression trajectory. Discussion We extend literature by identifying genetic contributions to epigenetic aging as a depression diathesis in adolescence. Findings also highlight the detrimental role of adverse life events in exacerbating genetic risk for the development of depression in adolescence.
Collapse
Affiliation(s)
- Kit K. Elam
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, United States
| | - Jinni Su
- Psychology Department, Arizona State University, Tempe, AZ, United States
| | - Weisiyu Abraham Qin
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, United States
| | | |
Collapse
|
8
|
Portugalov A, Akirav I. FAAH Inhibition Reverses Depressive-like Behavior and Sex-Specific Neuroinflammatory Alterations Induced by Early Life Stress. Cells 2024; 13:1881. [PMID: 39594629 PMCID: PMC11593135 DOI: 10.3390/cells13221881] [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: 10/01/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Early life stress (ELS) increases predisposition to major depressive disorder (MDD), with neuroinflammation playing a crucial role. This study investigated the long-term effects of the fatty acid amide hydrolase (FAAH) inhibitor URB597 on ELS-induced depressive-like behavior and messenger RNA (mRNA) of pro-inflammatory cytokines in the medial prefrontal cortex (mPFC) and CA1 regions. We also assessed whether these gene expression alterations were present at the onset of URB597 treatment during late adolescence. ELS induced a depressive-like phenotype in adult male and female rats, which was reversed by URB597. In the mPFC, ELS downregulated nuclear factor kappa B1 (nfκb1) in both sexes, while URB597 normalized this expression exclusively in males. In females, ELS downregulated interleukin (il) 6 and tumor necrosis factor alpha (tnfα) but upregulated il1β and corticotropin-releasing factor (crf); URB597 normalized il6, il1β, and crf. In the CA1, ELS downregulated il1β and tnfα in males and upregulated il1β expression in females, which was reversed by URB597. Some of these effects began in late adolescence, including mPFC-nfκb1 expression in both sexes, mPFC-il6 and mPFC-il1β in females, CA1-il1β and CA1-tnfα in males, and CA1-il1β in females. These findings highlight URB597 as a therapeutic approach for reversing ELS-induced depressive-like behavior by associating with changes in the gene expression of neuroinflammatory cytokines, with notable sex differences.
Collapse
Affiliation(s)
- Anna Portugalov
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- School of Psychological Sciences, Department of Psychology, University of Haifa, Haifa 3498838, Israel;
- The Integrated Brain and Behavior Research Center (IBBRC), University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
9
|
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
|
10
|
Carvalho Silva R, Martini P, Hohoff C, Mattevi S, Bortolomasi M, Menesello V, Gennarelli M, Baune BT, Minelli A. DNA methylation changes in association with trauma-focused psychotherapy efficacy in treatment-resistant depression patients: a prospective longitudinal study. Eur J Psychotraumatol 2024; 15:2314913. [PMID: 38362742 PMCID: PMC10878335 DOI: 10.1080/20008066.2024.2314913] [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: 08/29/2023] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Background: Stressful events increase the risk for treatment-resistant depression (TRD), and trauma-focused psychotherapy can be useful for TRD patients exposed to early life stress (ELS). Epigenetic processes are known to be related to depression and ELS, but there is no evidence of the effects of trauma-focused psychotherapy on methylation alterations.Objective: We performed the first epigenome-wide association study to investigate methylation changes related to trauma-focused psychotherapies effects in TRD patients.Method: Thirty TRD patients assessed for ELS underwent trauma-focused psychotherapy, of those, 12 received trauma-focused cognitive behavioural therapy, and 18 Eye Movement Desensitization and Reprocessing (EMDR). DNA methylation was profiled with Illumina Infinium EPIC array at T0 (baseline), after 8 weeks (T8, end of psychotherapy) and after 12 weeks (T12 - follow-up). We examined differentially methylated CpG sites and regions, as well as pathways analysis in association with the treatment.Results: Main results obtained have shown 110 differentially methylated regions (DMRs) with a significant adjusted p-value area associated with the effects of trauma-focused psychotherapies in the entire cohort. Several annotated genes are related to inflammatory processes and psychiatric disorders, such as LTA, GFI1, ARID5B, TNFSF13, and LST1. Gene enrichment analyses revealed statistically significant processes related to tumour necrosis factor (TNF) receptor and TNF signalling pathway. Stratified analyses by type of trauma-focused psychotherapy showed statistically significant adjusted p-value area in 141 DMRs only for the group of patients receiving EMDR, with annotated genes related to inflammation and psychiatric disorders, including LTA, GFI1, and S100A8. Gene set enrichment analyses in the EMDR group indicated biological processes related to inflammatory response, particularly the TNF signalling pathway.Conclusion: We provide preliminary valuable insights into global DNA methylation changes associated with trauma-focused psychotherapies effects, in particular with EMDR treatment.
Collapse
Affiliation(s)
- Rosana Carvalho Silva
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Christa Hohoff
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
| | - Stefania Mattevi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Valentina Menesello
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Bernhard T. Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, University of Melbourne, Melbourne, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
11
|
Skinner MK. Epigenetic biomarkers for disease susceptibility and preventative medicine. Cell Metab 2024; 36:263-277. [PMID: 38176413 DOI: 10.1016/j.cmet.2023.11.015] [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: 08/15/2023] [Revised: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
The development of molecular biomarkers for disease makes it possible for preventative medicine approaches to be considered. Therefore, therapeutics, treatments, or clinical management can be used to delay or prevent disease development. The problem with genetic mutations as biomarkers is the low frequency with genome-wide association studies (GWASs), generally at best a 1% association of the patients with the disease. In contrast, epigenetic alterations have a high-frequency association of greater than 90%-95% of individuals with pathology in epigenome-wide association studies (EWASs). A wide variety of human diseases have been shown to have epigenetic biomarkers that are disease specific and that detect pathology susceptibility. This review is focused on the epigenetic biomarkers for disease susceptibility, and it distinct from the large literature on epigenetics of disease etiology or progression. The development of efficient epigenetic biomarkers for disease susceptibility will facilitate a paradigm shift from reactionary medicine to preventative medicine.
