1
|
Nöthling J, Womersley JS, Mhlongo S, Lombard C, Abrahams N, Seedat S, Hemmings SMJ. The relationship between childhood trauma, rs1360780 genotypes, FKBP5 intron 7 methylation and posttraumatic stress disorder in women who have experienced rape. Eur J Psychotraumatol 2025; 16:2485707. [PMID: 40242984 PMCID: PMC12006943 DOI: 10.1080/20008066.2025.2485707] [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: 09/06/2023] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Background: Posttraumatic stress disorder (PTSD) is a common sequela of rape. Dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis, a core regulator of the stress response, has been implicated in the aetiology and chronicity of PTSD. FK506 binding protein (FKBP5) is a co-chaperone and functional regulator of the glucocorticoid receptor and the HPA-axis.Objective: This study investigated main and interaction effects of childhood trauma and the FKBP5 rs1360780 genotype on longitudinal FKBP5 intron 7 methylation, and whether change in FKBP5 methylation over time was associated with PTSD symptom severity over time.Method: Women who experienced rape (n = 96) were recruited from post-rape care services in KwaZulu Natal, South Africa. Total PTSD symptom scores, derived from the Davidson Trauma Scale, were assessed at baseline, 3-months and 6-months post-rape. Methylation levels at five FKBP5 intron 7 CpG sites were determined using EpiTYPER Sequenom MassArray technology. Genotyping of rs1360980 was completed using the Agena MassArray genotyping system. Mixed linear regression models were used to analyse the data.Results: The interaction between rs1360780 genotype and childhood trauma was a significant predictor of FKBP5 methylation over time. There was a significant positive correlation between childhood trauma and methylation levels in participants with the CT and TT genotypes, while there was a significant negative correlation between childhood trauma and methylation in CC genotype carriers. FKBP5 methylation was not a predictor of PTSD scores over time.Conclusion: This is the first study to investigate longitudinal change in FKBP5 methylation in a demographically homogenous same-trauma sample. The findings implicate childhood trauma and FKBP5 rs1360980 genotype in the trajectory of FKBP5 methylation levels in the aftermath of rape. Further research is needed to investigate the longitudinal role of FKBP5 intron 7 methylation in relation to PTSD symptom trajectories post-rape.
Collapse
Affiliation(s)
- Jani Nöthling
- South African Medical Research Council, Gender and Health Research Unit,Cape Town, South Africa
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, South African Medical Research Council, Stellenbosch University, Cape Town, South Africa
| | - Jacqueline Samantha Womersley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, South African Medical Research Council, Stellenbosch University, Cape Town, South Africa
| | - Shibe Mhlongo
- South African Medical Research Council, Gender and Health Research Unit,Cape Town, South Africa
| | - Carl Lombard
- Biostatistics Unit, South African Medical Research Council, Cape Town, South Africa
- Division of Epidemiology and Biostatistics, Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | - Naeemah Abrahams
- South African Medical Research Council, Gender and Health Research Unit,Cape Town, South Africa
- School of Public Health and Family Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, South African Medical Research Council, Stellenbosch University, Cape Town, South Africa
| | - Sian Megan Joanne Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, South African Medical Research Council, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
2
|
Norrholm SD. An Update on the Psychiatric Genomics of Posttraumatic Stress Disorder (PTSD). Psychiatr Clin North Am 2025; 48:403-415. [PMID: 40348425 DOI: 10.1016/j.psc.2025.01.013] [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: 05/14/2025]
Abstract
Posttraumatic Stress Disorder (PTSD) is a prevalent psychiatric condition characterized by intrusive thoughts, hyperarousal, avoidance, and negative cognitive alterations following traumatic events. While a significant portion of individuals experience trauma, only 5% to 30% develop PTSD, with certain groups at higher risk. Research indicates that PTSD's pathophysiology involves altered fear processing, neuroendocrine dysfunction, and immune system changes. Genetic studies, particularly twin studies, suggest a heritability estimate of 30% to 40% for PTSD. Initially focused on gene-environment interactions, recent advancements in genome-wide association studies have identified significant genetic markers enhancing understanding of PTSD's genetic underpinnings.
Collapse
Affiliation(s)
- Seth Davin Norrholm
- Department of Psychiatry and Behavioral Neurosciences, Neuroscience Center for Anxiety, Stress and Trauma, Wayne State University School of Medicine, 6135 Woodward Avenue, Detroit, MI 48202, USA.
| |
Collapse
|
3
|
Andero R. Stress-induced changes in the molecular processes underlying fear memories: implications for PTSD and relevant animal models. Mol Psychiatry 2025; 30:2219-2227. [PMID: 39890919 PMCID: PMC12014489 DOI: 10.1038/s41380-025-02910-8] [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: 06/20/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Most of the fear literature on humans and animals tests healthy individuals. However, fear memories can differ between healthy individuals and those previously exposed to traumatic stress, such as a car accident, sexual abuse, military combat and personal assault. Traumatic stress can lead to post-traumatic stress disorder (PTSD) which presents alterations in fear memories, such as an impairment of fear extinction and extinction recall. PTSD-like animal models are exposed to a single highly stressful experience in the laboratory, such as stress immobilization or single-prolonged stress. Some days later, animals exposed to a PTSD-like model can be tested in fear procedures that help uncover molecular mechanisms of fear memories. In this review, there are discussed the molecular mechanisms in stress-induced fear memories of patients with PTSD and PTSD-like animal models. The focus is on the effects of estradiol and cortisol/corticosterone hormones and of different genes, such as FKBP prolyl isomerase 5 gene (FKBP5) - FK506 binding protein 51 (FKBP51), pituitary adenylate cyclase-activating peptide (PACAP) - pituitary adenylate cyclase-activating polypeptide type I receptor (PAC1R), endocannabinoid (eCB) system and the tropomyosin receptor kinase B (TrkB) - brain-derived neurotrophic factor (BDNF). The conclusion is that greater emphasis should be placed on investigating the molecular mechanisms of fear memories in PTSD, through direct testing of patients with PTSD or the use of relevant PTSD-like models.
Collapse
Affiliation(s)
- Raül Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
4
|
Sun K, Cao C. The effects of childhood maltreatment, recent interpersonal and noninterpersonal stress, and HPA-axis multilocus genetic variation on prospective changes in adolescent depressive symptoms: A multiwave longitudinal study. Dev Psychopathol 2025; 37:543-554. [PMID: 38389485 DOI: 10.1017/s0954579424000269] [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: 02/24/2024]
Abstract
Based on a multiwave, two-year prospective design, this study is the first to examine the extent to which multilocus hypothalamic-pituitary-adrenal axis (HPA axis)-related genetic variants, childhood maltreatment, and recent stress jointly predicted prospective changes in adolescent depressive symptoms. A theory-driven multilocus genetic profile score (MGPS) was calculated to combine the effects of six common polymorphisms within HPA-axis related genes (CRHR1, NR3C1, NR3C2, FKBP5, COMT, and HTR1A) in a sample of Chinese Han adolescents (N = 827; 50.2% boys; Mage = 16.45 ± 1.36 years). The results showed that the three-way interaction of HPA-axis related MGPS, childhood maltreatment and recent interpersonal, but not noninterpersonal, stress significantly predicted prospective changes in adolescent depressive symptoms. For adolescents with high but not low HPA-axis related MGPS, exposure to severe childhood maltreatment predisposed individuals more vulnerable to recent interpersonal stress, exhibiting greater prospective changes in adolescent depressive symptoms. The findings provide preliminary evidence for the cumulative risk mechanism regarding gene-by-environment-by-environment (G × E1 × E2) interactions that underlie the longitudinal development of adolescent depressive symptoms and show effects specific to interpersonal stress.
Collapse
Affiliation(s)
- Kexin Sun
- School of Nursing and Rehabilitation, Shandong University, Jinan, China
| | - Cong Cao
- School of Nursing and Rehabilitation, Shandong University, Jinan, China
| |
Collapse
|
5
|
Liu Z, Zhai G. Cardiometabolic index and major depressive disorder: Stroke and diabetes as mediators. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111340. [PMID: 40147810 DOI: 10.1016/j.pnpbp.2025.111340] [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: 09/27/2024] [Revised: 03/08/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe affective disorder that is clearly linked to stroke and diabetes. This study aimed to investigate the mediating role of stroke and diabetes in the association between the cardiometabolic index (CMI) and MDD. METHODS This cross-sectional study analyzed data from 8312 participants in the National Health and Nutrition Examination Survey (NHANES, 2005-2018). MDD was diagnosed using the Patient Health Questionnaire-9 (PHQ-9 score > 10). Associations were evaluated using multivariate logistic/linear regression, stratified interaction analyses, restricted cubic spline (RCS) models for nonlinearity, and bootstrap mediation testing. RESULTS There was a robust positive correlation between the incidence of MDD [OR = 1.36 (95 % CI: 1.21-1.51)] and the PHQ-9 score [β = 0.55 (95 % CI: 0.37-0.73)], with a one-unit increase in CMI. The participants in CMIQ4 had a 64 % greater risk of stroke than did the participants in CMIQ1 [OR = 1.64 (95 % CI: 1.17-2.29)]. The forest plot shows that the results remained stable under the grouping of stroke, diabetes, race, gender, and age. Moreover, stroke and diabetes both exhibited partial mediating roles, with indirect effects accounting for 4.03 % and 5.37 % of the total effect, respectively. Through RCS analysis, a nonlinear correlation was observed between CMI and MDD and between CMI and diabetes. There is a linear relationship between stroke and MDD, and maintaining CMI levels below 0.518 may mitigate the risk of MDD. CONCLUSION Stroke and diabetes partially mediated the associations between CMI and MDD. However, additional prospective studies are warranted to scrutinize the impact of CMI on MDD.
Collapse
Affiliation(s)
- Zhenyu Liu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, No. 82, Xinhua South Road, Tongzhou District, Beijing 101199, China.
| | - Guangyao Zhai
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, No. 82, Xinhua South Road, Tongzhou District, Beijing 101199, China.
| |
Collapse
|
6
|
Tomlinson CJ, Ryniker L, Cook HM, Schwartz RM, Non AL. Epigenetics in persons living with HIV: trauma, coping, and FKBP5 and SLC6A4 methylation. Epigenomics 2025; 17:297-307. [PMID: 40069093 PMCID: PMC11970741 DOI: 10.1080/17501911.2025.2476389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
AIM People living with HIV (PLWH) have an increased risk for lifetime trauma and mental health difficulties. However, no studies have evaluated stress-related genes in relation to early-life adversity, lifetime trauma, or post-traumatic stress disorder (PTSD) in PLWH. METHODS Using bisulfite pyrosequencing, we evaluated DNA methylation (DNAm) in intron 7 of FKBP5, a glucocorticoid feedback regulator, and in the promoter of SLC6A4, the serotonin transporter gene, in whole blood of a random sample of 70 PLWH recruited from an HIV program, and 51 individuals 2 years later (n = 48 at both time points). Exploratory regression analyses were conducted with DNAm in relation to trauma exposure, mental health symptoms, and coping strategies. RESULTS Higher DNAm at one site of SLC6A4 was associated with lower levels of anxiety (B = -0.62 (SE = 0.23), p = 0.0109), depression (B = -0.06 (SE = 0.03), p = 0.0435), and PTSD symptoms at baseline (B = -0.03 (SE = 0.01), p = 0.0374). DNAm at FKBP5 was negatively associated with measures of anxiety (B = -0.30 (SE = 0.07), p = 0.0001) and depression symptoms (B = -0.2 (SE = 0.10), p = 0.0103). Various coping strategies were also associated with sites in both genes across time points, e.g. self-blame and substance use. CONCLUSION Our findings generate intriguing hypotheses linking mental health symptoms and DNA methylation, to be replicated with larger samples.
