1
|
Wen S, Zeng Y, Xu Y, Xu S, Chen W, Wang G, Zhang W, Song H. Association of childhood maltreatment and adverse lifetime experiences with post-injury psychopathology: evidence from the China Severe Trauma Cohort. BMC Med 2025; 23:29. [PMID: 39838452 PMCID: PMC11753082 DOI: 10.1186/s12916-025-03861-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Adverse life experiences have been associated with increased susceptibilities to psychopathology in later life. However, their impact on psychological responses following physical trauma remains largely unexplored. METHODS Based on the China Severe Trauma Cohort, we conducted a cohort study of 2937 patients who were admitted to the Trauma Medical Center of West China Hospital between June 2020 and August 2023. Adverse life experiences, including childhood maltreatment (5 subtypes, measured by Childhood Trauma Questionnaire-Short Form) and adverse lifetime experiences (17 subtypes, by Life Events Checklist) were assessed. Generalized linear mixed models were used to examine the associations of childhood maltreatment and adverse lifetime experiences with symptoms of psychopathology measured at multiple time-points after the index injury (i.e., at recruitment, 1-, 3-, 6-, and 12-month follow-ups), adjusted for important confounders. We further stratified the analyses by level of genetic predisposition to a given psychological symptom quantified by polygenic risk score (PRS) based on publicly available GWAS summary statistics. Mediation analyses were performed to assess the role of adverse lifetime experiences in connecting childhood maltreatment and post-injury psychopathology. RESULTS The mean age of participants was 47.95 years with a predominance of males (61.39%). During the whole follow-up period, the incidence of symptoms of stress-related disorders, anxiety, and depression was 13.86%, 29.89%, and 36.57%, respectively. We observed associations between the cumulative number of those studied adversities and increased risk of post-injury psychopathology, particularly stress-related disorder (odds ratio [OR] = 2.78, 95% confidence interval [CI] 1.87-4.12 for ≥ 2 vs no childhood maltreatment; 2.65 [1.67-4.20] for ≥ 4 vs 0-1 adverse lifetime experiences). By subtype, positive associations were observed for most studied life adversities, with the most pronounced estimates for childhood emotional abuse (ORs = 1.71-2.52) and lifetime life-threatening illness/injury (ORs = 1.87-2.89). We found basically comparable estimates among traumatized individuals with different PRSs for studied psychopathology. Moreover, adverse lifetime experiences may partially (mediation proportion: 22.52-27.48%) explain the associations between various childhood maltreatment and post-injury psychopathology. CONCLUSIONS Both childhood maltreatment and adverse lifetime experiences were associated with post-injury psychopathology, irrespective of genetic susceptibility. Such findings highlight the importance of close surveillance and timely psychological interventions for injury patients with adverse life experiences.
Collapse
Affiliation(s)
- Shu Wen
- Trauma Medical Center, Department of Orthopedics Surgery and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
- Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Yu Zeng
- Med-X Center for Informatics, Sichuan University, Chengdu, China
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yueyao Xu
- Med-X Center for Informatics, Sichuan University, Chengdu, China
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shishi Xu
- Med-X Center for Informatics, Sichuan University, Chengdu, China
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Endocrinology & Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Wenwen Chen
- Med-X Center for Informatics, Sichuan University, Chengdu, China
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Guanglin Wang
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, Sichuan University, Chengdu, China.
- Department of Orthopedics, Orthopedics Research Institute, West China Hospital, Sichuan University, Chengdu, China.
| | - Wei Zhang
- Med-X Center for Informatics, Sichuan University, Chengdu, China.
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Huan Song
- Med-X Center for Informatics, Sichuan University, Chengdu, China.
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland.
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Forum DMK, Bjerregaard C, Thomsen PH. The significance of DNA methylation of the NR3C1 gene encoding the glucocorticoid receptor for developing resilience in individuals exposed to early life stress. Nord J Psychiatry 2025; 79:1-14. [PMID: 39773140 DOI: 10.1080/08039488.2024.2436987] [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: 04/08/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE To analyze and interpret why some individuals are resilient to ELS while others are susceptible, resulting in psychiatric outcome later in life, with a focus on the role of DNAm of the NR3C1 gene as a mediating mechanism between ELS and the risk of psychiatric outcomes. We hypothesized that a high level of mental resilience to ELS, expressed as lower incidence of psychiatric outcomes, was associated with attenuated NR3C1 DNAm levels. MATERIALS AND METHODS The first authors conducted a systematic search on PubMed to identify primary research studies. Abstract were screened and full-text were reviewed to assess the eligibility for inclusion. Consensus on assessment was reached after discussion of eligibility criteria. Studies were sorted based on whether they investigated the association between ELS and NR3C1 DNAm in 1) individuals exposed compared to unexposed to ELS both without a psychiatric outcome or in 2) individuals exposed to ELS with a psychiatric outcome compared to exposed individuals without a psychiatric outcome. RESULTS AND CONCLUSION Seven studies met the eligibility criteria. The results were inconsistent; two studies supported our hypothesis, two studies indicated that increased NR3C1 DNAm mediated resilience to ELS, and three studies found no association.