Collapse
Affiliation(s)
- Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA.
| |
Collapse
|
12
|
Dósa Z, Nieto-Gonzalez JL, Elfving B, Hougaard KS, Holm MM, Wegener G, Jensen K. Reduction in hippocampal GABAergic transmission in a low birth weight rat model of depression. Acta Neuropsychiatr 2023; 35:315-327. [PMID: 36896595 DOI: 10.1017/neu.2023.18] [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] [Indexed: 03/11/2023]
Abstract
Prenatal stress is believed to increase the risk of developing neuropsychiatric disorders, including major depression. Adverse genetic and environmental impacts during early development, such as glucocorticoid hyper-exposure, can lead to changes in the foetal brain, linked to mental illnesses developed in later life. Dysfunction in the GABAergic inhibitory system is associated with depressive disorders. However, the pathophysiology of GABAergic signalling is poorly understood in mood disorders. Here, we investigated GABAergic neurotransmission in the low birth weight (LBW) rat model of depression. Pregnant rats, exposed to dexamethasone, a synthetic glucocorticoid, during the last week of gestation, yielded LBW offspring showing anxiety- and depressive-like behaviour in adulthood. Patch-clamp recordings from dentate gyrus granule cells in brain slices were used to examine phasic and tonic GABAA receptor-mediated currents. The transcriptional levels of selected genes associated with synaptic vesicle proteins and GABAergic neurotransmission were investigated. The frequency of spontaneous inhibitory postsynaptic currents (sIPSC) was similar in control and LBW rats. Using a paired-pulse protocol to stimulate GABAergic fibres impinging onto granule cells, we found indications of decreased probability of GABA release in LBW rats. However, tonic GABAergic currents and miniature IPSCs, reflecting quantal vesicle release, appeared normal. Additionally, we found elevated expression levels of two presynaptic proteins, Snap-25 and Scamp2, components of the vesicle release machinery. The results suggest that altered GABA release may be an essential feature in the depressive-like phenotype of LBW rats.
Collapse
Affiliation(s)
- Zita Dósa
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karin Sørig Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Mai Marie Holm
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Pharmaceutical Research Center of Excellence, North-West University, Potchefstroom, South Africa
| | - Kimmo Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
13
|
Carvalho Silva R, Pisanu C, Maffioletti E, Menesello V, Bortolomasi M, Gennarelli M, Baune BT, Squassina A, Minelli A. Biological markers of sex-based differences in major depressive disorder and in antidepressant response. Eur Neuropsychopharmacol 2023; 76:89-107. [PMID: 37595325 DOI: 10.1016/j.euroneuro.2023.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/20/2023]
Abstract
Major depressive disorder (MDD) presents different clinical features in women and men, with women being more affected and responding differently to antidepressant treatment. Specific molecular mechanisms underlying these differences are not well studied and this narrative review aims at providing an overview of the neurobiological features underlying sex-differences in biological systems involved in MDD pathophysiology and response to antidepressant treatment, focusing on human studies. The majority of the reviewed studies were performed through candidate gene approaches, focusing on biological systems involved in MDD pathophysiology, including the stress response, inflammatory and immune, monoaminergic, neurotrophic, gamma-aminobutyric acid and glutamatergic, and oxytocin systems. The influence of the endocrine system and sex-specific hormone effects are also discussed. Genome, epigenome and transcriptome-wide approaches are less frequently performed and most of these studies do not focus on sex-specific alterations, revealing a paucity of omics studies directed to unravel sex-based differences in MDD. Few studies about sex-related differences in antidepressant treatment response have been conducted, mostly involving the inflammatory system, with less evidence on the monoaminergic system and sparse evidence in omics approaches. Our review covers the importance of accounting for sex-differences in research, optimizing patient stratification for a more precise diagnostic and individualized treatment for women and men.
Collapse
Affiliation(s)
- Rosana Carvalho Silva
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - Elisabetta Maffioletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Valentina Menesello
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Bernhard T Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany; Department of Psychiatry, Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, B3H 2E2, Canada
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
14
|
Walter HJ, Abright AR, Bukstein OG, Diamond J, Keable H, Ripperger-Suhler J, Rockhill C. Clinical Practice Guideline for the Assessment and Treatment of Children and Adolescents With Major and Persistent Depressive Disorders. J Am Acad Child Adolesc Psychiatry 2023; 62:479-502. [PMID: 36273673 DOI: 10.1016/j.jaac.2022.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To enhance the quality of care and clinical outcomes for children and adolescents with major depressive disorder (MDD) and persistent depressive disorder (PDD). The aims are as follows: (1) to summarize empirically based guidance about the psychosocial and psychopharmacologic treatment of MDD and PDD in children and adolescents; and (2) to summarize expert-based guidance about the assessment of these disorders as an integral part of treatment, and the implementation of empirically based treatments for these disorders in clinical practice. METHOD Statements about the treatment of MDD and PDD are based upon empirical evidence derived from a critical systematic review of the scientific literature conducted by the Research Triangle Institute International-University of North Carolina at Chapel Hill (RTI-UNC) Evidence-based Practice Center under contract with the Agency for Healthcare Research and Quality (AHRQ). Evidence from meta-analyses published since the AHRQ/RTI-UNC review is also presented to support or refute the AHRQ findings. Guidance about the assessment and clinical implementation of treatments for MDD and PDD is informed by expert opinion and consensus as presented in previously published clinical practice guidelines, chapters in leading textbooks of child and adolescent psychiatry, the DSM-5-TR, and government-affiliated prescription drug information websites. RESULTS Psychotherapy (specifically, cognitive-behavioral and interpersonal therapies) and selective serotonin reuptake inhibitor (SSRI) medication have some rigorous (randomized controlled trials, meta-analyses) empirical support as treatment options. Because effective treatment outcomes are predicated in part upon accuracy of the diagnosis, depth of the clinical formulation, and breadth of the treatment plan, comprehensive, evidence-based assessment may enhance evidence-based treatment outcomes. CONCLUSION Disproportionate to the magnitude of the problem, there are significant limitations in the quality and quantity of rigorous empirical support for the etiology, assessment, and treatment of depression in children and adolescents. In the context of a protracted severe shortage of child and adolescent-trained behavioral health specialists, the demonstration of convenient, efficient, cost-effective, and user-friendly delivery mechanisms for safe and effective treatment of MDD and PDD is a key research need. Other research priorities include the sequencing and comparative effectiveness of depression treatments, delineation of treatment mediators and moderators, effective approaches to treatment nonresponders and disorder relapse/recurrence, long-term effects and degree of suicide risk with SSRI use, and the discovery of novel pharmacologic or interventional treatments.