Collapse
Affiliation(s)
- Cassidy J. Tomlinson
- Department of Anthropology, University of California San Diego, La Jolla, CA, USA
| | - Laura Ryniker
- Department of Occupational Medicine, Epidemiology and Prevention, Northwell Health, Great Neck, NY, USA
- Center for Traumatic Stress, Resilience and Recovery at Northwell Health, Great Neck, NY, USA
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Haley M. Cook
- Department of Occupational Medicine, Epidemiology and Prevention, Northwell Health, Great Neck, NY, USA
- Center for Traumatic Stress, Resilience and Recovery at Northwell Health, Great Neck, NY, USA
| | - Rebecca M. Schwartz
- Department of Occupational Medicine, Epidemiology and Prevention, Northwell Health, Great Neck, NY, USA
- Center for Traumatic Stress, Resilience and Recovery at Northwell Health, Great Neck, NY, USA
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Amy L. Non
- Department of Anthropology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Liu Z, Zhai G. Cardiometabolic index and major depressive disorder: Stroke and diabetes as mediators. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111340. [DOI: https:/doi.org/10.1016/j.pnpbp.2025.111340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2025]
|
8
|
Krasner H, Ong CV, Hewitt P, Vida TA. From Stress to Synapse: The Neuronal Atrophy Pathway to Mood Dysregulation. Int J Mol Sci 2025; 26:3219. [PMID: 40244068 PMCID: PMC11989442 DOI: 10.3390/ijms26073219] [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: 12/20/2024] [Revised: 03/02/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Mood disorders, including major depressive disorder and bipolar disorder, are among the most prevalent mental health conditions globally, yet their underlying mechanisms remain incompletely understood. This review critically examines the neuronal atrophy hypothesis, which posits that chronic stress and associated neurobiological changes lead to structural and functional deficits in critical brain regions, contributing to mood disorder pathogenesis. Key mechanisms explored include dysregulation of neurotrophic factors such as brain-derived neurotrophic factor (BDNF), elevated glucocorticoids from stress responses, neuroinflammation mediated by cytokines, and mitochondrial dysfunction disrupting neuronal energy metabolism. These processes collectively impair synaptic plasticity, exacerbate structural atrophy, and perpetuate mood dysregulation. Emerging evidence from neuroimaging, genetic, and epigenetic studies underscores the complexity of these interactions and highlights the role of environmental factors such as early-life stress and urbanization. Furthermore, therapeutic strategies targeting neuroplasticity, including novel pharmacological agents, lifestyle interventions, and anti-inflammatory treatments, are discussed as promising avenues for improving patient outcomes. Advancing our understanding of the neuronal atrophy hypothesis could lead to more effective, sustainable interventions for managing mood disorders and mitigating their global health burden.
Collapse
Affiliation(s)
| | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, 625 Shadow Lane, Las Vegas, NV 89106, USA; (H.K.); (C.V.O.); (P.H.)
| |
Collapse
|
9
|
Banushi B, Collova J, Milroy H. Epigenetic Echoes: Bridging Nature, Nurture, and Healing Across Generations. Int J Mol Sci 2025; 26:3075. [PMID: 40243774 PMCID: PMC11989090 DOI: 10.3390/ijms26073075] [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: 03/06/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Trauma can impact individuals within a generation (intragenerational) and future generations (transgenerational) through a complex interplay of biological and environmental factors. This review explores the epigenetic mechanisms that have been correlated with the effects of trauma across generations, including DNA methylation, histone modifications, and non-coding RNAs. These mechanisms can regulate the expression of stress-related genes (such as the glucocorticoid receptor (NR3C1) and FK506 binding protein 5 (FKBP5) gene), linking trauma to biological pathways that may affect long-term stress regulation and health outcomes. Although research using model organisms has elucidated potential epigenetic mechanisms underlying the intergenerational effects of trauma, applying these findings to human populations remains challenging due to confounding variables, methodological limitations, and ethical considerations. This complexity is compounded by difficulties in establishing causality and in disentangling epigenetic influences from shared environmental factors. Emerging therapies, such as psychedelic-assisted treatments and mind-body interventions, offer promising avenues to address both the psychological and potential epigenetic aspects of trauma. However, translating these findings into effective interventions will require interdisciplinary methods and culturally sensitive approaches. Enriched environments, cultural reconnection, and psychosocial interventions have shown the potential to mitigate trauma's impacts within and across generations. By integrating biological, social, and cultural perspectives, this review highlights the critical importance of interdisciplinary frameworks in breaking cycles of trauma, fostering resilience, and advancing comprehensive healing across generations.
Collapse
Affiliation(s)
- Blerida Banushi
- School of Indigenous Studies, The University of Western Australia, Crawley, WA 6009, Australia; (J.C.); (H.M.)
| | | | | |
Collapse
|
10
|
Lugenbühl JF, Snijders C, Pernia CD, Estruch MS, Kenis G, Daskalakis NP. Corticosteroid-regulated gene transcription in SH-SY5Y-derived neurons: Insights into the mineralocorticoid and glucocorticoid receptor-mediated response. J Neuroendocrinol 2025:e70021. [PMID: 40108866 DOI: 10.1111/jne.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Post-traumatic stress disorder (PTSD) and major depressive disorder (MDD) are debilitating stress-related psychiatric disorders that can develop following exposure to traumatic events or chronic stress in some individuals. The neurobiological processes leading to disease remain largely unknown. Among others, these disorders are characterized by a dysregulated hypothalamic-pituitary-adrenal axis, which is regulated by the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). This leads to altered downstream corticosteroid-induced gene expression. In vitro models are promising tools to investigate specific neurobiological underpinnings of the stress response in the brain. Here, we investigated the suitability of SH-SY5Y-derived neurons as a cost-efficient system to study the role of GR and MR in the neuronal stress response. SH-SY5Y-derived neurons were characterized, exposed to corticosteroids, and analyzed on transcriptomic and proteomic levels. We show that (i) these neurons express sufficient and seemingly functional GR and MR to allow the study of corticosteroid-induced transcription, (ii) three corticosteroids cortisol, dexamethasone, and aldosterone, induced similar transcriptomic effects, (iii) the antagonist spironolactone mildly attenuated the effects of dexamethasone in FKBP5, DUSP1, and SUPV3L1. Mifepristone did not significantly alter the effect of aldosterone. (iv) Integrating transcriptomic alterations of these corticosteroid-exposed neurons with those of iPSC-derived neurons exposed to dexamethasone showed concordant corticosteroid-induced effects in the two in vitro systems. To determine translational validity, we compared the gene expression in these neurons with the transcriptome of postmortem brain samples from individuals with PTSD and MDD, yielding stronger negative correlations of corticosteroid effects in SH-SY5Y-derived neurons with PTSD signatures than with MDD signatures. Upon further refinement and validation, SH-SY5Y-derived neurons may serve as a simplistic tool to study neuronal corticosteroid-induced gene expression and the implicated molecular networks around GR and MR. Strengthening our insight into these receptors' functions improves our understanding of the hypothalamic-pituitary-adrenal axis, which is commonly altered in stress-related psychiatric disorders such as PTSD and MDD.
Collapse
Affiliation(s)
- Justina F Lugenbühl
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Clara Snijders
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Cameron D Pernia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Marina Soliva Estruch
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Gunter Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs) and European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML) Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
11
|
van Doeselaar L, Abromeit A, Stark T, Menegaz D, Ballmann M, Mitra S, Yang H, Rehawi G, Huettl RE, Bordes J, Narayan S, Harbich D, Deussing JM, Rammes G, Czisch M, Knauer-Arloth J, Eder M, Lopez JP, Schmidt MV. FKBP51 in glutamatergic forebrain neurons promotes early life stress inoculation in female mice. Nat Commun 2025; 16:2529. [PMID: 40087272 PMCID: PMC11912546 DOI: 10.1038/s41467-025-57952-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Early life stress (ELS) can increase vulnerability to psychiatric disorders, but also trigger resilience. FKBP51 has been associated with an increased risk for developing psychiatric disorders, specifically in interaction with ELS exposure. Here, the contribution of FKBP51 in glutamatergic forebrain neurons to the long-term consequences of ELS was investigated in both sexes. In female wild-type Fkbp5lox/lox mice, ELS exposure led to an anxiolytic phenotype and improved memory performance in a stressful context, however this ELS effect was absent in Fkbp5Nex mice. These interactive FKBP51 x ELS effects in female mice were also reflected in reduced brain region volumes, and on structural and electrophysiological properties of CA1 pyramidal neurons of the dorsal hippocampus. In contrast, the behavioral, structural and functional effects in male ELS mice were less pronounced and independent of FKBP51. RNA sequencing of the hippocampus revealed the transcription factor 4 (TCF4) as a potential regulator of the female interactive effects. Cre-dependent viral overexpression of TCF4 in female Nex-Cre mice led to similar beneficial effects on behavior as the ELS exposure. This study demonstrates a sex-specific role for FKBP51 in mediating the adaptive effects of ELS on emotional regulation, cognition, and neuronal function, implicating TCF4 as a downstream effector.
Collapse
Affiliation(s)
- Lotte van Doeselaar
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Alexandra Abromeit
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Tibor Stark
- Core Unit Neuroimaging, Max Planck Institute of Psychiatry, Munich, Germany
- Emotion Research Department, Max Planck Institute of Psychiatry, Munich, Germany
| | - Danusa Menegaz
- Core Unit Electrophysiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Markus Ballmann
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum Rechts der Isar, Munich, Germany
| | - Shiladitya Mitra
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Huanqing Yang
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Ghalia Rehawi
- Department Genes & Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rosa-Eva Huettl
- Core Unit Virus Production, Max Planck Institute of Psychiatry, Munich, Germany
| | - Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Daniela Harbich
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Genetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Gerhard Rammes
- Klinik für Anaesthesiologie und Intensivmedizin der Technischen Universität München, Klinikum Rechts der Isar, Munich, Germany
| | - Michael Czisch
- Core Unit Neuroimaging, Max Planck Institute of Psychiatry, Munich, Germany
| | - Janine Knauer-Arloth
- Department Genes & Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Matthias Eder
- Core Unit Electrophysiology, Max Planck Institute of Psychiatry, Munich, Germany
| | - Juan Pablo Lopez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
12
|
Arancibia M, Manterola M, Ríos U, Moya PR, Moran-Kneer J, Bustamante ML. The rs1360780 Variant of FKBP5: Genetic Variation, Epigenetic Regulation, and Behavioral Phenotypes. Genes (Basel) 2025; 16:325. [PMID: 40149476 PMCID: PMC11941772 DOI: 10.3390/genes16030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
FKBP5 has been of special scientific interest in the behavioral sciences since it has been involved in the pathophysiology of several mental disorders. It is a gene with pleiotropic effects which encodes the protein FKBP5, a cochaperone that decreases glucocorticoid receptor (GR) affinity for glucocorticoids by competing with FKBP4, altering the GR chaperone complex, and impairing GR activation. As a key modulator of the stress response, FKBP5 plays a critical role in regulating cortisol levels in the organism. The FKBP5 gene is regulated through a combination of transcriptional, epigenetic, post-transcriptional, and environmental mechanisms, as well as genetic polymorphisms that influence its transcription and stress responsiveness. Notably, the rs1360780 T-allele in FKBP5 significantly affects FKBP5 regulation and has been linked to stress-related disorders by influencing transcription and stress responsiveness. In this narrative review, we aim to provide an overview of the role played by the single-nucleotide polymorphism rs1360780 in the FKBP5 locus in gene expression, its epigenetic regulation, and the impact of early stress in its functioning. We discuss some brain regions with differential expression of FKBP5 and some behavioral phenotypes linked to the locus. The T-allele of rs1360780 is considered a risk variant, as it leads to high FKBP5 induction, which delays negative feedback and increases GR resistance. This results in states of relative hypercortisolemia and brain morphofunctional alterations, particularly in regions sensitive to glucocorticoid activity during critical periods of neurodevelopment. Additionally, exposure to childhood maltreatment is associated with demethylation of the glucocorticoid response elements of FKBP5, further increasing its expression levels. Among the psychological dimensions analyzed in which FKBP5 is involved are neurocognition, aggression, suicidality, and social cognition. At the level of mental disorders, the gene may play a role in the pathogenesis of post-traumatic stress disorder, depression, and bipolar disorder. In psychotic disorders, its role is less clear. This knowledge enhances the understanding of disease mechanisms that operate through psychopathological dimensions, and highlights the need to design specific, person-centered psychopharmacological and environmental therapeutic interventions.
Collapse
Affiliation(s)
- Marcelo Arancibia
- Department of Psychiatry, Faculty of Medicine, School of Medicine, Universidad de Valparaíso, Valparaíso 2360002, Chile; (M.A.)
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
- Program of Human Genetics, Biomedical Science Institute, Universidad de Chile, Independencia 8380453, Chile
| | - Marcia Manterola
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
- Program of Human Genetics, Biomedical Science Institute, Universidad de Chile, Independencia 8380453, Chile
| | - Ulises Ríos
- Department of Psychiatry, Faculty of Medicine, School of Medicine, Universidad de Valparaíso, Valparaíso 2360002, Chile; (M.A.)
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
| | - Pablo R. Moya
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
- Institute of Physiology, Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Javier Moran-Kneer
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
- Department of Clinical Psychology, School of Psychology, Faculty of Social Sciences, Universidad de Valparaíso, Valparaíso 2341369, Chile
| | - M. Leonor Bustamante
- Center for Translational Studies in Stress and Mental Health (C-ESTRES), Faculty of Sciences, Universidad de Valparaíso, Valparaíso 2360102, Chile; (M.M.); (P.R.M.); (J.M.-K.)