Collapse
Affiliation(s)
- Ditte Mathilde Klith Forum
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| | - Camilla Bjerregaard
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| | - Per Hove Thomsen
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| |
Collapse
|
3
|
Fan X, Sun L, Qin Y, Liu Y, Wu S, Du L. The Role of HSP90 Molecular Chaperones in Depression: Potential Mechanisms. Mol Neurobiol 2025; 62:708-717. [PMID: 38896156 DOI: 10.1007/s12035-024-04284-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
Major depressive disorder (MDD) is characterized by high rates of disability and death and has become a public health problem that threatens human life and health worldwide. HPA axis disorder and neuroinflammation are two common biological abnormalities in MDD patients. Hsp90 is an important molecular chaperone that is widely distributed in the organism. Hsp90 binds to the co-chaperone and goes through a molecular chaperone cycle to complete its regulation of the client protein. Numerous studies have demonstrated that Hsp90 regulates how the HPA axis reacts to stress and how GR, the HPA axis' responsive substrate, matures. In addition, Hsp90 exhibits pro-inflammatory effects that are closely related to neuroinflammation in MDD. Currently, Hsp90 inhibitors have made some progress in the treatment of a variety of human diseases, but they still need to be improved. Further insight into the role of Hsp90 in MDD provides new ideas for the development of new antidepressant drugs targeting Hsp90.
Collapse
Affiliation(s)
- Xuyuan Fan
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Lei Sun
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Ye Qin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Yuan Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Shusheng Wu
- Department of the Central Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| |
Collapse
|
4
|
Liu C, Gershon ES. Endophenotype 2.0: updated definitions and criteria for endophenotypes of psychiatric disorders, incorporating new technologies and findings. Transl Psychiatry 2024; 14:502. [PMID: 39719446 PMCID: PMC11668880 DOI: 10.1038/s41398-024-03195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Recent genetic studies have linked numerous loci to psychiatric disorders. However, the biological pathways that connect these genetic associations to psychiatric disorders' specific pathophysiological processes are largely unclear. Endophenotypes, first defined over five decades ago, are heritable traits, independent of disease state that are associated with a disease, encompassing a broad range of neurophysiological, biochemical, endocrinological, neuroanatomical, cognitive, and neuropsychological characteristics. Considering the advancements in genetics and genomics over recent decades, we propose a revised definition of endophenotypes as 'genetically influenced phenotypes linked to disease or treatment characteristics and their related events.' We also updated endophenotype criteria to include (1) reliable measurement, (2) association with the disease or its related events, and (3) genetic mediation. 'Genetic mediation' is introduced to differentiate between causality and pleiotropic effects and allows non-linear relationships. Furthermore, this updated Endophenotype 2.0 framework expands to encompass genetically regulated responses to disease-related factors, including environmental risks, illness progression, treatment responses, and resilience phenotypes, which may be state-dependent. This broadened definition paves the way for developing new endophenotypes crucial for genetic analyses in psychiatric disorders. Integrating genetics, genomics, and diverse endophenotypes into multi-dimensional mechanistic models is vital for advancing our understanding of psychiatric disorders. Crucially, elucidating the biological underpinnings of endophenotypes will enhance our grasp of psychiatric genetics, thereby improving disease risk prediction and treatment approaches.