Collapse
|
15
|
Mendonça MS, Mangiavacchi PM, Mendes AV, Loureiro SR, Martín-Santos R, Glória LS, Marques W, De Marco SPG, Kanashiro MM, Hallak JEC, Crippa JAS, Rios ÁFL. DNA methylation in regulatory elements of the FKBP5 and NR3C1 gene in mother-child binomials with depression. J Affect Disord 2023; 331:287-299. [PMID: 36933666 DOI: 10.1016/j.jad.2023.03.031] [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: 09/27/2022] [Revised: 02/24/2023] [Accepted: 03/11/2023] [Indexed: 03/20/2023]
Abstract
BACKGROUND The FKBP5 and NR3C1 genes play an important role in stress response, thus impacting mental health. Stress factor exposure in early life, such as maternal depression, may contribute to epigenetic modifications in stress response genes, increasing the susceptibility to different psychopathologies. The present study aimed to evaluate the DNA methylation profile in maternal-infant depression in regulatory regions of the FKBP5 gene and the alternative promoter of the NR3C1 gene. METHODS We evaluated 60 mother-infant pairs. The levels of DNA methylation were analyzed by the MSRED-qPCR technique. RESULTS We observed an increased DNA methylation profile in the NR3C1 gene promoter in children with depression and children exposed to maternal depression (p < 0.05). In addition, we observed a correlation of DNA methylation between mothers and offspring exposed to maternal depression. This correlation shows a possible intergenerational effect of maternal MDD exposure on the offspring. For FKBP5, we found a decrease in DNA methylation at intron 7 in children exposed to maternal MDD during pregnancy and a correlation of DNA methylation between mothers and children exposed to maternal MDD (p < 0.05). LIMITATIONS Although the individuals of this study are a rare group, the sample size of the study was small, and we evaluated the DNA methylation of only one CpG site for each region. CONCLUSION These results indicate changes in DNA methylation levels in regulatory regions of FKBP5 and NR3C1 in the mother-child MDD context and represent a potential target of studies to understand the depression etiology and how it occurs between generations.
Collapse
Affiliation(s)
- Mariana S Mendonça
- Laboratory of Biotechnology, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil
| | - Paula M Mangiavacchi
- Laboratory of Reproduction and Animal Breeding, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil
| | - Ana V Mendes
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Sonia R Loureiro
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Rocio Martín-Santos
- Instituto Nacional de Ciência e Tecnologia Translacional em Medicina Translational Medicine (INCT-TM), National Council for Scientific and Technological Development, São Paulo, Brazil; Department of Psychiatry and Psychology, Hospital Clínic, Institut d' Investigacions Biomedicas August Pi I Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Instituto de Neurociencias, University of Barcelona, Barcelona 08036, Spain
| | - Leonardo S Glória
- Laboratory of Animal Science, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil
| | - Wilson Marques
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Silmara P G De Marco
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Milton M Kanashiro
- Laboratory of Recognition Biology, North Fluminense State University (UENF), Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil
| | - Jaime E C Hallak
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina Translational Medicine (INCT-TM), National Council for Scientific and Technological Development, São Paulo, Brazil
| | - José A S Crippa
- Department of Neurosciences and Behavioral Sciences, USP, Ribeirão Preto, São Paulo 14051-140, Brazil; Instituto Nacional de Ciência e Tecnologia Translacional em Medicina Translational Medicine (INCT-TM), National Council for Scientific and Technological Development, São Paulo, Brazil
| | - Álvaro F L Rios
- Laboratory of Biotechnology, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Campos dos Goytacazes, Rio de Janeiro 28013-602, Brazil.
| |
Collapse
|
16
|
Roy B, Dwivedi Y. An insight into the sprawling microverse of microRNAs in depression pathophysiology and treatment response. Neurosci Biobehav Rev 2023; 146:105040. [PMID: 36639069 PMCID: PMC9974865 DOI: 10.1016/j.neubiorev.2023.105040] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/06/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Stress-related neuropathologies are pivotal in developing major depressive disorder (MDD) and are often governed by gene-regulatory changes. Being a stress-responsive gene-regulatory factor, microRNAs (miRNAs) have tremendous biomolecular potential to define an altered gene-regulatory landscape in the MDD brain. MiRNAs' regulatory roles in the MDD brain are closely aligned with changes in plasticity, neurogenesis, and stress-axis functions. MiRNAs act at the epigenetic interface between stress-induced environmental stimuli and cellular pathologies by triggering large-scale gene expression changes in a highly coordinated fashion. The parallel changes in peripheral circulation may provide an excellent opportunity for miRNA to devise more effective treatment strategies and help explore their potential as biomarkers in treatment response. This review discusses the role of miRNAs as epigenetic modifiers in the etiopathogenesis of MDD. Concurrently, key research is highlighted to show the progress in using miRNAs as predictive biomarkers for treatment response.