- Program of Human Genetics, Biomedical Science Institute, Universidad de Chile, Independencia 8380453, Chile
| |
Collapse
|
13
|
Seah C, Sidamon-Eristoff AE, Huckins LM, Brennand KJ. Implications of gene × environment interactions in post-traumatic stress disorder risk and treatment. J Clin Invest 2025; 135:e185102. [PMID: 40026250 PMCID: PMC11870735 DOI: 10.1172/jci185102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Exposure to traumatic stress is common in the general population. Variation in the brain's molecular encoding of stress potentially contributes to the heterogeneous clinical outcomes in response to traumatic experiences. For instance, only a minority of those exposed to trauma will develop post-traumatic stress disorder (PTSD). Risk for PTSD is at least partially heritable, with a growing number of genetic factors identified through GWAS. A major limitation of genetic studies is that they capture only the genetic component of risk, whereas PTSD by definition requires an environmental traumatic exposure. Furthermore, the extent, timing, and type of trauma affects susceptibility. Here, we discuss the molecular mechanisms of PTSD risk together with gene × environment interactions, with a focus on how either might inform genetic screening for individuals at high risk for disease, reveal biological mechanisms that might one day yield novel therapeutics, and impact best clinical practices even today. To close, we discuss the interaction of trauma with sex, gender, and race, with a focus on the implications for treatment. Altogether, we suggest that predicting, preventing, and treating PTSD will require integrating both genotypic and environmental information.
Collapse
Affiliation(s)
- Carina Seah
- Department of Genetics and Genomics and
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anne Elizabeth Sidamon-Eristoff
- Department of Psychiatry, Division of Molecular Psychiatry
- Interdepartmental Neuroscience Program, Wu Tsai Institute, and
- MD-PhD Program, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Kristen J. Brennand
- Department of Genetics and Genomics and
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Division of Molecular Psychiatry
- Interdepartmental Neuroscience Program, Wu Tsai Institute, and
| |
Collapse
|
14
|
Hanson T, Spencer S, Harker SA, Barry F, Burton P, Beauchemin J, Mennenga SE, Braden BB, D'Sa V, Koinis-Mitchell D, Deoni SC, Lewis CR. Peripheral DNA Methylation of Cortisol- and Serotonin-Related Genes Predicts Hippocampal Volume in a Pediatric Population. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100421. [PMID: 39867566 PMCID: PMC11758844 DOI: 10.1016/j.bpsgos.2024.100421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hippocampal volume increases throughout early development and is an important indicator of cognitive abilities and mental health. However, hippocampal development is highly vulnerable to exposures during development, as seen by smaller hippocampal volume and differential epigenetic programming in genes implicated in mental health. However, few studies have investigated hippocampal volume in relation to the peripheral epigenome across development, and even less is known about potential genetic moderators. Therefore, in this study, we explored relationships between hippocampal volume and peripheral DNA methylation of mental health-related genes, specifically NR3C1, FKBP5, and SLC6A4, throughout early development and whether these associations were moderated by age or genotype. Methods Bilateral hippocampal volume was computed from T2-weighted images through FreeSurfer, and DNA methylation was measured from saliva using the Illumina MethylationEPIC microarray in a pediatric population (N = 248, females = 112, meanage = 5.13 years, SDage = 3.60 years). Results Multiple linear regression and bootstrapping analyses revealed that DNA methylation of NR3C1, FKBP5, and SLC6A4 was associated with hippocampal volume and that these relationships were moderated by age and gene-specific variants. Conclusions These findings support the validity of peripheral DNA methylation profiles for indirectly assessing hippocampal volume and development and underscore the importance of genotype and age considerations in research. Therefore, peripheral epigenetic profiles may be a promising avenue for investigating the impacts of early-life stress on brain structure and subsequent mental health outcomes.
Collapse
Affiliation(s)
- Taena Hanson
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Sophia Spencer
- Department of Psychology, Arizona State University, Tempe, Arizona
| | | | - Fatoumata Barry
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Phoebe Burton
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | | | - B. Blair Braden
- College of Health Solutions, Arizona State University, Tempe, Arizona
| | - Viren D'Sa
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Daphne Koinis-Mitchell
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Sean C.L. Deoni
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
- Advanced Baby Imaging Laboratory, Rhode Island Hospital, Providence, Rhode Island
| | - Candace R. Lewis
- Department of Psychology, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Neurogenomics, Translational Genomics Research Institute, Phoenix, Arizona
| |
Collapse
|
15
|
Johnson JL. FKBP51 functions in the regulation of circadian rhythm and Alzheimer's disease. Cell Stress Chaperones 2025; 30:81-83. [PMID: 39933601 PMCID: PMC11889542 DOI: 10.1016/j.cstres.2025.02.002] [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: 01/15/2025] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The FK506-binding protein 51 (FKBP51) is an important regulator of glucocorticoid receptor activity and an Hsp90 cochaperone. FKBP51 has previously been identified as a drug target due to its roles in stress-related disorders and pain tolerance. Two recent publications in Cell Stress and Chaperones further explore FKBP51 functions. To understand whether FKBP51 plays a role in sleep disturbances linked to stress disorders, one study examined the role of FKBP51 in regulating the circadian rhythm. Broadening the range of Hsp90 cochaperone function, the other article summarized the role of multiple cochaperones in Alzheimer's disease, discussing how cochaperones affect both Aβ and tau. They emphasize the role of FKBP51 in promoting tau pathogenesis and discuss the small molecule LA1011, which binds Hsp90 and competes with Hsp90-FKBP51 interaction. Further studies with LA1011 may lead to new treatments for Alzheimer's disease and will help clarify the contributions of FKBP51 to human disorders.
Collapse
Affiliation(s)
- Jill L Johnson
- Department of Biological Sciences and the Center for Reproductive Biology, University of Idaho, Moscow, ID 83844, USA.
| |
Collapse
|
16
|
Goltser-Dubner T, Benarroch F, Lavon M, Amer R, Canetti L, Giesser R, Kianski E, Martin J, Pevzner D, Blum Weinberg P, Ben-Ari A, Bar-Nitsan M, Alon S, Yshai S, Lotan A, Galili-Weisstub E, Segman R, Shalev A. Childhood trauma cortisol and immune cell glucocorticoid transcript levels are associated with increased risk for suicidality in adolescence. Mol Psychiatry 2025:10.1038/s41380-025-02923-3. [PMID: 39994424 DOI: 10.1038/s41380-025-02923-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 12/28/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Rising adolescent suicide rates present a growing unmet need. Childhood trauma (CT) has been associated with altered cortisol dynamics and immune cell glucocorticoid reactivity, yet their additive longer-term contributions to later suicide outcomes are less clear. The current study compared CT scores, resting salivary free cortisol and mononuclear cell gene expression levels of the nuclear receptor, subfamily 3, member 1 (NR3C1) coding the glucocorticoid receptor, and its co-chaperons FKBP prolyl isomerase 5 (FKBP5) and KIT Ligand (KITLG), between a cohort of adolescents presenting with a suicidal crisis requiring hospital treatment, and matched healthy controls. Childhood trauma scores and glucocorticoid measures were significantly altered among suicidal adolescents, and CT scores correlated with mononuclear cell glucocorticoid transcripts. Both CT scores and glucocorticoid measures explained substantial additive portions of the variance in adolescent suicidality. Long-term perturbations in cortisol dynamics and immune cell glucocorticoid response elements denote dysregulated immune stress reactivity, and may possess value in prediction and point to modifiable-risk factors in prevention of clinically significant suicidality during the brittle period of adolescence, years after childhood trauma exposure.
Collapse
Affiliation(s)
- Tanya Goltser-Dubner
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fortu Benarroch
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Lavon
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Reaan Amer
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Laura Canetti
- Department of Psychology, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Giesser
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ella Kianski
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Josef Martin
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dalya Pevzner
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pnina Blum Weinberg
- The Donald Cohen Child and Adolescent Psychiatry Department, Eitanim Psychiatric Hospital, The Jerusalem Mental Health Center, Jerusalem, Israel
| | - Amichai Ben-Ari
- Department of Behavioral Sciences, Ariel University, Ariel, Israel
| | | | - Shaked Alon
- Department of Behavioral Sciences, Ariel University, Ariel, Israel
| | - Shai Yshai
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Lotan
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esti Galili-Weisstub
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronen Segman
- Molecular Psychiatry Laboratory, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Amit Shalev
- The Herman-Danna Department of Child and Adolescent Psychiatry, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
17
|
Yang TC, Tsai JP, Hsu H, Chen YC, Liaw YC, Hsu SY, Yang HJ, Liaw YP. Delving Into the Interaction Between Exercise and Diabetes on Methylation of the FKBP5 Gene. J Diabetes Res 2025; 2025:1162708. [PMID: 40017583 PMCID: PMC11865466 DOI: 10.1155/jdr/1162708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Objective: FKBP5 is a critical gene involved in regulating the hypothalamic-pituitary-adrenal (HPA) axis and stress response. Aberrant DNA methylation at FKBP5 cytosine-phosphate-guanine (CpG) sites, such as cg22363520 and cg00862770, has been implicated in mental health disorders and metabolic diseases, including Type 2 diabetes. Exercise is a modulator of DNA methylation and metabolic health. This study investigates the interaction between exercise, diabetes, and FKBP5 methylation at cg22363520 and cg00862770 and explores their implications for mental health and disease development. Materials and Methods: FKBP5 methylation levels at cg22363520 and cg00862770 were analyzed in a cohort stratified by diabetes and exercise. Multiple linear regression models assessed the main effects and interactions of exercise and diabetes on FKBP5 methylation, with further stratified analyses for site-specific effects. Results: Exercise and diabetes showed significant and site-specific effects on FKBP5 methylation at cg22363520 and cg00862770. At cg22363520, exercise significantly reduced methylation levels in nondiabetic participants (β = -0.00195, p = 0.0157), while no significant effect was observed in diabetic individuals. Conversely, at cg00862770, exercise significantly decreased methylation levels in diabetic participants (β = -0.00611, p = 0.0081), with no significant effect in the nondiabetic group. Diabetes itself was associated with increased FKBP5 methylation at both sites, particularly in individuals without regular exercise. Additionally, significant interaction effects between exercise and diabetes were identified for both cg22363520 (p = 0.0336) and cg00862770 (p = 0.0021), highlighting the interplay between metabolic status and physical activity in regulating FKBP5 methylation. Conclusion: This study demonstrates that the effects of exercise on FKBP5 methylation are site-specific and influenced by diabetes status. Exercise reduces methylation at cg22363520 in nondiabetics and at cg00862770 in diabetics, indicating its role in modulating epigenetic regulation of stress and metabolic pathways. These findings underscore the interplay between exercise, diabetes, and FKBP5 methylation, with potential implications for improving mental health and metabolic outcomes.
Collapse
Affiliation(s)
- Teng-Chi Yang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Department of Medical Affairs, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
| | - Jen Pi Tsai
- Division of Nephrology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi School Foundation, Hualien County, Taiwan
| | - Honda Hsu
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi School Foundation, Hualien County, Taiwan
- Division of Plastic Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
| | - Yen-Chung Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Department of Neurology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Yi-Chia Liaw
- Neurological Institute, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Shu Yi Hsu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Hao Jan Yang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Institute of Medicine, Chug Shan Medical University, Taichung City, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
18
|
Taylor WW, Korobkova L, Bhinderwala N, Dias BG. Toward Understanding and Halting Legacies of Trauma. Biol Psychiatry 2025:S0006-3223(25)00108-8. [PMID: 39956254 DOI: 10.1016/j.biopsych.2025.02.010] [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: 05/15/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Echoes of natural and anthropogenic stressors not only reverberate within the physiology, biology, and neurobiology of the generation directly exposed to them but also within the biology of future generations. With the intent of understanding this phenomenon, significant efforts have been made to establish how exposure to psychosocial stress, chemicals, over- and undernutrition, and chemosensory experiences exert multigenerational influences. From these studies, we are gaining new appreciation for how negative environmental events experienced by one generation impact future generations. In this review, we first outline the need to operationally define dimensions of negative environmental events in the laboratory and the routes by which the impact of such events are felt through generations. Next, we discuss molecular processes that cause the effects of negative environmental events to be initiated in the exposed generation and then perpetuated across generations. Finally, we discuss how legacies of flourishing can be engineered to halt or reverse multigenerational influences of negative environmental events. In summary, this review synthesizes our current understanding of the concept, causes, and consequences of multigenerational echoes of stress and looks for opportunities to halt them.