Collapse
Affiliation(s)
- Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
- School of Life Sciences, Central South University, Changsha, China.
| | - Elliot S Gershon
- Departments of Psychiatry and Human Genetics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
5
|
Feng Y, Zhang S, Liao X, Jia Y, Yang Y, Zhang W. Association between bullying victimization and mental health problems among Chinese left-behind children: a cross-sectional study from the adolescence mental health promotion cohort. Front Psychiatry 2024; 15:1440821. [PMID: 39575193 PMCID: PMC11578976 DOI: 10.3389/fpsyt.2024.1440821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024] Open
Abstract
Background Left-behind children (LBC) refer to those who have been separated from at least one parent for six months or more due to parental migration for work. This phenomenon poses a significant threat to the mental health of over 61 million LBC in China. This study aims to compare the prevalence of mental health symptoms between LBC and non-left-behind children (non-LBC) and to explore the predictive effect of bullying victimization on adolescent mental health problems. Methods In 2019, we conducted a cross-sectional analysis involving 28,036 children and adolescents in Mianyang City, Sichuan Province, China, with ages ranging from 8 to 19 years. Mental health symptoms were assessed using the 7-item Generalized Anxiety Disorder (GAD-7) scale and the 9-item Patient Health Questionnaire (PHQ-9). The Delaware Bullying Victimization Scale-Student (DBVS-S) was employed to gather data on experiences of bullying victimization. Information on self-injury was collected by inquiring whether participants had engaged in self-injurious behavior and the reasons for such behavior. Multivariable logistic regression was utilized to analyze the risk and protective factors associated with mental health symptoms, with a particular focus on different types of bullying victimization. Results Compared to non-left-behind children (non-LBC), left-behind children (LBC) exhibited a higher prevalence of mental health issues: anxiety symptoms (24.0% vs. 18.0%, p<0.001), depressive symptoms (27.9% vs. 19.4%, p<0.001), and self-injurious behavior (17.7% vs. 12.2%, p<0.001). Among LBC, physical bullying was identified as the most significant predictor of anxiety symptoms (OR = 1.62). Additionally, LBC who experienced verbal bullying had a higher risk of depressive symptoms (OR = 2.23) and self-injurious behaviors (OR = 1.54). Enhanced family functioning, positive teacher-student relationships, and strong peer relationships were found to offer protective effects against mental health problems. Conclusion Our results suggested that LBC experienced a higher incidence of mental health symptoms, particularly among those who had been victims of bullying. This underscores the urgent need for supportive strategies focused on the school environment and interpersonal relationships to mitigate negative mental health outcomes for LBC.
Collapse
Affiliation(s)
- Yuan Feng
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Simai Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Med-X Center for Informatics, Sichuan University, Chengdu, China
| | - Xiao Liao
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuge Jia
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yang
- School of Management and the Key Laboratory of Process Optimization and Intelligent Decision-Making, Ministry of Education, Hefei University of Technology, Hefei, China
| | - Wei Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Tang L, Zhao P, Pan C, Song Y, Zheng J, Zhu R, Wang F, Tang Y. Epigenetic molecular underpinnings of brain structural-functional connectivity decoupling in patients with major depressive disorder. J Affect Disord 2024; 363:249-257. [PMID: 39029702 DOI: 10.1016/j.jad.2024.07.110] [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: 10/04/2023] [Revised: 06/24/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is progressively recognized as a stress-related disorder characterized by aberrant brain network dynamics, encompassing both structural and functional domains. Yet, the intricate interplay between these dynamic networks and their molecular underpinnings remains predominantly unexplored. METHODS Both structural and functional networks were constructed using multimodal neuroimaging data from 183 MDD patients and 300 age- and gender-matched healthy controls (HC). structural-functional connectivity (SC-FC) coupling was evaluated at both the connectome- and nodal-levels. Methylation data of five HPA axis key genes, including NR3C1, FKBP5, CRHBP, CRHR1, and CRHR2, were analyzed using Illumina Infinium Methylation EPIC BeadChip. RESULTS We observed a significant reduction in SC-FC coupling at the connectome-level in patients with MDD compared to HC. At the nodal level, we found an imbalance in SC-FC coupling, with reduced coupling in cortical regions and increased coupling in subcortical regions. Furthermore, we identified 23 differentially methylated CpG sites on the HPA axis, following adjustment for multiple comparisons and control of age, gender, and medication status. Notably, three CpG sites on NR3C1 (cg01294526, cg19457823, and cg23430507), one CpG site on FKBP5 (cg25563198), one CpG site on CRHR1 (cg26656751), and one CpG site on CRHR2 (cg18351440) exhibited significant associations with SC-FC coupling in MDD patients. CONCLUSIONS These findings provide valuable insights into the connection between micro-scale epigenetic changes in the HPA axis and SC-FC coupling at macro-scale connectomes. They unveil the mechanisms underlying increased susceptibility to MDD resulting from chronic stress and may suggest potential pharmacological targets within the HPA-axis for MDD treatment.