Collapse
Affiliation(s)
- Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
17
|
Gathier AW, Verhoeven JE, van Oppen PC, Penninx BWJH, Merkx MJM, Dingemanse P, Stehouwer KMKS, van den Bulck CMM, Vinkers CH. Design and rationale of the REStoring mood after early life trauma with psychotherapy (RESET-psychotherapy) study: a multicenter randomized controlled trial on the efficacy of adjunctive trauma-focused therapy (TFT) versus treatment as usual (TAU) for adult patients with major depressive disorder (MDD) and childhood trauma. BMC Psychiatry 2023; 23:41. [PMID: 36650502 PMCID: PMC9843991 DOI: 10.1186/s12888-023-04518-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a common, recurrent mental disorder and a leading cause of disability worldwide. A large part of adult MDD patients report a history of childhood trauma (CT). Patients with MDD and CT are assumed to represent a clinically and neurobiologically distinct MDD subtype with an earlier onset, unfavorable disease course, stress systems' dysregulations and brain alterations. Currently, there is no evidence-based treatment strategy for MDD that specifically targets CT. Given the central role of trauma in MDD patients with CT, trauma-focused therapy (TFT), adjunctive to treatment as usual (TAU), may be efficacious to alleviate depressive symptoms in this patient population. METHODS The RESET-psychotherapy study is a 12-week, single-blind, randomized controlled trial testing the efficacy of TFT in 158 adults with moderate to severe MDD, as a 'stand-alone' depression diagnosis or superimposed on a persistent depressive disorder (PDD), and CT. TFT (6-10 sessions of Eye Movement Desensitization and Reprocessing and/or imagery rescripting) + TAU is compared to TAU only. Assessments, including a wide range of psychological/psychiatric and biological characteristics, take place before randomization (T0), during treatment (T1), at post-treatment (T2) and at 6-month follow-up (T3). Pre-post treatment stress-related biomarkers in hair (cortisol) and blood (epigenetics and inflammation) will be assessed to better understand working mechanisms of TFT. A subgroup of 60 participants will undergo structural and functional Magnetic Resonance Imaging (MRI) assessments to determine pre-post treatment brain activity. The primary outcome is self-reported depression symptom severity at post-treatment, measured with the 30-item Inventory of Depressive Symptomatology - Self Report (IDS-SR). DISCUSSION If adjunctive TFT efficaciously alleviates depressive symptoms in MDD patients with CT, this novel treatment strategy could pave the way for a more personalized and targeted MDD treatment. TRIAL REGISTRATION ClinicalTrials.gov, registered at 08-12-2021, number of identification: NCT05149352.
Collapse
Affiliation(s)
- Anouk W. Gathier
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Boelelaan 1117, Amsterdam, The Netherlands ,Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
| | - Josine E. Verhoeven
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Boelelaan 1117, Amsterdam, The Netherlands ,Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands ,grid.420193.d0000 0004 0546 0540GGZ inGeest Mental Health Care, Amsterdam, The Netherlands
| | - Patricia C. van Oppen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Boelelaan 1117, Amsterdam, The Netherlands ,Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands ,grid.420193.d0000 0004 0546 0540GGZ inGeest Mental Health Care, Amsterdam, The Netherlands
| | - Brenda W. J. H. Penninx
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Boelelaan 1117, Amsterdam, The Netherlands ,Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | | | - Pieter Dingemanse
- grid.413664.2Altrecht GGZ, Nieuwe Houtenseweg 12, Utrecht, The Netherlands
| | | | | | - Christiaan H. Vinkers
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Department of Psychiatry, Boelelaan 1117, Amsterdam, The Netherlands ,Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands ,grid.420193.d0000 0004 0546 0540GGZ inGeest Mental Health Care, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Kanes SJ, Dennie L, Perera P. Targeting the Arginine Vasopressin V 1b Receptor System and Stress Response in Depression and Other Neuropsychiatric Disorders. Neuropsychiatr Dis Treat 2023; 19:811-828. [PMID: 37077711 PMCID: PMC10106826 DOI: 10.2147/ndt.s402831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
A healthy stress response is critical for good mental and overall health and promotes neuronal growth and adaptation, but the intricately balanced biological mechanisms that facilitate a stress response can also result in predisposition to disease when that equilibrium is disrupted. The hypothalamic-pituitary-adrenal (HPA) axis neuroendocrine system plays a critical role in the body's response and adaptation to stress, and vasopressinergic regulation of the HPA axis is critical to maintaining system responsiveness during chronic stress. However, exposure to repeated or excessive physical or emotional stress or trauma can shift the body's stress response equilibrium to a "new normal" underpinned by enduring changes in HPA axis function. Exposure to early life stress due to adverse childhood experiences can also lead to lasting neurobiological changes, including in HPA axis function. HPA axis impairment in patients with depression is considered among the most reliable findings in biological psychiatry, and chronic stress has been shown to play a major role in the pathogenesis and onset of depression and other neuropsychiatric disorders. Modulating HPA axis activity, for example via targeted antagonism of the vasopressin V1b receptor, is a promising approach for patients with depression and other neuropsychiatric disorders associated with HPA axis impairment. Despite favorable preclinical indications in animal models, demonstration of clinical efficacy for the treatment of depressive disorders by targeting HPA axis dysfunction has been challenging, possibly due to the heterogeneity and syndromal nature of depressive disorders. Measures of HPA axis function, such as elevated cortisol levels, may be useful biomarkers for identifying patients who may benefit from treatments that modulate HPA axis activity. Utilizing clinical biomarkers to identify subsets of patients with impaired HPA axis function who may benefit is a promising next step in fine-tuning HPA axis activity via targeted antagonism of the V1b receptor.