Collapse
Affiliation(s)
- William Wesley Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Laura Korobkova
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Nabeel Bhinderwala
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Brian George Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California; Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
19
|
Obliosca JM, Vest O, Patel D, Ferguson T, Smith K, Christy D, Powers A, Smith AK, Xu Y, Tison CK. Surface Plasmon Resonance Imaging-Based Platform Enables Detection of Single, Site-Specific 5-Methylcytosine Associated with Post-traumatic Stress Disorder (PTSD). ACS Pharmacol Transl Sci 2025; 8:522-532. [PMID: 39974633 PMCID: PMC11833716 DOI: 10.1021/acsptsci.4c00628] [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: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 02/21/2025]
Abstract
While identification of epigenetic changes in individuals with psychiatric dysfunctions such as post-traumatic stress disorder (PTSD) is paramount to genomic research, there is no rapid and simplified way to detect an epigenetic marker such as DNA methylation in genes. Here, we introduce a faster, simpler method to detect methylation in the form of 5-methylcytosine (5mC, termed as PTSD-associated base) in known CpG sites using nanoenhanced surface plasmon resonance imaging-based epigenetic assay (EpiNanoSPRi). This assay platform simultaneously detects a panel of single, site-specific PTSD bases in target genes or regions using an anti-5mC antibody and a universal nanoenhancer on a gold-coated sensing chip. Not only can EpiNanoSPRi identify 5mC at the single-base level, but it also can quantify the extent of DNA methylation. Our method is superior and more practical to bisulfite-based DNA sequencing techniques as it will significantly reduce DNA methylation identification from 4 days (e.g., DNA Sequencing) to 9 h without massive analysis workflow. This platform can potentially be applied to diagnose other psychiatric disorders such as Alzheimer's, Parkinson's, dementia, schizophrenia, and Huntington's diseases.
Collapse
Affiliation(s)
- Judy M. Obliosca
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Olivia Vest
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Dimpal Patel
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Tammy Ferguson
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Kelsi Smith
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Dan Christy
- Systems
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Abigail Powers
- Emory
University School of Medicine, 101 Woodruff Circle WMB, Atlanta, Georgia 30322, United States
| | - Alicia K. Smith
- Emory
University School of Medicine, 101 Woodruff Circle WMB, Atlanta, Georgia 30322, United States
| | - Yang Xu
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| | - Christopher K. Tison
- Biotech
Group, Luna Labs USA, LLC., 706 Forest Street, Suite A, Charlottesville, Virginia 22903, United States
| |
Collapse
|
20
|
Merrill SM, Konwar C, Fraihat Z, Parent J, Dajani R. Molecular insights into trauma: A framework of epigenetic pathways to resilience through intervention. MED 2025; 6:100560. [PMID: 39708797 DOI: 10.1016/j.medj.2024.11.013] [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: 04/22/2024] [Revised: 10/01/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Experiences of complex trauma and adversity, especially for children, are ongoing global crises necessitating adaptation. Bioadaptability to adversity and its health consequences emphasizes the dynamism of adaptation to trauma and the potential for research to inform intervention strategies. Epigenetic variability, particularly DNA methylation, associates with chronic adversity while allowing for resilience and adaptability. Epigenetics, including age- and site-specific changes in DNA methylation, gene-environment interactions, pharmacological responses, and biomarker characterization and evaluation, may aid in understanding trauma responses and promoting well-being by facilitating psychological and biological adaptation. Understanding these molecular processes provides a foundation for a biologically adaptive framework to shift public health strategies from restorative to long-term adaptation and resilience. Psychological, cultural, and biological trauma must be addressed in innovative interventions for vulnerable populations, particularly children and adolescents. Understanding molecular changes may provide a biopsychosocial perspective for culturally sensitive, evidence-based interventions that promote resilience and thriving in new settings.
Collapse
Affiliation(s)
- Sarah M Merrill
- Department of Psychology, University of Massachusetts Lowell, Lowell, MA, USA.
| | - Chaini Konwar
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Zaid Fraihat
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Justin Parent
- Department of Psychology, University of Massachusetts Lowell, Lowell, MA, USA; Department of Psychology, College of Health Sciences, University of Rhode Island, Kingston, RI, USA; Emma Pendleton Bradley Hospital, East Providence, RI, USA
| | - Rana Dajani
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan.
| |
Collapse
|
21
|
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
|
22
|
Zhen-Duan J, Canenguez KM, Wilson AE, Gu Y, Valluri HG, Chavez AD, Argentieri MA, Schachter AB, Wu H, Baccarelli AA, Daviglus ML, Wassertheil-Smoller S, Warner ET, Shields AE. Religion, spirituality, and DNA methylation in HPA-axis genes among Hispanic/Latino adults. Epigenomics 2025; 17:155-166. [PMID: 39707707 PMCID: PMC11812325 DOI: 10.1080/17501911.2024.2442293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
AIM Investigate associations between religion and spirituality (R&S) and DNA methylation of four HPA-axis genes (i.e. 14 CpG sites) among 992 adults from the Hispanic Community Health Study/Study of Latinos cohorts. METHODS We assessed 1) the association between R&S measures and mean percent methylation overall and stratified by nativity status (US-born or immigrant) and 2) if interactions between R&S and methylation differed by nativity status. RESULTS Among individuals with the FKBP5 CC genotype, increased spirituality scores were associated with significantly lower methylation levels among immigrants, compared to US-born participants. Organizational religiosity (e.g. service attendance) was associated with increased FKBP5 (CC genotype) methylation among immigrants. CONCLUSION R&S may influence HPA-axis functioning differently based on nativity status; a finding that could offer insight into mechanisms leading to health disparities.
Collapse
Affiliation(s)
- Jenny Zhen-Duan
- Disparities Research Unit, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Katia M. Canenguez
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anna E. Wilson
- Disparities Research Unit, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Health Policy Research Center, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yue Gu
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Harshitha G. Valluri
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Alejandra D. Chavez
- Disparities Research Unit, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - M. Austin Argentieri
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anna Boonin Schachter
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Martha L. Daviglus
- Institute for Minority Health Research, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | | | - Erica T. Warner
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alexandra E. Shields
- Harvard/MGH Center on Genomics, Vulnerable Populations, and Health Disparities, Mongan Institute, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Mongan Institute, Clinical Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
23
|
Kovarova V, Bordes J, Mitra S, Narayan S, Springer M, Brix LM, Deussing JM, Schmidt MV. Deep phenotyping reveals CRH and FKBP51-dependent behavioral profiles following chronic social stress exposure in male mice. Neuropsychopharmacology 2025; 50:556-567. [PMID: 39438757 PMCID: PMC11736030 DOI: 10.1038/s41386-024-02008-9] [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: 05/15/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
The co-chaperone FKBP51, encoded by FKBP5 gene, is recognized as a psychiatric risk factor for anxiety and depressive disorders due to its crucial role in the stress response. Another key modulator in stress response regulation is the corticotropin releasing hormone (CRH), which is co-expressed with FKBP51 in many stress-relevant brain-regions and cell-types. Together, they intricately influence the balance of the hypothalamic-pituitary-adrenal (HPA) axis, one of the primary stress response systems. Previous research underscores the potential moderating effects these genes have on the regulation of the stressful life events towards the vulnerability of major depressive disorder (MDD). However, the specific function of FKBP51 in CRH-expressing neurons remains largely unexplored. Here, through deep behavioral phenotyping, we reveal heightened stress effects in mice lacking FKBP51 in CRH co-expressing neurons (CRHFKBP5-/-), particularly evident in social contexts. Our findings highlight the importance of considering cell-type specificity and context in comprehending stress responses and advocate for the utilization of machine-learning-driven phenotyping of mouse models. By elucidating these intricacies, we lay down the groundwork for personalized interventions aimed at enhancing stress resilience and individual well-being.
Collapse
Affiliation(s)
- Veronika Kovarova
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Shiladitya Mitra
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Sowmya Narayan
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 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, Munich, Germany
| | - Lea Maria Brix
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804, Munich, Germany
| | - Jan M Deussing
- Research Group Molecular Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
24
|
Smeeth D, Ecker S, Chervova O, McEwen F, Karam E, Beck S, Pluess M. War Exposure and DNA Methylation in Syrian Refugee Children and Adolescents. JAMA Psychiatry 2025; 82:191-200. [PMID: 39565630 PMCID: PMC11579893 DOI: 10.1001/jamapsychiatry.2024.3714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/16/2024] [Indexed: 11/21/2024]
Abstract
Importance Exposure to war is associated with poor mental health outcomes. Adverse and traumatic experiences can lead to long-lasting DNA methylation changes, potentially mediating the link between adversity and mental health. To date, limited studies have investigated the impact of war on DNA methylation in children or adolescents, hampering our understanding of the biological impact of war exposure. Objective To identify salivary DNA methylation differences associated with war exposure in refugee children and adolescents. Design, Setting, and Participants This cohort study included Syrian refugee children and adolescents, and their primary caregiver were recruited from tented settlements in Lebanon. Data collection was carried out in 2 waves, 1 year apart, from October 2017 to January 2018 and October 2018 to January 2019. Children and their caregiver were interviewed, and children provided saliva samples for DNA extraction. Data analysis was conducted in 2022, 2023, and 2024. Exposure War exposure assessed by interviewing children and their caregiver using the War Events Questionnaire. Main Outcomes and Measures Salivary DNA methylation levels were assayed with the Infinium MethylationEPIC BeadChip (Illumina). Epigenetic aging acceleration was estimated using a set of preexisting epigenetic aging clocks. A literature search was conducted to identify previously reported DNA methylation correlates of childhood trauma. Results The study population included 1507 children and adolescents (mean [SD] age, 11.3 [2.4] years; age range, 6-19 years; 793 female [52.6%]). A total of 1449 children provided saliva samples for DNA extraction in year 1, and 872 children provided samples in year 2. Children who reported war events had a number of differentially methylated sites and regions. Enrichment analyses indicated an enrichment of gene sets associated with transmembrane transport, neurotransmission, and intracellular movement in genes that exhibited differential methylation. Sex-stratified analyses found a number of sex-specific DNA methylation differences associated with war exposure. Only 2 of 258 (0.8%) previously reported trauma-associated DNA methylation sites were associated with war exposure (B = -0.004; 95% CI, -0.005 to -0.003; Bonferroni P = .04 and B = -0.005; 95% CI, -0.006 to -0.004; Bonferroni P = .03). Any war exposure or bombardment was nominally associated with decreased epigenetic age using the Horvath multitissue clock (B = -0.39; 95% CI, -0.63 to -0.14; P = .007 and B = -0.42; 95% CI, -0.73 to -0.11; P = .002). Conclusions and Relevance In this cohort of Syrian refugee children and adolescents, war exposure was associated with a small number of distinct differences in salivary DNA methylation.
Collapse
Affiliation(s)
- Demelza Smeeth
- Department of Biological and Experimental Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
- School of Psychology, University of Surrey, Guildford, United Kingdom
| | - Simone Ecker
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Olga Chervova
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Fiona McEwen
- Department of Biological and Experimental Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
- Department of War Studies, King’s College London, London, United Kingdom
| | - Elie Karam
- Department of Psychiatry and Clinical Psychology, Balamand University, St Georges Hospital University Medical Center, Institute for Development, Research, Advocacy and Applied Care, Beirut, Lebanon
| | - Stephan Beck
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Michael Pluess
- Department of Biological and Experimental Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
- School of Psychology, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
25
|
Logue E, Nemeroff CB. Sex Differences in the Associations Among Early Life Adversity, Inflammation, and Cognition. Biomolecules 2025; 15:161. [PMID: 40001464 PMCID: PMC11853676 DOI: 10.3390/biom15020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Early life adversity (ELA) has long been recognized to negatively impact a variety of health outcomes, with increasingly recognized long-term implications for neurocognitive function. ELA may affect the brain through multiple mechanisms, including chronic inflammation. One potential moderator of the pathway from ELA to neuroinflammation to cognitive dysfunction is sex. ELA may leave females potentially even more vulnerable to cognitive impairment in later life. This review discusses the influence of ELA on cognitive function across much of the lifespan, how inflammation is implicated in this process, and the current state of knowledge regarding sex differences in these relationships. We conclude with a discussion of unanswered questions and suggestions for future research, including the incorporation of genetic data.