Collapse
Affiliation(s)
- Lili Tang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China; Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Pengfei Zhao
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China
| | - Chunyu Pan
- School of Computer Science and Engineering, Northeastern University, Shenyang, PR China
| | - Yanzhuo Song
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China
| | - Rongxin Zhu
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, PR China.
| | - Yanqing Tang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
7
|
Zhang X, Fan L, Yang L, Jin X, Liu H, Lei H, Song X, Zhang Z, Zhang F, Song J. DAPK1 mediates cognitive dysfunction and neuronal apoptosis in PSD rats through the ERK/CREB/BDNF signaling pathway. Behav Brain Res 2024; 471:115064. [PMID: 38777261 DOI: 10.1016/j.bbr.2024.115064] [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: 02/21/2024] [Revised: 04/21/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024]
Abstract
Post-stroke depression (PSD) is one of the most common mental sequelae after a stroke and can damage the brain. Although PSD has garnered increasing attention in recent years, the precise mechanism remains unclear. Studies have indicated that the expression of DAPK1 is elevated in various neurodegenerative conditions, including depression, ischemic stroke, and Alzheimer's disease. However, the specific molecular mechanism of DAPK1-mediated cognitive dysfunction and neuronal apoptosis in PSD rats is unclear. In this study, we established a rat model of PSD, and then assessed depression-like behaviors and cognitive dysfunction in rats using behavioral tests. In addition, we detected neuronal apoptosis and analyzed the expression of DAPK1 protein and proteins related to the ERK/CREB/BDNF signaling pathway. The findings revealed that MCAO combined with CUMS can induce more severe depression-like behaviors and cognitive dysfunction in rats, while overexpression of DAPK1 may hinder the downstream ERK/CREB/BDNF signaling pathways, resulting in neuronal loss and exacerbation of brain tissue damage. In this study, we will focus on DAPK1 and explore its role in PSD.
Collapse
Affiliation(s)
- Xinyue Zhang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Lifei Fan
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Lina Yang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China
| | - Xuejiao Jin
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Huanhuan Liu
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Hao Lei
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Xiaojia Song
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China
| | - Zhaohui Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Fuping Zhang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China; Henan Collaborative Innovation Center of Prevention and treatment of mental disorder, Xinxiang 453000, China; Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou 451162, China.
| | - Jinggui Song
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Provincial Psychiatric Hospital, Xinxiang 453000, China; Henan Provincial Key Laboratory of Biological Psychiatry (Xinxiang Medical College), Xinxiang 453000, China; Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou 451162, China.
| |
Collapse
|
8
|
Kumar R, Nuñez NA, Joshi N, Joseph B, Verde A, Seshadri A, Cuellar Barboza AB, Prokop LJ, Medeiros GC, Singh B. Metabolomic biomarkers for (R, S)-ketamine and (S)-ketamine in treatment-resistant depression and healthy controls: A systematic review. Bipolar Disord 2024; 26:321-330. [PMID: 38326104 DOI: 10.1111/bdi.13412] [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: 02/09/2024]
Abstract
BACKGROUND Ketamine is increasingly used for treatment-resistant depression (TRD) while its mechanism of action is still being investigated. In this systematic review, we appraise the current evidence of metabolomic biomarkers for racemic ketamine and esketamine in patients with TRD and healthy controls (HCs). METHODS A comprehensive search of several databases (Ovid MEDLINE®, Embase, and Epub Ahead of Print) was performed from each database's inception to June 29, 2022, in any language, was conducted. We included studies wherein the metabolomic biomarkers for racemic ketamine or esketamine were investigated in TRD or HCs. Our main outcomes were to examine changes in metabolites among patients treated with ketamine/esketamine and explore the association with response to ketamine/esketamine. RESULTS A total of 1859 abstracts were screened of which 11 were included for full-text review. Of these, a total of five articles were included (N = 147), including three RCTs (n = 129) and two open-label trials (n = 18). All studies used racemic ketamine; one study additionally used esketamine. The included studies evaluated patients with treatment-resistant bipolar depression (n = 22), unipolar depression (n = 91), and HCs (n = 34). The included studies reported alteration in several metabolites including acylcarnitines, lipids, kynurenine (KYN), and arginine with ketamine in TRD. Studies suggest the involvement of energy metabolism, KYN, and arginine pathways. In HCs, acetylcarnitine decreased post-infusion, whereas inconsistent findings were observed after the ketamine infusion in TRD patients. CONCLUSIONS This systematic review provides preliminary evidence that ketamine may cause changes in several important pathways involved in energy metabolism and inflammation. Larger and more rigorous studies are needed.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Nicolas A Nuñez