Collapse
Affiliation(s)
- Stephen J Kanes
- EmbarkNeuro, Oakland, CA, USA
- Correspondence: Stephen J Kanes, EmbarkNeuro, Inc, 1111 Broadway, Suite 1300, Oakland, CA, 94607, USA, Tel +1 610 757 7821, Email
| | | | | |
Collapse
|
19
|
Mahony C, O'Ryan C. A molecular framework for autistic experiences: Mitochondrial allostatic load as a mediator between autism and psychopathology. Front Psychiatry 2022; 13:985713. [PMID: 36506457 PMCID: PMC9732262 DOI: 10.3389/fpsyt.2022.985713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Molecular autism research is evolving toward a biopsychosocial framework that is more informed by autistic experiences. In this context, research aims are moving away from correcting external autistic behaviors and toward alleviating internal distress. Autism Spectrum Conditions (ASCs) are associated with high rates of depression, suicidality and other comorbid psychopathologies, but this relationship is poorly understood. Here, we integrate emerging characterizations of internal autistic experiences within a molecular framework to yield insight into the prevalence of psychopathology in ASC. We demonstrate that descriptions of social camouflaging and autistic burnout resonate closely with the accepted definitions for early life stress (ELS) and chronic adolescent stress (CAS). We propose that social camouflaging could be considered a distinct form of CAS that contributes to allostatic overload, culminating in a pathophysiological state that is experienced as autistic burnout. Autistic burnout is thought to contribute to psychopathology via psychological and physiological mechanisms, but these remain largely unexplored by molecular researchers. Building on converging fields in molecular neuroscience, we discuss the substantial evidence implicating mitochondrial dysfunction in ASC to propose a novel role for mitochondrial allostatic load in the relationship between autism and psychopathology. An interplay between mitochondrial, neuroimmune and neuroendocrine signaling is increasingly implicated in stress-related psychopathologies, and these molecular players are also associated with neurodevelopmental, neurophysiological and neurochemical aspects of ASC. Together, this suggests an increased exposure and underlying molecular susceptibility to ELS that increases the risk of psychopathology in ASC. This article describes an integrative framework shaped by autistic experiences that highlights novel avenues for molecular research into mechanisms that directly affect the quality of life and wellbeing of autistic individuals. Moreover, this framework emphasizes the need for increased access to diagnoses, accommodations, and resources to improve mental health outcomes in autism.
Collapse
Affiliation(s)
| | - Colleen O'Ryan
- Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
20
|
Herzberg MP, Tillman R, Kandala S, Barch DM, Luby J. Preschool Depression and Hippocampal Volume: The Moderating Role of Family Income. J Am Acad Child Adolesc Psychiatry 2022; 61:1362-1371. [PMID: 35523377 PMCID: PMC10845235 DOI: 10.1016/j.jaac.2022.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 10/31/2022]
Abstract
OBJECTIVE Depression and low socioeconomic status have both been associated with hippocampal volume alterations. Whether these factors interact to predict neurobehavioral outcomes has not been adequately studied. The authors investigated family income as a moderator of the relationship between depression and hippocampal volume in a longitudinal sample. METHOD Longitudinal behavioral data, beginning at preschool age, and behavioral and neuroimaging data from school age to adolescence were used to assess the impact of preschool only and total preschool to adolescent depression symptoms on hippocampal volumes using family income as a moderator (N = 176). RESULTS Depression severity during the preschool period interacted with family income to predict hippocampal volumes at the intercept (ie, age 13 years; B = -0.078, p = .003). Interaction decomposition revealed that only individuals with relatively high family income exhibited smaller hippocampal volume with increasing depression severity (B = -0.146, p = .005). Family income was associated with hippocampus volumes only in individuals with low to moderate preschool depression severity (B = 0.289, p = .007 and B = 0.169, p = .030, respectively). CONCLUSION Preschool depression severity interacts with family income to predict hippocampal volume across development, such that the effects of early depression are evident only in those with higher income. These findings suggest that hippocampal volume may not be an effective marker of risk for depression at different levels of socioeconomic status, and emphasizes the importance of the environmental context when assessing risk markers for depression. Future research should explore how socioeconomic stress may eclipse the effects of depression on hippocampal development, setting alternative neurodevelopmental risk trajectories.
Collapse
Affiliation(s)
- Max P Herzberg
- Washington University in St. Louis, St. Louis, Missouri.
| | | | | | | | - Joan Luby
- Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
21
|
Uliana DL, Zhu X, Gomes FV, Grace AA. Using animal models for the studies of schizophrenia and depression: The value of translational models for treatment and prevention. Front Behav Neurosci 2022; 16:935320. [PMID: 36090659 PMCID: PMC9449416 DOI: 10.3389/fnbeh.2022.935320] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Animal models of psychiatric disorders have been highly effective in advancing the field, identifying circuits related to pathophysiology, and identifying novel therapeutic targets. In this review, we show how animal models, particularly those based on development, have provided essential information regarding circuits involved in disorders, disease progression, and novel targets for intervention and potentially prevention. Nonetheless, in recent years there has been a pushback, largely driven by the US National Institute of Mental Health (NIMH), to shift away from animal models and instead focus on circuits in normal subjects. This has been driven primarily from a lack of discovery of new effective therapeutic targets, and the failure of targets based on preclinical research to show efficacy. We discuss why animal models of complex disorders, when strongly cross-validated by clinical research, are essential to understand disease etiology as well as pathophysiology, and direct new drug discovery. Issues related to shortcomings in clinical trial design that confound translation from animal models as well as the failure to take patient pharmacological history into account are proposed to be a source of the failure of what are likely effective compounds from showing promise in clinical trials.