Collapse
Affiliation(s)
- Erin Logue
- Department of Psychiatry and Behavioral Sciences, Mulva Clinic for Neurosciences, The University of Texas at Austin Dell Medical School, Austin, TX 78712, USA
- Department of Neurology, Mulva Clinic for Neurosciences, The University of Texas at Austin Dell Medical School, Austin, TX 78712, USA
| | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Mulva Clinic for Neurosciences, The University of Texas at Austin Dell Medical School, Austin, TX 78712, USA
- Institute for Early Life Adversity, The University of Texas at Austin Dell Medical School, Austin, TX 78712, USA
| |
Collapse
|
26
|
Polli A, Nijs J, Thienpont B. Epigenetics as the molecular substrate of multimodal lifestyle approaches for patients with persistent pain. Braz J Phys Ther 2025; 29:101170. [PMID: 39742735 PMCID: PMC11751397 DOI: 10.1016/j.bjpt.2024.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Affiliation(s)
- Andrea Polli
- Pain in Motion (PiM) international research group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Rehabilitation Sciences & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium; Department of Public Health and Primary Care, Centre for Environment & Health, KU Leuven, Leuven, Belgium; Flanders Research Foundation - FWO, Belgium
| | - Jo Nijs
- Pain in Motion (PiM) international research group, Department of Physiotherapy, Human Physiology and Anatomy, Faculty of Rehabilitation Sciences & Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium; Department of Physical Medicine and Physiotherapy, University Hospital Brussels, Brussels, Belgium; Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium; KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven 3000, Leuven, Belgium; KU Leuven Cancer Institute (LKI), KU Leuven 3000, Leuven, Belgium
| |
Collapse
|
27
|
Castro-Quintas A, Palma-Gudiel H, Eixarch E, San Martín González N, Röh S, Sauer S, Rex-Haffner M, Monteserin-Garcia JL, de la Fuente-Tomás L, Crispi F, Garcia Portilla MP, Binder EB, Fañanas L. Placental epigenetic signatures of maternal distress in glucocorticoid-related genes and newborn outcomes: A study of Spanish primiparous women. Eur Neuropsychopharmacol 2025; 90:36-47. [PMID: 39504602 DOI: 10.1016/j.euroneuro.2024.10.001] [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: 04/24/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Maternal stress during pregnancy can impact offspring health, increasing the risk of neuropsychiatric disorders. The human placenta plays a crucial role in understanding this effect, influencing fetal programming as it connects maternal and fetal circulation. Our hypothesis centers on maternal stress influencing children's outcomes through placental DNA methylation, targeting three cortisol-regulating genes: NR3C1, FKBP5, and HSD11B2. In this pilot study, chorionic villi and maternal decidua placental layers from 45 mother-infant dyads (divided into two groups based on high/low maternal stress exposure) were analyzed for DNA methylation at the genes of interest via targeted bisulfite sequencing. Pregnant women provided four saliva samples throughout a day for cortisol determinations and were assessed for the presence of depressive symptoms at each of the three trimesters of pregnancy. Newborns underwent neurodevelopmental assessments and salivary cortisol evaluations at 7 weeks. Increased maternal diurnal cortisol levels in the first trimester of pregnancy was significantly associated with elevated DNA methylation at exon 1D of the NR3C1 gene and lower DNA methylation at intron 7 of the FKBP5 gene, both in chorionic villi samples. Elevated DNA methylation at introns 1 and 7 of FKBP5 in the maternal decidua were strongly linked to an anticipated delivery. DNA methylation at the HSD11B2 promoter region was uniformly low across all placental samples. No associations with newborn neurodevelopment were found. These results emphasize the importance of exploring layer-specific methylation differences at distinct pregnancy stages, highlighting the complex interplay between maternal stress, placental epigenetic modifications, and fetal development throughout the prenatal period.
Collapse
Affiliation(s)
- Agueda Castro-Quintas
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain
| | - Helena Palma-Gudiel
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain
| | - Elisenda Eixarch
- Department of Surgery and Surgical specializations, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research on Rare Diseases (CIBER of Rare Diseases, CIBERER), Madrid, Spain
| | - Nerea San Martín González
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain
| | - Simone Röh
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Susann Sauer
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Monika Rex-Haffner
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jose Luis Monteserin-Garcia
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain
| | - Lorena de la Fuente-Tomás
- Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain; Department of Psychology, University of Oviedo, Oviedo, Spain
| | - Fatima Crispi
- Department of Surgery and Surgical specializations, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain; BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research on Rare Diseases (CIBER of Rare Diseases, CIBERER), Madrid, Spain
| | - Maria Paz Garcia Portilla
- Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain; Department of Psychiatry, University of Oviedo, Oviedo, Spain; Servicio de Salud del Principado de Asturias (SESPA), Oviedo, Spain
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lourdes Fañanas
- Department of Evolutionary Biology, Ecology and Environmental Sciences, Faculty of Biology, University of Barcelona, Barcelona, Spain; Biomedicine Institute of the University of Barcelona (IBUB), Barcelona, Spain; Health Institut Carlos III, Network Centre for Biomedical Research in Mental Health (CIBER of Mental Health, CIBERSAM), Madrid, Spain.
| |
Collapse
|
28
|
Hartmann J, Klengel C, Dillmann LJ, Hisey EE, Hafner K, Shukla R, Soliva Estruch M, Bajaj T, Ebert T, Mabbott KG, Rostin L, Philipsen A, Carlezon WA, Gisabella B, McCullumsmith RE, Vergis JM, Klengel T, Berretta S, Daskalakis NP, Pantazopoulos H, Gassen NC, Ressler KJ. SKA2 enhances stress-related glucocorticoid receptor signaling through FKBP4-FKBP5 interactions in neurons. Proc Natl Acad Sci U S A 2024; 121:e2417728121. [PMID: 39705315 DOI: 10.1073/pnas.2417728121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/15/2024] [Indexed: 12/22/2024] Open
Abstract
Genes involved in regulating the hypothalamic-pituitary-adrenal (HPA) axis, including the glucocorticoid receptor (GR), are linked to various stress-related psychopathologies including bipolar disorder as well as other mood and trauma-related disorders. The protein product of the cell cycle gene, SKA2, is a GR interaction partner in peripheral cells. However, the precise roles of SKA2 in stress and GR signaling in the brain, specifically in nonreplicating postmitotic neurons, and its involvement in HPA axis regulation remain unclear. Here, we demonstrate, using diverse in vitro cell assays, a mechanism by which SKA2 promotes GR signaling through enhancing GR-FKBP4 interaction leading to dissociation of FK506-bindingprotein 51 (FKBP5) from the complex. FKBP4 and FKBP5 are cochaperones known to regulate GR function in opposite directions. Notably in mice, SKA2 in Crh+ neurons of the paraventricular nucleus of the hypothalamus is crucial for HPA axis responsiveness and for maintaining the negative feedback loop underlying allostasis. Moreover, we show that SKA2 expression is increased in postmortem human hippocampus and amygdala from individuals with BD. Our study highlights a critical role of SKA2 in HPA axis function, adds to the understanding of the molecular basis of stress-related psychiatric disorders, and points to potential targets for intervention.
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Larissa J Dillmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Erin E Hisey
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
| | - Rammohan Shukla
- Department of Neuroscience, University of Wyoming, Laramie, WY 82071
| | - Marina Soliva Estruch
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Thomas Bajaj
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Tim Ebert
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Katharine G Mabbott
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Luise Rostin
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital, Bonn 53127, Germany
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | | | - John M Vergis
- Department of Neurosciences, University of Toledo, Toledo, OH 43614
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nils C Gassen
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich 80804, Germany
- Department of Psychiatry and Psychotherapy, Research Group Neurohomeostasis, University Hospital, Boon 53127, Germany
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478
| |
Collapse
|
29
|
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
|
30
|
Li Y, Zhang B, Yang Y, Su P, Samsom JN, Wong AHC, Liu F. Sex and Age Differences in Glucocorticoid Signaling After an Aversive Experience in Mice. Cells 2024; 13:2041. [PMID: 39768133 PMCID: PMC11674875 DOI: 10.3390/cells13242041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND glucocorticoids may play an important role in the formation of fear memory, which is relevant to the neurobiology of post-traumatic stress disorder (PTSD). In our previous study, we showed the glucocorticoid receptor (GR) forms a protein complex with FKBP51, which prevents translocation of GR into the nucleus to affect gene expression; this complex is elevated in PTSD patients and by fear-conditioned learning in mice, and disrupting this complex blocks the storage and retrieval of fear-conditioned memories. The timing of release of glucocorticoid relative to the formation of a traumatic memory could be important in this process, and remains poorly understood. METHODS AND RESULTS we mapped serum corticosterone over time after fear conditioning in cardiac blood samples from male and female mice, as well as adult and aged mice using ELISA. We show a significant alteration in serum corticosterone after conditioning; notably, levels spike after 30 min but drop lower than unconditioned controls after 24 h. We further investigate the effect of glucocorticoid on GR phosphorylation and localization in HEK 293T cells by Western blot. Hydrocortisone treatment promotes phosphorylation and nuclear translocation of GR. CONCLUSIONS these data contribute to our understanding of the processes linking stress responses to molecular signals and fear memory, which is relevant to understanding the shared mechanisms related to PTSD.
Collapse
Affiliation(s)
- Yun Li
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100054, China; (Y.L.); (P.S.)
| | - Bin Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Institute of Mental Health and Drug Discovery, School of Mental Health, Wenzhou Medical University, Ouhai District, Wenzhou 325000, China; (B.Z.); (Y.Y.)
| | - Youhua Yang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Institute of Mental Health and Drug Discovery, School of Mental Health, Wenzhou Medical University, Ouhai District, Wenzhou 325000, China; (B.Z.); (Y.Y.)
| | - Ping Su
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100054, China; (Y.L.); (P.S.)
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada; (J.N.S.); (A.H.C.W.)
| | - James Nicholas Samsom
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada; (J.N.S.); (A.H.C.W.)
| | - Albert H. C. Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada; (J.N.S.); (A.H.C.W.)
- Department of Pharmacology & Toxicology, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Institutes of Medical Science, University of Toronto, 1 King’s College Cir., Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Fang Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Institute of Mental Health and Drug Discovery, School of Mental Health, Wenzhou Medical University, Ouhai District, Wenzhou 325000, China; (B.Z.); (Y.Y.)
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada; (J.N.S.); (A.H.C.W.)
- Department of Pharmacology & Toxicology, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Institutes of Medical Science, University of Toronto, 1 King’s College Cir., Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Physiology, University of Toronto, 1 King’s College Cir., Toronto, ON M5S 1A8, Canada
| |
Collapse
|
31
|
Gebru NT, Hill SE, Blair LJ. Genetically engineered mouse models of FK506-binding protein 5. J Cell Biochem 2024; 125:e30374. [PMID: 36780339 PMCID: PMC10423308 DOI: 10.1002/jcb.30374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/25/2022] [Accepted: 01/15/2023] [Indexed: 02/14/2023]
Abstract
FK506 binding protein 51 (FKBP51) is a molecular chaperone that influences stress response. In addition to having an integral role in the regulation of steroid hormone receptors, including glucocorticoid receptor, FKBP51 has been linked with several biological processes including metabolism and neuronal health. Genetic and epigenetic alterations in the gene that encodes FKBP51, FKBP5, are associated with increased susceptibility to multiple neuropsychiatric disorders, which has fueled much of the research on this protein. Because of the complexity of these processes, animal models have been important in understanding the role of FKBP51. This review examines each of the current mouse models of FKBP5, which include whole animal knockout, conditional knockout, overexpression, and humanized mouse models. The generation of each model and observational details are discussed, including behavioral phenotypes, molecular changes, and electrophysiological alterations basally and following various challenges. While much has been learned through these models, there are still many aspects of FKBP51 biology that remain opaque and future studies are needed to help illuminate these current gaps in knowledge. Overall, FKBP5 continues to be an exciting potential target for stress-related disorders.
Collapse
Affiliation(s)
- Niat T. Gebru
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
| | - Shannon E. Hill
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
| | - Laura J. Blair
- USF Health Byrd Alzheimer’s Institute, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Department of Molecular Medicine, University of South Florida, 4001 E. Fletcher Ave. Tampa, Florida 33613, United States
- Research Service, James A. Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, United States
| |
Collapse
|
32
|
Tao Y, Jin M, Zhang H, Ran M, Xu H, Zou S, Deng F, Huang L, Zhang H, Wang X, Wang Y, Hou H, Liang S, Ma X, Yin L. PRKCB methylation: a potential biomarker of MDD with childhood chronic stress, a cross-sectional study in drug-naive, first-episode adolescent MDD. Epigenetics 2024; 19:2408159. [PMID: 39342638 PMCID: PMC11444515 DOI: 10.1080/15592294.2024.2408159] [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: 06/10/2024] [Revised: 08/29/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
The purpose of this study was to investigate the relationship between childhood chronic stress(CCS), Protein kinase C beta (PRKCB) methylation and adolescent major depressive disorder (MDD). After recruiting 100 adolescents with MDD and 50 healthy controls (HCs), we evaluated the severity of CCS. PRKCB methylation was assessed by pyrosequencing using whole blood-derived DNA. To explore the relationship between CCS, PRKCB and adolescent MDD, we conducted correlation analysis and regression analysis, and constructed multiplicative interaction models and generalized linear models. PRKCB methylation and CCS were both found to be associated with MDD, and CCS was associated with PRKCB methylation. No significant CCS-PRKCB methylation interactions were observed. However, we found the interaction of CCS and MDD on PRKCB methylation. Our results found that PRKCB methylation was influenced by CCS and the disease itself, and PRKCB methylation was significantly positively associated with MDD severity, suggesting that PRKCB methylation may be a potential biomarker for adolescent MDD. This study is a cross-sectional observational study, which cannot draw the conclusion of causality. Prospective cohort studies are needed to further examine the relationship between CCS, adolescent MDD, and PRKCB methylation.