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Neha Joshi
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Boney Joseph
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Alessandra Verde
- Section of Pediatrics, Department of Translational Medical Science, Federico II University, Naples, Italy
- Division of Medical Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Ashok Seshadri
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Larry J Prokop
- Mayo Medical Libraries, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Gustavo C Medeiros
- Department of Psychiatry & Behavioral Sciences, John Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Balwinder Singh
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Cánepa ET, Berardino BG. Epigenetic mechanisms linking early-life adversities and mental health. Biochem J 2024; 481:615-642. [PMID: 38722301 DOI: 10.1042/bcj20230306] [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: 02/19/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/15/2024]
Abstract
Early-life adversities, whether prenatal or postnatal exposure, have been linked to adverse mental health outcomes later in life increasing the risk of several psychiatric disorders. Research on its neurobiological consequences demonstrated an association between exposure to adversities and persistent alterations in the structure, function, and connectivity of the brain. Consistent evidence supports the idea that regulation of gene expression through epigenetic mechanisms are involved in embedding the impact of early-life experiences in the genome and mediate between social environments and later behavioral phenotypes. In addition, studies from rodent models and humans suggest that these experiences and the acquired risk factors can be transmitted through epigenetic mechanisms to offspring and the following generations potentially contributing to a cycle of disease or disease risk. However, one of the important aspects of epigenetic mechanisms, unlike genetic sequences that are fixed and unchangeable, is that although the epigenetic markings are long-lasting, they are nevertheless potentially reversible. In this review, we summarize our current understanding of the epigenetic mechanisms involved in the mental health consequences derived from early-life exposure to malnutrition, maltreatment and poverty, adversities with huge and pervasive impact on mental health. We also discuss the evidence about transgenerational epigenetic inheritance in mammals and experimental data suggesting that suitable social and pharmacological interventions could reverse adverse epigenetic modifications induced by early-life negative social experiences. In this regard, these studies must be accompanied by efforts to determine the causes that promote these adversities and that result in health inequity in the population.
Collapse
Affiliation(s)
- Eduardo T Cánepa
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| | - Bruno G Berardino
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and IQUIBICEN, CONICET, Buenos Aires, Argentina
| |
Collapse
|
10
|
Zhang Y, Yue W, Li J. The association of FKBP5 gene polymorphism with genetic susceptibility to depression and response to antidepressant treatment- a systematic review. BMC Psychiatry 2024; 24:274. [PMID: 38609904 PMCID: PMC11010372 DOI: 10.1186/s12888-024-05717-z] [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: 08/02/2023] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Given the inconsistencies in current studies regarding the impact of FKBP5 gene polymorphisms on depression, arising from variations in study methods, subjects, and treatment strategies, this paper provides a comprehensive review of the relationship between FKBP5 gene polymorphisms and genetic susceptibility to depression, as well as their influence on response to antidepressant treatment. METHODS Electronic databases were searched up to April 11, 2023, for all literature in English and Chinese on depression, FKBP5 gene polymorphisms, and antidepressant treatment. Data extraction and quality assessment were performed for key study characteristics. Qualitative methods were used to synthesize the study results. RESULTS A total of 21 studies were included, with the majority exhibiting average to moderate quality. Six SNPs (rs3800373, rs1360780, rs9470080, rs4713916, rs9296158, rs9394309) were broadly implicated in susceptibility to depression, while rs1360780 and rs3800373 were linked to antidepressant treatment sensitivity. Additionally, rs1360780 was associated with adverse reactions to antidepressant drug treatment. However, these associations were largely unconfirmed in replication studies. CONCLUSIONS Depression is recognized as a polygenic genetic disorder, with multiple genes contributing, each exerting relatively small effects. Future studies should explore not only multiple gene interactions but also epigenetic changes. Presently, research on FKBP5 in affective disorders remains notably limited, highlighting the necessity for further investigations in this domain.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Mental Health, Peking University Sixth Hospital, 100191, Beijing, China
- Tianjin Anding Hospital, Tianjin Municipal Mental Health Center, 300222, Tianjin, China
| | - Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, 100191, Beijing, China.