Collapse
Affiliation(s)
- Daniela L. Uliana
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiyu Zhu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A. Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
22
|
Chen J, Luo Q, Li Y, Wu Z, Lin X, Yao J, Yu H, Nie H, Du Y, Peng H, Wu H. Intrinsic brain abnormalities in female major depressive disorder patients with childhood trauma: A resting-state functional magnetic resonance imaging study. Front Neurosci 2022; 16:930997. [PMID: 36017185 PMCID: PMC9395929 DOI: 10.3389/fnins.2022.930997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Objective Childhood trauma is a strong predictor of major depressive disorder (MDD). Women are more likely to develop MDD than men. However, the neural basis of female MDD patients with childhood trauma remains unclear. We aimed to identify the specific brain regions that are associated with female MDD patients with childhood trauma. Methods We recruited 16 female MDD patients with childhood trauma, 16 female MDD patients without childhood trauma, and 20 age- and education level-matched healthy controls. All participants underwent resting-state functional magnetic resonance imaging (MRI). Regional brain activity was evaluated as the amplitude of low-frequency fluctuation (ALFF). Furthermore, functional connectivity (FC) analyses were performed on areas with altered ALFF to explore alterations in FC patterns. Results There was increased ALFF in the left middle frontal gyrus (MFG) and the right postcentral gyrus (PoCG) in MDD with childhood trauma compared with MDD without childhood trauma. The areas with significant ALFF discrepancies were selected as seeds for the FC analyses. There was increased FC between the left MFG and the bilateral putamen gyrus. Moreover, ALFF values were correlated with childhood trauma severity. Conclusion Our findings revealed abnormal intrinsic brain activity and FC patterns in female MDD patients with childhood trauma, which provides new possibilities for exploring the pathophysiology of this disorder in women.
Collapse
Affiliation(s)
- Juran Chen
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianyi Luo
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuhong Li
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiyao Wu
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyi Lin
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiazheng Yao
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiwen Yu
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiqin Nie
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingying Du
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongjun Peng
- Department of Clinical Psychology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Hongjun Peng,
| | - Huawang Wu
- Department of Radiology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
- Huawang Wu,
| |
Collapse
|
23
|
Packard K, Opendak M. Rodent models of early adversity: Impacts on developing social behavior circuitry and clinical implications. Front Behav Neurosci 2022; 16:918862. [PMID: 35990728 PMCID: PMC9385963 DOI: 10.3389/fnbeh.2022.918862] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Flexible and context-appropriate social functioning is key for survival across species. This flexibility also renders social behavior highly plastic, particularly during early development when attachment to caregiver can provide a template for future social processing. As a result, early caregiving adversity can have unique and lasting impacts on social behavior and even confer vulnerability to psychiatric disorders. However, the neural circuit mechanisms translating experience to outcome remain poorly understood. Here, we consider social behavior scaffolding through the lens of reward and threat processing. We begin by surveying several complementary rodent models of early adversity, which together have highlighted impacts on neural circuits processing social cues. We next explore these circuits underlying perturbed social functioning with focus on dopamine (DA) and its role in regions implicated in social and threat processing such as the prefrontal cortex (PFC), basolateral amygdala (BLA) and the lateral habenula (LHb). Finally, we turn to human populations once more to examine how altered DA signaling and LHb dysfunction may play a role in social anhedonia, a common feature in diagnoses such as schizophrenia and major depressive disorder (MDD). We argue that this translational focus is critical for identifying specific features of adversity that confer heightened vulnerability for clinical outcomes involving social cue processing.
Collapse
Affiliation(s)
- Katherine Packard
- Department of Neuroscience, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Maya Opendak
- Department of Neuroscience, Kennedy Krieger Institute, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
24
|
Lippard ETC, Nemeroff CB. Going beyond risk factor: Childhood maltreatment and associated modifiable targets to improve life-long outcomes in mood disorders. Pharmacol Biochem Behav 2022; 215:173361. [PMID: 35219755 DOI: 10.1016/j.pbb.2022.173361] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 01/26/2023]
Abstract
Childhood maltreatment increases risk for mood disorders and is associated with earlier onset-and more pernicious disease course following onset-of mood disorders. While the majority of studies to date have been cross-sectional, longitudinal studies are emerging and support the devastating role(s) childhood maltreatment has on development of, and illness course in, mood disorders. This manuscript extends prior reviews to emphasize more recent work, highlighting longitudinal data, and discusses treatment studies that provide clues to mechanisms that mediate disease risk, course, relapse, and treatment response. Evidence suggesting systemic inflammation, alterations in hypothalamic-pituitary-adrenal (HPA) axis function and corticotropin-releasing factor (CRF) neural systems, genetic and other familial factors as mechanisms that mediate risk and onset of, and illness course in, mood disorders following childhood maltreatment is discussed. Risky behaviors following maltreatment, e.g., substance use and unhealthy lifestyles, may further exacerbate alterations in the HPA axis, CRF neural systems, and systematic inflammation to contribute to a more pernicious disease course. More research on sex differences and the impact of maltreatment in vulnerable populations is needed. Future research needs to be aimed at leveraging knowledge on modifiable targets, going beyond childhood maltreatment as a risk factor, to inform prevention and treatment strategies and foster trauma-informed care.
Collapse
Affiliation(s)
- Elizabeth T C Lippard
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX, USA; Institute of Early Life Adversity Research, Dell Medical School, University of Texas, Austin, TX, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX, USA; Department of Psychology, University of Texas, Austin, TX, USA; Mulva Clinic for Neuroscience, Dell Medical School, University of Texas, Austin, TX, USA.