Collapse
Affiliation(s)
- Yuanmei Tao
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
| | - Meijiang Jin
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
| | - Hang Zhang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
| | - Maojia Ran
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
| | - Hanmei Xu
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Shoukang Zou
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Fang Deng
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Lijuan Huang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaolan Wang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yanping Wang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Huijin Hou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohong Ma
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
| | - Li Yin
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Department of Psychiatry, Sichuan Clinical Medical Research Center for Mental Disorder, Chengdu, Sichuan, China
- Institute for Systematic Genetics, Frontiers Science Center for Disease-Related Molecular Network, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Göver T, Slezak M. Targeting glucocorticoid receptor signaling pathway for treatment of stress-related brain disorders. Pharmacol Rep 2024; 76:1333-1345. [PMID: 39361217 PMCID: PMC11582215 DOI: 10.1007/s43440-024-00654-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 11/22/2024]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis plays a central role in governing stress-related disorders such as major depressive disorder (MDD), anxiety, and post-traumatic stress disorder. Chronic stress or early life trauma, known risk factors of disease, alter HPA axis activity and pattern of glucocorticoid (GC) secretion. These changes have consequences for physiological processes controlled by glucocorticoid receptor (GR) signaling, such as immune response and metabolism. In the brain, the aberrant GR signaling translates to altered behavior, making the GR pathway a viable target for therapies of stress-related disorders. One of the crucial elements of the pathway is FKBP5, a regulator of GR sensitivity and feedback control within the HPA axis, in which genetic variants were shown to moderate the risk of developing psychiatric conditions. The difficulty in targeting the GR-FKBP5 pathway stems from tailoring the intervention to specific brain regions and cell types, in the context of personalized genetic variations in GR and GR-associated genes, like FKBP5. The development of selective inhibitors, antagonists, and approaches based on targeted protein degradation offer insights into mechanistic aspects of disease and pave the way for improved therapy. These strategies can be employed either independently or in conjunction with conventional medications. Concomitant advancements in personalized drug screening (e.g. in vitro models exploiting induced pluripotent stem cells, iPSCs) bring the potential for optimization of therapy aiming to rescue central deficits originating from the HPA imbalance. In this mini-review, we discuss potential therapeutic strategies targeting GR signaling in stress-related disorders, with a focus on personalized approaches and advancements in drug development.
Collapse
Affiliation(s)
- Tansu Göver
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, ul. Stabłowicka 147, 54-066, Wroclaw, Poland
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chałubińskiego 3A, 50-368, Wroclaw, Poland
| | - Michal Slezak
- Lukasiewicz Research Network - PORT Polish Center for Technology Development, ul. Stabłowicka 147, 54-066, Wroclaw, Poland.
| |
Collapse
|
34
|
Zgajnar N, Lagadari M, Gallo LI, Piwien-Pilipuk G, Galigniana MD. Mitochondrial-nuclear communication by FKBP51 shuttling. J Cell Biochem 2024; 125:e30386. [PMID: 36815347 DOI: 10.1002/jcb.30386] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/24/2023] [Accepted: 02/03/2023] [Indexed: 02/24/2023]
Abstract
The HSP90-binding immunophilin FKBP51 is a soluble protein that shows high homology and structural similarity with FKBP52. Both immunophilins are functionally divergent and often show antagonistic actions. They were first described in steroid receptor complexes, their exchange in the complex being the earliest known event in steroid receptor activation upon ligand binding. In addition to steroid-related events, several pleiotropic actions of FKBP51 have emerged during the last years, ranging from cell differentiation and apoptosis to metabolic and psychiatric disorders. On the other hand, mitochondria play vital cellular roles in maintaining energy homeostasis, responding to stress conditions, and affecting cell cycle regulation, calcium signaling, redox homeostasis, and so forth. This is achieved by proteins that are encoded in both the nuclear genome and mitochondrial genes. This implies active nuclear-mitochondrial communication to maintain cell homeostasis. Such communication involves factors that regulate nuclear and mitochondrial gene expression affecting the synthesis and recruitment of mitochondrial and nonmitochondrial proteins, and/or changes in the functional state of the mitochondria itself, which enable mitochondria to recover from stress. FKBP51 has emerged as a serious candidate to participate in these regulatory roles since it has been unexpectedly found in mitochondria showing antiapoptotic effects. Such localization involves the tetratricopeptide repeats domains of the immunophilin and not its intrinsic enzymatic activity of peptidylprolyl-isomerase. Importantly, FKBP51 abandons the mitochondria and accumulates in the nucleus upon cell differentiation or during the onset of stress. Nuclear FKBP51 enhances the enzymatic activity of telomerase. The mitochondrial-nuclear trafficking is reversible, and certain situations such as viral infections promote the opposite trafficking, that is, FKBP51 abandons the nucleus and accumulates in mitochondria. In this article, we review the latest findings related to the mitochondrial-nuclear communication mediated by FKBP51 and speculate about the possible implications of this phenomenon.
Collapse
Affiliation(s)
- Nadia Zgajnar
- Instituto de Biología y Medicina Experimental (IBYME)/CONICET, Buenos Aires, Argentina
| | - Mariana Lagadari
- Instituto de Ciencia y Tecnología de Alimentos de Entre Ríos, Concordia, Argentina
| | - Luciana I Gallo
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFYBYNE)/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Mario D Galigniana
- Instituto de Biología y Medicina Experimental (IBYME)/CONICET, Buenos Aires, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
35
|
Kremer TL, Chen J, Buhl A, Berhe O, Bilek E, Geiger LS, Ma R, Moessnang C, Reichert M, Reinhard I, Schwarz K, Schweiger JI, Streit F, Witt SH, Zang Z, Zhang X, Nöthen MM, Rietschel M, Ebner-Priemer UW, Schwarz E, Meyer-Lindenberg A, Braun U, Tost H. Multimodal Associations of FKBP5 Methylation With Emotion-Regulatory Brain Circuits. Biol Psychiatry 2024; 96:858-867. [PMID: 38460581 DOI: 10.1016/j.biopsych.2024.03.003] [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: 08/16/2023] [Revised: 02/02/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Understanding the biological processes that underlie individual differences in emotion regulation and stress responsivity is a key challenge for translational neuroscience. The gene FKBP5 is a core regulator in molecular stress signaling that is implicated in the development of psychiatric disorders. However, it remains unclear how FKBP5 DNA methylation in peripheral blood is related to individual differences in measures of neural structure and function and their relevance to daily-life stress responsivity. METHODS Here, we characterized multimodal correlates of FKBP5 DNA methylation by combining epigenetic data with neuroimaging and ambulatory assessment in a sample of 395 healthy individuals. RESULTS First, we showed that FKBP5 demethylation as a psychiatric risk factor was related to an anxiety-associated reduction of gray matter volume in the ventromedial prefrontal cortex, a brain area that is involved in emotion regulation and mental health risk and resilience. This effect of epigenetic upregulation of FKBP5 on neuronal structure is more pronounced where FKBP5 is epigenetically downregulated at baseline. Leveraging 208 functional magnetic resonance imaging scans during a well-established emotion-processing task, we found that FKBP5 DNA methylation in peripheral blood was associated with functional differences in prefrontal-limbic circuits that modulate affective responsivity to daily stressors, which we measured using ecological momentary assessment in daily life. CONCLUSIONS Overall, we demonstrated how FKBP5 contributes to interindividual differences in neural and real-life affect regulation via structural and functional changes in prefrontal-limbic brain circuits.
Collapse
Affiliation(s)
- Thomas L Kremer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Anais Buhl
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Oksana Berhe
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Edda Bilek
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Lena S Geiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ren Ma
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolin Moessnang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus Reichert
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany; Department of eHealth and Sports Analytics, Ruhr University Bochum, Bochum, Germany
| | - Iris Reinhard
- Department of Biostatistics, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kristina Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Janina I Schweiger
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Fabian Streit
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Zhenxiang Zang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Xiaolong Zhang
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Marcella Rietschel
- DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ulrich W Ebner-Priemer
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Mental mHealth Lab, Chair of Applied Psychology, Institute of Sports and Sports Science, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany
| | - Urs Braun
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany; Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Heike Tost
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DZPG (German Center for Mental Health), partner site Mannheim/Heidelberg/Ulm, Germany.
| |
Collapse
|
36
|
Jin Z, Xing Y, Duan P, Bi Y, Li X, Feng W, Zhang B. Revealing the molecular links between coronary heart disease and cognitive impairment: the role of aging-related genes and therapeutic potential of stellate ganglion block. Biogerontology 2024; 26:16. [PMID: 39609308 PMCID: PMC11604741 DOI: 10.1007/s10522-024-10159-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] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Coronary heart disease (CHD) and cognitive impairment frequently co-occur in aging populations, yet the molecular mechanisms linking these conditions remain unclear. This study aims to elucidate the roles of key aging-related genes (ARGs), specifically FKBP5 and DDIT3, in the pathophysiology of CHD and cognitive impairment, and to evaluate the therapeutic potential of stellate ganglion block (SGB). Using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing (bulk RNA-seq) data, we identified FKBP5 and DDIT3 as pivotal genes upregulated in both conditions. Experimental findings show that SGB effectively modulates these ARG-related pathways through autonomic regulation, specifically suppressing estrogen and NF-κB signaling pathways, thereby reducing the expression of pro-inflammatory cytokines such as SRC, MMP2, FKBP5, IRAK1, and MYD88, while upregulating the vasodilation-related gene NOS3. This modulation improved endothelial and cardiac function and enhanced cerebral blood flow (CBF), leading to cognitive improvement. Behavioral assessments, including novel object recognition (NOR) and Morris water maze (MWM) tests, demonstrated that SGB-treated rats outperformed untreated MI rats, with significant cognitive recovery over time. Further support from laser Doppler flowmetry (LDF) and electroencephalogram (EEG) analyses revealed increased left frontal blood flow and stabilized neural activity, indicating a favorable neurophysiological environment for cognitive rehabilitation. Our findings suggest that left stellate ganglion block (LSGB) provides both cardiac and cognitive benefits through targeted gene modulation, establishing its therapeutic potential for addressing the intersecting pathologies of CHD and cognitive impairment.
Collapse
Affiliation(s)
- Zhehao Jin
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuling Xing
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Pengyu Duan
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China
| | - Yonghong Bi
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Xiaoyan Li
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Weiyu Feng
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China
| | - Bing Zhang
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China.
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
37
|
Clarke SA, Eng PC, Comninos AN, Lazarus K, Choudhury S, Tsang C, Meeran K, Tan TM, Dhillo WS, Abbara A. Current Challenges and Future Directions in the Assessment of Glucocorticoid Status. Endocr Rev 2024; 45:795-817. [PMID: 38795365 PMCID: PMC11581704 DOI: 10.1210/endrev/bnae016] [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: 08/04/2023] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 05/27/2024]
Abstract
Glucocorticoid (GC) hormones are secreted in a circadian and ultradian rhythm and play a critical role in maintaining physiological homeostasis, with both excess and insufficient GC associated with adverse effects on health. Current assessment of GC status is primarily clinical, often in conjunction with serum cortisol values, which may be stimulated or suppressed depending on the GC disturbance being assessed. In the setting of extreme perturbations in cortisol levels ie, markedly low or high levels, symptoms and signs of GC dysfunction may be overt. However, when disturbances in cortisol GC status values are less extreme, such as when assessing optimization of a GC replacement regimen, signs and symptoms can be more subtle or nonspecific. Current tools for assessing GC status are best suited to identifying profound disturbances but may lack sensitivity for confirming optimal GC status. Moreover, single cortisol values do not necessarily reflect an individual's GC status, as they are subject to inter- and intraindividual variation and do not take into account the pulsatile nature of cortisol secretion, variation in binding proteins, or local tissue concentrations as dictated by 11beta-hydroxysteroid dehydrogenase activity, as well as GC receptor sensitivity. In the present review, we evaluate possible alternative methods for the assessment of GC status that do not solely rely on the measurement of circulating cortisol levels. We discuss the potential of changes in metabolomic profiles, micro RNA, gene expression, and epigenetic and other novel biomarkers such as growth differentiating factor 15 and osteocalcin, which could in the future aid in the objective classification of GC status.