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), 100191, Beijing, China.
- NHC Key Laboratory of Mental Health, Peking University, 100191, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, 100871, Beijing, China.
- Chinese Institute for Brain Research, 102206, Beijing, China.
| | - Jie Li
- Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 300222, Tianjin, China.
| |
Collapse
|
11
|
Vinberg M, McIntyre RS, Giraldi A, Coello K. Struggling Can Also Show on the Inside: Current Knowledge of the Impact of Childhood Maltreatment on Biomarkers in Mood Disorders. Neuropsychiatr Dis Treat 2024; 20:583-595. [PMID: 38496323 PMCID: PMC10944138 DOI: 10.2147/ndt.s383322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
The link between childhood maltreatment and mood disorders is complex and involves multiple bio-psycho-social factors that affect multiple molecular pathways. The present narrative review aims to clarify the current understanding of the impact of childhood maltreatment on biomarkers in patients with mood disorders and their first-degree relatives. Neurotransmitters, such as serotonin, dopamine, norepinephrine, and hormones (eg the stress hormone cortisol), play a crucial role in regulating mood and emotion. Childhood maltreatment can alter and affect the levels and functioning of these neurotransmitters in the brain; further, childhood maltreatment can lead to structural and connectivity changes in the brain, hence contributing to the development of mood disorders and moderating illness presentation and modifying response to treatments. Childhood maltreatment information, therefore, appears mandatory in treatment planning and is a critical factor in therapeutic algorithms. Further research is needed to fully understand these pathways and develop new treatment modalities for individuals with mood disorders who have experienced childhood maltreatment and effective preventive interventions for individuals at risk of developing mood disorders.
Collapse
Affiliation(s)
- Maj Vinberg
- Mental Health Centre Northern Zealand, the Early Multimodular Prevention, and Intervention Research Institution (EMPIRI) – Mental Health Services CPH, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Annamaria Giraldi
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Sexological Clinic, Mental Health Center Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Klara Coello
- Copenhagen Affective Disorder Research Centre (CADIC), Psychiatric Centre Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
12
|
Hoffmann F, Heim C. [Emotional Abuse in Childhood and Adolescence: Biological Embedding and Clinical Implications]. Prax Kinderpsychol Kinderpsychiatr 2024; 73:4-27. [PMID: 38275227 DOI: 10.13109/prkk.2024.73.1.4] [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: 01/27/2024]
Abstract
Emotional abuse, defined as degrading, manipulative, or neglectful behaviors by caregivers, represents a common adverse experience for children and adolescents, often co-occurring with other maltreatment types. Exposure to emotional abuse significantly affects mental health across the lifespan and is particularly associated with elevated depression risk.This review examinesmechanisms, by which emotional abuse influences brain development and the neuroendocrine stress response system and discusses the roles of genetic vulnerability and epigenetic processes in contributing to an elevated mental health risk. Emotional abuse has similar effects on brain networks responsible for emotion processing and regulation as other maltreatment types.Moreover, it uniquely affects networks related to self-relevant information and socio-cognitive processes. Furthermore, emotional abuse is associated with an impaired recovery of the neuroendocrine response to acute stress. Similar to other maltreatment types, emotional abuse is associated with epigenetic changes in genes regulating the neuroendocrine stress response system that are implicated in increased mental health risk.These findings suggest that emotional abuse has equally detrimental effects on children'smental health as physical or sexual abuse, warranting broader societal awareness and enhanced early detection efforts. Early interventions should prioritize emotion regulation, social cognition, self-esteemenhancement, and relationship- oriented approaches for victims of emotional abuse.