| | - Charles B Nemeroff
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, University of Texas, Austin, TX, USA; Institute of Early Life Adversity Research, Dell Medical School, University of Texas, Austin, TX, USA; Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, TX, USA; Mulva Clinic for Neuroscience, Dell Medical School, University of Texas, Austin, TX, USA
| |
Collapse
|
25
|
Li QS, Galbraith D, Morrison RL, Trivedi MH, Drevets WC. Circulating microRNA associated with future relapse status in major depressive disorder. Front Psychiatry 2022; 13:937360. [PMID: 36061300 PMCID: PMC9428445 DOI: 10.3389/fpsyt.2022.937360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Major depressive disorder (MDD) is an episodic condition with relapsing and remitting disease course. Elucidating biomarkers that can predict future relapse in individuals responding to an antidepressant treatment holds the potential to identify those patients who are prone to illness recurrence. The current study explored relationships between relapse risk in recurrent MDD and circulating microRNAs (miRNAs) that participate in RNA silencing and post-transcriptional regulation of gene expression. Serum samples were acquired from individuals with a history of recurrent MDD who were followed longitudinally in the observational study, OBSERVEMDD0001 (ClinicalTrials.gov Identifier: NCT02489305). Circulating miRNA data were obtained in 63 participants who relapsed ("relapsers") and 154 participants who did not relapse ("non-relapsers") during follow-up. The miRNA was quantified using the ID3EAL™ miRNA Discovery Platform from MiRXES measuring 575 circulating miRNAs using a patented qPCR technology and normalized with a standard curve from spike-in controls in each plate. The association between miRNAs and subsequent relapse was tested using a linear model, adjusting for age, gender, and plate. Four miRNAs were nominally associated with relapse status during the observational follow-up phase with a false discover rate adjusted p-value < 0.1. Enrichment analysis of experimentally validated targets revealed 112 significantly enriched pathways, including neurogenesis, response to cytokine, neurotrophin signaling, vascular endothelial growth factor signaling, relaxin signaling, and cellular senescence pathways. These data suggest these miRNAs putatively associated with relapse status may have the potential to regulate genes involved in multiple signaling pathways that have previously been associated with MDD. If shown to be significant in a larger, independent sample, these data may hold potential for developing a miRNA signature to identify patients likely to relapse, allowing for earlier intervention.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, Titusville, NJ, United States.,JRD Data Science, Janssen Research and Development, LLC, Titusville, NJ, United States
| | | | - Randall L Morrison
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, Titusville, NJ, United States
| | - Madhukar H Trivedi
- Department of Psychiatry, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Wayne C Drevets
- Neuroscience Therapeutic Area, Janssen Research and Development, LLC, San Diego, CA, United States
| |
Collapse
|
26
|
Deuter CE, Smit J, Kaczmarczyk M, Wingenfeld K, Otte C, Kuehl LK. Approach-avoidance tendencies in depression and childhood trauma: No effect of noradrenergic stimulation. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2021; 8:100077. [PMID: 35757673 PMCID: PMC9216672 DOI: 10.1016/j.cpnec.2021.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/18/2021] [Accepted: 07/30/2021] [Indexed: 11/17/2022] Open
Abstract
Adverse childhood experiences (ACE) are a major risk factor for major depressive disorder (MDD) in later life. Both conditions are characterized by dysregulations in the noradrenergic system related which again could represent a mediating mechanism for deficits in affective processing and behavioral functioning. In this double-blind, placebo-controlled study we tested the hypothesis that ACE and MDD are characterized by aberrant approach-avoidance (AA) tendencies and that these are mitigated after noradrenergic stimulation with yohimbine. In a mixed-measures, fully crossed design, participants (N = 131, 73 women) with/without MDD and with/without ACE received a single-dose of yohimbine or placebo on different days, followed by an AA task. We found modulation of AA tendencies by the emotional valence of target images, yet there were no effects of group or treatment. From these results, we conclude that AA tendencies are not critically affected by MDD or ACE and that the noradrenergic system is not substantially involved in this behavior.
Collapse
|
27
|
Onaka T, Takayanagi Y. The oxytocin system and early-life experience-dependent plastic changes. J Neuroendocrinol 2021; 33:e13049. [PMID: 34713517 PMCID: PMC9286573 DOI: 10.1111/jne.13049] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023]
Abstract
Early-life experience influences social and emotional behaviour in adulthood. Affiliative tactile stimuli in early life facilitate the development of social and emotional behaviour, whereas early-life adverse stimuli have been shown to increase the risk of various diseases in later life. On the other hand, oxytocin has been shown to have organizational actions during early-life stages. However, the detailed mechanisms of the effects of early-life experience and oxytocin remain unclear. Here, we review the effects of affiliative tactile stimuli during the neonatal period and neonatal oxytocin treatment on the activity of the oxytocin-oxytocin receptor system and social or emotional behaviour in adulthood. Both affiliative tactile stimuli and early-life adverse stimuli in the neonatal period acutely activate the oxytocin-oxytocin receptor system in the brain but modulate social behaviour and anxiety-related behaviour apparently in an opposite direction in adulthood. Accumulating evidence suggests that affiliative tactile stimuli and exogenous application of oxytocin in early-life stages induce higher activity of the oxytocin-oxytocin receptor system in adulthood, although the effects are dependent on experimental procedures, sex, dosages and brain regions examined. On the other hand, early-life stressful stimuli appear to induce reduced activity of the oxytocin-oxytocin receptor system, possibly leading to adverse actions in adulthood. It is possible that activation of a specific oxytocin system can induce beneficial actions against early-life maltreatments and thus could be used for the treatment of developmental psychiatric disorders.
Collapse
Affiliation(s)
- Tatsushi Onaka
- Division of Brain and NeurophysiologyDepartment of PhysiologyJichi Medical UniversityTochigiJapan
| | - Yuki Takayanagi
- Division of Brain and NeurophysiologyDepartment of PhysiologyJichi Medical UniversityTochigiJapan
| |
Collapse
|
28
|
Targeting metabotropic glutamate receptors for the treatment of depression and other stress-related disorders. Neuropharmacology 2021; 196:108687. [PMID: 34175327 DOI: 10.1016/j.neuropharm.2021.108687] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/15/2022]
Abstract
The discovery of robust antidepressant effects of ketamine in refractory patients has led to increasing focus on agents targeting glutamatergic signaling as potential novel antidepressant strategy. Among the agents targeting the glutamatergic system, compounds acting at metabotropic glutamate (mGlu) receptors are among the most promising agents under studies for depressive disorders. Further, the receptor diversity, distinct distribution in the CNS, and ability to modulate the glutamatergic neurotransmission in the brain areas implicated in mood disorders make them an exciting target for stress-related disorders. In preclinical models, antidepressant and anxiolytic effects of mGlu5 negative allosteric modulators (NAMs) have been reported. Interestingly, mGlu2/3 receptor antagonists show fast and sustained antidepressant-like effects similar to that of ketamine in rodents. Excitingly, they can also induce antidepressant effects in the animal models of treatment-resistant depression and are devoid of the side-effects associated with ketamine. Unfortunately, clinical trials of both mGlu5 and mGlu2/3 receptor NAMs have been inconclusive, and additional trials using other compounds with suitable preclinical and clinical properties are needed. Although group III mGlu receptors have gained less attention, mGlu7 receptor ligands have been shown to induce antidepressant-like effects in rodents. Collectively, compounds targeting mGlu receptors provide an alternative approach to fill the outstanding clinical need for safer and more efficacious antidepressants. This article is part of the special Issue on "Glutamate Receptors - mGluRs".