Collapse
Affiliation(s)
- Sophie A Clarke
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Pei Chia Eng
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
- Department of Endocrinology, National University of Singapore, Singapore
| | - Alexander N Comninos
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Katharine Lazarus
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Sirazum Choudhury
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Christie Tsang
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
| | - Karim Meeran
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Tricia M Tan
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Waljit S Dhillo
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| | - Ali Abbara
- Section of Investigative Medicine, Imperial College London, London W12 ONN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London W6 8RF, UK
| |
Collapse
|
38
|
Lee A, Thuras P, Baller J, Jiao C, Guo B, Erbes CR, Polusny MA, Liu C, Wu B, Lim KO, Bishop JR. Serotonin Transporter (SLC6A4) and FK506-Binding Protein 5 (FKBP5) Genotype and Methylation Relationships with Response to Meditation in Veterans with PTSD. Mol Neurobiol 2024; 61:9608-9622. [PMID: 38671329 DOI: 10.1007/s12035-024-04096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/04/2024] [Indexed: 04/28/2024]
Abstract
Meditation-based interventions are novel and effective non-pharmacologic treatments for veterans with PTSD. We examined relationships between treatment response, early life trauma exposure, DNA polymorphisms, and methylation in the serotonin transporter (SLC6A4) and FK506-binding protein 5 (FKBP5) genes. DNA samples and clinical outcomes were examined in 72 veterans with PTSD who received meditation-based therapy in two separate studies of mindfulness-based stress reduction (MBSR) and Transcendental Meditation (TM). The PTSD Checklist was administered to assess symptoms at baseline and after 9 weeks of meditation intervention. We examined the SLC6A4 promoter (5HTTLPR_L/S insertion/deletion + rs25531_A/G) polymorphisms according to previously defined gene expression groups, and the FKBP5 variant rs1360780 previously associated with PTSD disease risk. Methylation for CpG sites of SLC6A4 (28 sites) and FKBP5 (45 sites) genes was quantified in DNA samples collected before and after treatment. The 5HTTLPR LALA high expression genotype was associated with greater symptom improvement in participants exposed to early life trauma (p = 0.015). Separately, pre to post-treatment change of DNA methylation in a group of nine FKBP5 CpG sites was associated with greater symptom improvement (OR = 2.8, 95% CI 1.1-7.1, p = 0.027). These findings build on a wealth of existing knowledge regarding epigenetic and genetic relationships with PTSD disease risk to highlight the potential importance of SLC6A4 and FKBP5 for treatment mechanisms and as biomarkers of symptom improvement.
Collapse
Affiliation(s)
- Adam Lee
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Room 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN, 55455, USA
| | - Paul Thuras
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Joshua Baller
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Chuan Jiao
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Krebs, Université Paris Cité, 75014, Paris, France
| | - Bin Guo
- Division of Biostatistics, University of Minnesota, Minneapolis, MN, USA
| | - Christopher R Erbes
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Center for Care Delivery and Outcomes Research, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Melissa A Polusny
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Center for Care Delivery and Outcomes Research, Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
| | - Chunyu Liu
- Department of Psychiatry, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Baolin Wu
- Department of Epidemiology and Biostatistics, Program in Public Health, University of California-Irvine, Irvine, CA, USA
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Geriatric Research, Education, and Clinical Center, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Room 7-115 Weaver-Densford Hall, 308 Harvard St SE, Minneapolis, MN, 55455, USA.
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
39
|
Karlbauer VN, Martins J, Rex-Haffner M, Sauer S, Roeh S, Dittrich K, Doerr P, Klawitter H, Entringer S, Buss C, Winter SM, Heim C, Czamara D, Binder EB. Prenatal exposures and cell type proportions are main drivers of FKBP5 DNA methylation in maltreated and non-maltreated children. Neurobiol Stress 2024; 33:100687. [PMID: 39640002 PMCID: PMC11617920 DOI: 10.1016/j.ynstr.2024.100687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
DNA methylation in peripheral tissues may be a relevant biomarker of risk for developing mental disorders after exposure to early life adversity. Genes involved in HPA axis regulation, such as FKBP5, might play a key role. In this study, we aimed to identify the main drivers of salivary FKBP5 methylation in a cohort of 162 maltreated and non-maltreated children aged 3-5 years at two measurement timepoints. We combined data from a targeted bisulfite sequencing approach for fine-mapping 49 CpGs in regulatory regions of FKBP5 and epigenetic scores for exposure to alcohol, cigarette smoke, and glucocorticoids derived from the EPICv1 microarray. Most variability of methylation in the FKBP5 locus was explained by estimated cell type proportions as well as epigenetic exposure scores, most prominently by the glucocorticoid exposure score. While not surviving correction for multiple testing, we replicated previously reported associations of FKBP5 methylation with CM. We also detected synergistic effects of both rs1360780 genotype and the glucocorticoid exposure score on FKBP5 hypomethylation. These effects were identified in the 3'TAD, a distal regulatory region of FKBP5 which is not extensively covered in Illumina arrays, emphasizing the need for fine mapping approaches. Additionally, the epigenetic glucocorticoid exposure score was associated with childhood maltreatment, maternal mental disorders, and pregnancy complications, thereby highlighting the role of glucocorticoid signaling in the epigenetic consequences of early adversity. These results underscore the need to assess cell type heterogeneity in targeted assessments of DNA methylation and show the impact of exposures beyond just childhood maltreatment such as glucocorticoid exposure.
Collapse
Affiliation(s)
- Vera N. Karlbauer
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität Munich, Germany
| | - Jade Martins
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Monika Rex-Haffner
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Susann Sauer
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Simone Roeh
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Katja Dittrich
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Child and Adolescent Psychiatry, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Peggy Doerr
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Child and Adolescent Psychiatry, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Heiko Klawitter
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Campus Charité Mitte, Luisenstraße 57, 10117 Berlin, Germany
| | - Sonja Entringer
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Campus Charité Mitte, Luisenstraße 57, 10117 Berlin, Germany
- Deutsches Zentrum für Psychische Gesundheit (DZPG), LMU Klinikum. Klinik für Psychiatrie und Psychotherapie, Nußbaumstr, 80336 München & Virchowweg 23, 10117 Berlin, Germany
- Development, Health and Disease Research Program, Department of Pediatrics, University of California Irvine, Irvine, USA
| | - Claudia Buss
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Campus Charité Mitte, Luisenstraße 57, 10117 Berlin, Germany
- Deutsches Zentrum für Psychische Gesundheit (DZPG), LMU Klinikum. Klinik für Psychiatrie und Psychotherapie, Nußbaumstr, 80336 München & Virchowweg 23, 10117 Berlin, Germany
- Development, Health and Disease Research Program, Department of Pediatrics, University of California Irvine, Irvine, USA
| | - Sibylle M. Winter
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Department of Child and Adolescent Psychiatry, Augustenburger Platz 1, 13353 Berlin, Germany
- Deutsches Zentrum für Psychische Gesundheit (DZPG), LMU Klinikum. Klinik für Psychiatrie und Psychotherapie, Nußbaumstr, 80336 München & Virchowweg 23, 10117 Berlin, Germany
| | - Christine Heim
- Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Medical Psychology, Campus Charité Mitte, Luisenstraße 57, 10117 Berlin, Germany
- Deutsches Zentrum für Psychische Gesundheit (DZPG), LMU Klinikum. Klinik für Psychiatrie und Psychotherapie, Nußbaumstr, 80336 München & Virchowweg 23, 10117 Berlin, Germany
- Cluster of Excellence NeuroCure (EXC25), Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30329, USA
| | - Darina Czamara
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
| | - Elisabeth B. Binder
- Dept. Genes and Environment, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany
- Deutsches Zentrum für Psychische Gesundheit (DZPG), LMU Klinikum. Klinik für Psychiatrie und Psychotherapie, Nußbaumstr, 80336 München & Virchowweg 23, 10117 Berlin, Germany
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30329, USA
| |
Collapse
|
40
|
Núñez-Ríos DL, Nagamatsu ST, Martínez-Magaña JJ, Hurd Y, Rompala G, Krystal JH, Montalvo-Ortiz JL. Mapping the epigenomic landscape of post-traumatic stress disorder in human cortical neurons. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315258. [PMID: 39484232 PMCID: PMC11527063 DOI: 10.1101/2024.10.11.24315258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The study conducted a comprehensive genome-wide analysis of differential 5mC and 5hmC modifications at both CpG and non-CpG sites in postmortem orbitofrontal neurons from 25 PTSD cases and 13 healthy controls. It was observed that PTSD patients exhibit a greater number of differential 5hmC sites compared to 5mC sites. Specifically, individuals with PTSD tend to show hyper-5mC/5hmC at CpG sites, particularly within CpG islands and promoter regions, and hypo-5mC/5hmC at non-CpG sites, especially within intragenic regions. Functional enrichment analysis indicated distinct yet interconnected roles for 5mC and 5hmC in PTSD. The 5mC marks primarily regulate cell-cell adhesion processes, whereas 5hmC marks are involved in embryonic morphogenesis and cell fate commitment. By integrating published PTSD findings from central and peripheral tissues through multi-omics approaches, several biological mechanisms were prioritized, including developmental processes, HPA axis regulation, and immune responses. Based on the consistent enrichment in developmental processes, we hypothesize that if epigenetic changes occur during early developmental stages, they may increase the risk of developing PTSD following trauma exposure. Conversely, if these epigenetic changes occur in adulthood, they may influence neuronal apoptosis and survival mechanisms.
Collapse
Affiliation(s)
- Diana L. Núñez-Ríos
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Jose Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | - Yasmin Hurd
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Gregory Rompala
- Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - John H. Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| | | | - Janitza L. Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- National Center of Post-Traumatic Stress Disorder, VA CT Healthcare System, West Haven, CT, USA
| |
Collapse
|
41
|
Kodila ZN, Shultz SR, Yamakawa GR, Mychasiuk R. Critical Windows: Exploring the Association Between Perinatal Trauma, Epigenetics, and Chronic Pain. Neuroscientist 2024; 30:574-596. [PMID: 37212380 PMCID: PMC11439237 DOI: 10.1177/10738584231176233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Chronic pain is highly prevalent and burdensome, affecting millions of people worldwide. Although it emerges at any point in life, it often manifests in adolescence. Given that adolescence is a unique developmental period, additional strains associated with persistent and often idiopathic pain lead to significant long-term consequences. While there is no singular cause for the chronification of pain, epigenetic modifications that lead to neural reorganization may underpin central sensitization and subsequent manifestation of pain hypersensitivity. Epigenetic processes are particularly active during the prenatal and early postnatal years. We demonstrate how exposure to various traumas, such as intimate partner violence while in utero or adverse childhood experiences, can significantly influence epigenetic regulation within the brain and in turn modify pain-related processes. We provide compelling evidence that the burden of chronic pain is likely initiated early in life, often being transmitted from mother to offspring. We also highlight two promising prophylactic strategies, oxytocin administration and probiotic use, that have the potential to attenuate the epigenetic consequences of early adversity. Overall, we advance understanding of the causal relationship between trauma and adolescent chronic pain by highlighting epigenetic mechanisms that underlie this transmission of risk, ultimately informing how to prevent this rising epidemic.
Collapse
Affiliation(s)
- Zoe N. Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
- Health Sciences, Vancouver Island University, Nanaimo, Canada
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
42
|
Dahrendorff J, Currier G, Uddin M. Leveraging DNA methylation to predict treatment response in major depressive disorder: A critical review. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32985. [PMID: 38650309 DOI: 10.1002/ajmg.b.32985] [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: 09/26/2023] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024]
Abstract
Major depressive disorder (MDD) is a debilitating and prevalent mental disorder with a high disease burden. Despite a wide array of different treatment options, many patients do not respond to initial treatment attempts. Selection of the most appropriate treatment remains a significant clinical challenge in psychiatry, highlighting the need for the development of biomarkers with predictive utility. Recently, the epigenetic modification DNA methylation (DNAm) has emerged to be of great interest as a potential predictor of MDD treatment outcomes. Here, we review efforts to date that seek to identify DNAm signatures associated with treatment response in individuals with MDD. Searches were conducted in the databases PubMed, Scopus, and Web of Science with the concepts and keywords MDD, DNAm, antidepressants, psychotherapy, cognitive behavior therapy, electroconvulsive therapy, transcranial magnetic stimulation, and brain stimulation therapies. We identified 32 studies implicating DNAm patterns associated with MDD treatment outcomes. The majority of studies (N = 25) are focused on selected target genes exploring treatment outcomes in pharmacological treatments (N = 22) with a few studies assessing treatment response to electroconvulsive therapy (N = 3). Additionally, there are few genome-scale efforts (N = 7) to characterize DNAm patterns associated with treatment outcomes. There is a relative dearth of studies investigating DNAm patterns in relation to psychotherapy, electroconvulsive therapy, or transcranial magnetic stimulation; importantly, most existing studies have limited sample sizes. Given the heterogeneity in both methods and results of studies to date, there is a need for additional studies before existing findings can inform clinical decisions.
Collapse
Affiliation(s)
- Jan Dahrendorff
- Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Glenn Currier
- Department of Psychiatry and Behavioral Neurosciences, University of South Florida, Tampa, Florida, USA
| | - Monica Uddin
- Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
43
|
Otten J, Dan S, Rostin L, Profetto AE, Lardenoije R, Klengel T. Spatial transcriptomics reveals modulation of transcriptional networks across brain regions after auditory threat conditioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614979. [PMID: 39386587 PMCID: PMC11463379 DOI: 10.1101/2024.09.25.614979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Prior research has demonstrated genome-wide transcriptional changes related to fear and anxiety across species, often focusing on individual brain regions or cell types. However, the extent of gene expression differences across brain regions and how these changes interact at the level of transcriptional connectivity remains unclear. To address this, we performed spatial transcriptomics RNAseq analyses in an auditory threat conditioning paradigm in mice. We generated a spatial transcriptomic atlas of a coronal mouse brain section covering cortical and subcortical regions, corresponding to histologically defined regions. Our finding revealed widespread transcriptional responses across all brain regions examined, particularly in the medial and lateral habenula, and the choroid plexus. Network analyses highlighted altered transcriptional connectivity between cortical and subcortical regions, emphasizing the role of steroidogenic factor 1. These results provide new insights into the transcriptional networks involved in auditory threat conditioning, enhancing our understanding of molecular and neural mechanisms underlying fear and anxiety disorders.
Collapse
|
44
|
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
|
45
|
Castro RTA, Gardini E, Iliadis SI, Ehlert U, Kallak TK, Skalkidou A. Personality vulnerability to depression, resilience, and depressive symptoms: epigenetic markers among perinatal women. Ups J Med Sci 2024; 129:10603. [PMID: 39257475 PMCID: PMC11385460 DOI: 10.48101/ujms.v129.10603] [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: 02/16/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 09/12/2024] Open
Abstract
Background We examined differences in DNA methylation patterns in the NR3C1 and FKBP5 genes in relation to personality vulnerability to depression, resilience, and perinatal depressive symptoms, whilst also considering possible moderating effects of childhood traumatic events. Methods N = 160 perinatal women were assessed at late pregnancy and 1 year postpartum for personality vulnerability to depression, resilience, depressive symptoms, and childhood traumatic events with self-reported questionnaires. NR3C1 and FKBP5 methylation markers were analyzed via sodium bisulfite sequencing. Associations of methylation markers with the above mentioned variables were tested using multivariable regressions. Results NR3C1 methylation at CpGs 1, 4 and average methylation sites were negatively associated with resilience; NR3C1 methylation at CpG 2 was positively associated with postpartum depressive symptoms; methylation at CpG 4 was positively associated with prenatal depressive symptoms. The interaction between current distress due to interpersonal traumatic events and NR3C1 CpG sites in relation to personality vulnerability was significant on CpG sites 3 and 4, whereas the interaction between current distress due to total traumatic events and NR3C1 in relation to personality vulnerability was significant on CpG site 2. FKBP5 showed no significant associations with the outcomes. Conclusions This study identified associations between NR3C1 methylation and resilience as well as perinatal depressive symptoms. Interestingly, an interaction between early trauma and personality vulnerability was noted. Our findings on these specific DNA methylation markers may, if replicated and integrated into risk prediction models, contribute to early diagnosis of mothers at risk, targeted health promotion, and early interventions.
Collapse
Affiliation(s)
- Rita T Amiel Castro
- Department of Clinical Psychology and Psychotherapy, University of Zurich, Institute of Psychology, Zurich, Switzerland
| | - Elena Gardini
- Department of Clinical Psychology and Psychotherapy, University of Zurich, Institute of Psychology, Zurich, Switzerland
| | - Stavros I Iliadis
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Ulrike Ehlert
- Department of Clinical Psychology and Psychotherapy, University of Zurich, Institute of Psychology, Zurich, Switzerland
| | | | - Alkistis Skalkidou
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| |
Collapse
|
46
|
Collins JM, Keane JM, Deady C, Khashan AS, McCarthy FP, O'Keeffe GW, Clarke G, Cryan JF, Caputi V, O'Mahony SM. Prenatal stress impacts foetal neurodevelopment: Temporal windows of gestational vulnerability. Neurosci Biobehav Rev 2024; 164:105793. [PMID: 38971516 DOI: 10.1016/j.neubiorev.2024.105793] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Prenatal maternal stressors ranging in severity from everyday occurrences/hassles to the experience of traumatic events negatively impact neurodevelopment, increasing the risk for the onset of psychopathology in the offspring. Notably, the timing of prenatal stress exposure plays a critical role in determining the nature and severity of subsequent neurodevelopmental outcomes. In this review, we evaluate the empirical evidence regarding temporal windows of heightened vulnerability to prenatal stress with respect to motor, cognitive, language, and behavioural development in both human and animal studies. We also explore potential temporal windows whereby several mechanisms may mediate prenatal stress-induced neurodevelopmental effects, namely, excessive hypothalamic-pituitary-adrenal axis activity, altered serotonin signalling and sympathetic-adrenal-medullary system, changes in placental function, immune system dysregulation, and alterations of the gut microbiota. While broadly defined developmental windows are apparent for specific psychopathological outcomes, inconsistencies arise when more complex cognitive and behavioural outcomes are considered. Novel approaches to track molecular markers reflective of the underlying aetiologies throughout gestation to identify tractable biomolecular signatures corresponding to critical vulnerability periods are urgently required.
Collapse
Affiliation(s)
- James M Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - James M Keane
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Clara Deady
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Fergus P McCarthy
- The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Obstetrics and Gynaecology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland.
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; The Irish Centre for Maternal and Child Health Research (INFANT), Cork University Maternity Hospital, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | - Valentina Caputi
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| | | |
Collapse
|
47
|
Creasey N, Leijten P, Tollenaar MS, Boks MP, Overbeek G. DNA methylation variation after a parenting program for child conduct problems: Findings from a randomized controlled trial. Child Dev 2024; 95:1462-1477. [PMID: 38436454 DOI: 10.1111/cdev.14090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
This study investigated associations of the Incredible Years (IY) parenting program with children's DNA methylation. Participants were 289 Dutch children aged 3-9 years (75% European ancestry, 48% female) with above-average conduct problems. Saliva was collected 2.5 years after families were randomized to IY or care as usual (CAU). Using an intention-to-treat approach, confirmatory multiple-regression analyses revealed no significant differences between the IY and CAU groups in children's methylation levels at the NR3C1 and FKBP5 genes. However, exploratory epigenome-wide analyses revealed nine differentially methylated regions between groups, coinciding with SLAMF1, MITF, FAM200B, PSD3, SNX31, and CELSR1. The study provides preliminary evidence for associations of IY with children's salivary methylation levels and highlights the need for further research into biological outcomes of parenting programs.
Collapse
Affiliation(s)
- Nicole Creasey
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Patty Leijten
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke S Tollenaar
- Institute of Psychology & Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | - Marco P Boks
- Institute of Psychology & Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
- Department of Psychiatry, Brain Center University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Geertjan Overbeek
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
48
|
Debs SR, Rothmond DA, Zhu Y, Weickert CS, Purves-Tyson TD. Molecular evidence of altered stress responsivity related to neuroinflammation in the schizophrenia midbrain. J Psychiatr Res 2024; 177:118-128. [PMID: 39004003 DOI: 10.1016/j.jpsychires.2024.07.004] [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: 12/04/2023] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Stress and inflammation are risk factors for schizophrenia. Chronic psychosocial stress is associated with subcortical hyperdopaminergia, a core feature of schizophrenia. Hyperdopaminergia arises from midbrain neurons, leading us to hypothesise that changes in stress response pathways may occur in this region. To identify whether transcriptional changes in glucocorticoid and mineralocorticoid receptors (NR3C1/GR, NR3C2/MR) or other stress signalling molecules (FKBP4, FKBP5) exist in schizophrenia midbrain, we measured gene expression in the human brain (N = 56) using qRT-PCR. We assessed whether alterations in these mRNAs were related to previously identified high/low inflammatory status. We investigated relationships between stress-related transcripts themselves, and between FKBP5 mRNA, dopaminergic, and glial cell transcripts in diagnostic and inflammatory subgroups. Though unchanged by diagnosis, GR mRNA levels were reduced in high inflammatory compared to low inflammatory schizophrenia cases (p = 0.026). We found no effect of diagnosis or inflammation on MR mRNA. FKBP4 mRNA was decreased and FKBP5 mRNA was increased in schizophrenia (p < 0.05). FKBP5 changes occurred in high inflammatory (p < 0.001), whereas FKBP4 changes occurred in low inflammatory schizophrenia cases (p < 0.05). The decrease in mRNA encoding the main stress receptor (GR), as well as increased transcript levels of the stress-responsive negative regulator (FKBP5), may combine to blunt the midbrain response to stress in schizophrenia when neuroinflammation is present. Negative correlations between FKBP5 mRNA and dopaminergic transcripts in the low inflammatory subgroup suggest higher levels of FKBP5 mRNA may also attenuate dopaminergic neurotransmission in schizophrenia even when inflammation is absent. We report alterations in GR-mediated stress signalling in the midbrain in schizophrenia.
Collapse
Affiliation(s)
- Sophie R Debs
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Debora A Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia
| | - Yunting Zhu
- Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Tertia D Purves-Tyson
- Preclinical Neuropsychiatry Laboratory, Neuroscience Research Australia, Randwick, New South Wales, 2031, Australia; Discipline of Psychiatry & Mental Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, 2052, Australia.
| |
Collapse
|
49
|
Nikolić T, Bogosavljević MV, Stojković T, Kanazir S, Lončarević-Vasiljković N, Radonjić NV, Popić J, Petronijević N. Effects of Antipsychotics on the Hypothalamus-Pituitary-Adrenal Axis in a Phencyclidine Animal Model of Schizophrenia. Cells 2024; 13:1425. [PMID: 39272997 PMCID: PMC11394463 DOI: 10.3390/cells13171425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Schizophrenia (SCH) is a mental disorder that requires long-term antipsychotic treatment. SCH patients are thought to have an increased sensitivity to stress. The dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, observed in SCH, could include altered levels of glucocorticoids, glucocorticoid receptors (GRs), and associated proteins. The perinatal administration of phencyclidine (PCP) to rodents represents an animal model of SCH. This study investigated the effects of perinatal PCP exposure and subsequent haloperidol/clozapine treatment on corticosterone levels measured by ELISA and the expression of GR-related proteins (GR, pGR, HSP70, HSP90, FKBP51, and 11β-Hydroxysteroid dehydrogenase-11β-HSD) determined by Western blot, in different brain regions of adult rats. Six groups of male rats were treated on the 2nd, 6th, 9th, and 12th postnatal days (PN), with either PCP or saline. Subsequently, one saline and one PCP group received haloperidol/clozapine from PN day 35 to PN day 100. The results showed altered GR sensitivity in the rat brain after PCP exposure, which decreased after haloperidol/clozapine treatment. These findings highlight disturbances in the HPA axis in a PCP-induced model of SCH and the potential protective effects of antipsychotics. To the best of our knowledge, this is the first study to investigate the effects of antipsychotic drugs on the HPA axis in a PCP animal model of SCH.
Collapse
Affiliation(s)
- Tatjana Nikolić
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (T.N.); (M.V.B.)
| | - Milica Velimirović Bogosavljević
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (T.N.); (M.V.B.)
| | - Tihomir Stojković
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (T.N.); (M.V.B.)
| | - Selma Kanazir
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, 11000 Belgrade, Serbia;
| | - Nataša Lončarević-Vasiljković
- iNOVA4Health, NOVA Medical School|Faculdade Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Campo dos Mártires da Pátria, 1169-056 Lisbon, Portugal;
| | - Nevena V. Radonjić
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jelena Popić
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4, Canada;
| | - Nataša Petronijević
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (T.N.); (M.V.B.)
| |
Collapse
|
50
|
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
|