Collapse
|
29
|
Mourtzi N, Sertedaki A, Charmandari E. Glucocorticoid Signaling and Epigenetic Alterations in Stress-Related Disorders. Int J Mol Sci 2021; 22:5964. [PMID: 34073101 PMCID: PMC8198182 DOI: 10.3390/ijms22115964] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Stress is defined as a state of threatened or perceived as threatened homeostasis. The well-tuned coordination of the stress response system is necessary for an organism to respond to external or internal stressors and re-establish homeostasis. Glucocorticoid hormones are the main effectors of stress response and aberrant glucocorticoid signaling has been associated with an increased risk for psychiatric and mood disorders, including schizophrenia, post-traumatic stress disorder and depression. Emerging evidence suggests that life-stress experiences can alter the epigenetic landscape and impact the function of genes involved in the regulation of stress response. More importantly, epigenetic changes induced by stressors persist over time, leading to increased susceptibility for a number of stress-related disorders. In this review, we discuss the role of glucocorticoids in the regulation of stress response, the mechanism through which stressful experiences can become biologically embedded through epigenetic alterations, and we underline potential associations between epigenetic changes and the development of stress-related disorders.
Collapse
Affiliation(s)
- Niki Mourtzi
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
| | - Amalia Sertedaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
30
|
Merz MP, Turner JD. Is early life adversity a trigger towards inflammageing? Exp Gerontol 2021; 150:111377. [PMID: 33905877 DOI: 10.1016/j.exger.2021.111377] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
There are many 'faces' of early life adversity (ELA), such as childhood trauma, institutionalisation, abuse or exposure to environmental toxins. These have been implicated in the onset and severity of a wide range of chronic non-communicable diseases later in life. The later-life disease risk has a well-established immunological component. This raises the question as to whether accelerated immune-ageing mechanistically links early-life adversity to the lifelong health trajectory resulting in either 'poor' or 'healthy' ageing. Here we examine observational and mechanistic studies of ELA and inflammageing, highlighting common and distinct features in these two life stages. Many biological processes appear in common including reduction in telomere length, increased immunosenescence, metabolic distortions and chronic (viral) infections. We propose that ELA shapes the developing immune, endocrine and nervous system in a non-reversible way, creating a distinct phenotype with accelerated immunosenescence and systemic inflammation. We conclude that ELA might act as an accelerator for inflammageing and age-related diseases. Furthermore, we now have the tools and cohorts to be able to dissect the interaction between ELA and later life phenotype. This should, in the near future, allow us to identify the ecological and mechanistic processes that are involved in 'healthy' or accelerated immune-ageing.
Collapse
Affiliation(s)
- Myriam P Merz
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 2 avenue de Université, L-4365 Esch-sur-Alzette, Luxembourg
| | - Jonathan D Turner
- Immune Endocrine and Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29 rue Henri Koch, L-4354 Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
31
|
Deuter CE, Otte C, Wingenfeld K, Kuehl LK. Yohimbine-Induced Reactivity of Heart Rate Variability in Unmedicated Depressed Patients With and Without Adverse Childhood Experience. Front Psychiatry 2021; 12:734904. [PMID: 34975560 PMCID: PMC8717379 DOI: 10.3389/fpsyt.2021.734904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022] Open
Abstract
Stressful life events play a role in the pathogenesis of major depressive disorder (MDD) and many patients with MDD were exposed to developmental stress due to adverse childhood experiences (ACE). Furthermore, dysregulation of the autonomic nervous system and higher incidence of cardiovascular disease are found in MDD. In MDD, and independently in individuals with ACE, abnormalities in heart rate variability (HRV) have been reported. While these are often confounded, we systematically investigated them with a study which included MDD patients with/without ACE as well as healthy individuals with/without ACE. With this study, we investigated the influence of noradrenergic stimulation on HRV reactivity in unmedicated participants in a randomized, double-blind, repeated measures design. Our sample consisted of men and women with MDD and ACE (n = 25), MDD without ACE (n = 24), healthy participants with ACE (n = 27), and without ACE (n = 48). Participants received a 10 mg single dose of the alpha-2 antagonist yohimbine that increases noradrenergic activity or placebo on 2 separate days, with ECG recordings before and after drug administration at defined intervals. We found lower basal HRV in MDD and ACE: patients with MDD had reduced RMSSD whereas participants with ACE had lower LF-HRV. Contrary to our hypothesis, there was no effect of yohimbine. With this study, we were able to replicate previous findings on HRV differences in MDD and ACE. From the null effect of yohimbine, we conclude that the yohimbine-induced sympathetic activation is not a significant driver of HRV in MDD and ACE.
Collapse
Affiliation(s)
- Christian Eric Deuter
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Christian Otte
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Katja Wingenfeld
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Linn Kristina Kuehl
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Department of Psychiatry and Psychotherapy, Berlin, Germany.,Department of Psychology, Clinical Psychology and Psychotherapy, MSB Medical School Berlin, Berlin, Germany
| |
Collapse
|