Collapse
Affiliation(s)
- Ferdinand Hoffmann
- Charité - Universitätsmedizin Berlin, Gliedkörperschaft der Freien Universität Berlin und der Humboldt-Universität zu Berlin, Institut für Medizinische Psychologie Deutschland
| | - Christine Heim
- Institut für Medizinische Psychologie Charité - Universitätsmedizin Berlin Luisenstr. 57 10117 Berlin Deutschland
| |
Collapse
|
13
|
Cascino G, Monteleone AM. Early traumatic experiences and the hypothalamus-pituitary-adrenal axis in people with eating disorders: A narrative review. Psychoneuroendocrinology 2024; 159:106665. [PMID: 37944210 DOI: 10.1016/j.psyneuen.2023.106665] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Exposure to trauma during childhood is a non-specific risk factor for psychiatric disorders, including eating disorders (EDs), over the life course. Moreover, an association between stressful life events and the onset/maintenance of EDs has been documented. Therefore, the hypothalamus-pituitary-adrenal (HPA) axis, namely the main component of the endogenous stress response system, has been proposed to be implicated in the pathophysiology of EDs. In this narrative review the current knowledge concerning the effects of early trauma exposure on the HPA axis activity and their putative role in the pathophysiology of EDs will be illustrated. Research findings corroborate the idea that childhood trauma exposure has long-lasting dysregulating effects on the activity of the HPA axis, which may contribute to the biological background of the early trauma-related risk for the development of EDs across the life span. Moreover, literature data support the existence of a "maltreated ecophenotype" in EDs characterized by specific clinic and neuroendocrine features, which may have important implications in treatment programming for such a type of patients.
Collapse
Affiliation(s)
- Giammarco Cascino
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", University of Salerno, Salerno, Italy.
| | | |
Collapse
|
14
|
Reyna NC, Clark BJ, Hamilton DA, Pentkowski NS. Anxiety and Alzheimer's disease pathogenesis: focus on 5-HT and CRF systems in 3xTg-AD and TgF344-AD animal models. Front Aging Neurosci 2023; 15:1251075. [PMID: 38076543 PMCID: PMC10699143 DOI: 10.3389/fnagi.2023.1251075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/25/2023] [Indexed: 02/12/2024] Open
Abstract
Dementia remains one of the leading causes of morbidity and mortality in older adults. Alzheimer's disease (AD) is the most common type of dementia, affecting over 55 million people worldwide. AD is characterized by distinct neurobiological changes, including amyloid-beta protein deposits and tau neurofibrillary tangles, which cause cognitive decline and subsequent behavioral changes, such as distress, insomnia, depression, and anxiety. Recent literature suggests a strong connection between stress systems and AD progression. This presents a promising direction for future AD research. In this review, two systems involved in regulating stress and AD pathogenesis will be highlighted: serotonin (5-HT) and corticotropin releasing factor (CRF). Throughout the review, we summarize critical findings in the field while discussing common limitations with two animal models (3xTg-AD and TgF344-AD), novel pharmacotherapies, and potential early-intervention treatment options. We conclude by highlighting promising future pharmacotherapies and translational animal models of AD and anxiety.
Collapse
Affiliation(s)
- Nicole C. Reyna
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | | | | | | |
Collapse
|
15
|
Eichenauer H, Ehlert U. The association between prenatal famine, DNA methylation and mental disorders: a systematic review and meta-analysis. Clin Epigenetics 2023; 15:152. [PMID: 37716973 PMCID: PMC10505322 DOI: 10.1186/s13148-023-01557-y] [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: 05/02/2023] [Accepted: 08/14/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Undernutrition in pregnant women is an unfavorable environmental condition that can affect the intrauterine development via epigenetic mechanisms and thus have long-lasting detrimental consequences for the mental health of the offspring later in life. One epigenetic mechanism that has been associated with mental disorders and undernutrition is alterations in DNA methylation. The effect of prenatal undernutrition on the mental health of adult offspring can be analyzed through quasi-experimental studies such as famine studies. The present systematic review and meta-analysis aims to analyze the association between prenatal famine exposure, DNA methylation, and mental disorders in adult offspring. We further investigate whether altered DNA methylation as a result of prenatal famine exposure is prospectively linked to mental disorders. METHODS We conducted a systematic search of the databases PubMed and PsycINFO to identify relevant records up to September 2022 on offspring whose mothers experienced famine directly before and/or during pregnancy, examining the impact of prenatal famine exposure on the offspring's DNA methylation and/or mental disorders or symptoms. RESULTS The systematic review showed that adults who were prenatally exposed to famine had an increased risk of schizophrenia and depression. Several studies reported an association between prenatal famine exposure and hyper- or hypomethylation of specific genes. The largest number of studies reported differences in DNA methylation of the IGF2 gene. Altered DNA methylation of the DUSP22 gene mediated the association between prenatal famine exposure and schizophrenia in adult offspring. Meta-analysis confirmed the increased risk of schizophrenia following prenatal famine exposure. For DNA methylation, meta-analysis was not suitable due to different microarrays/data processing approaches and/or unavailable data. CONCLUSION Prenatal famine exposure is associated with an increased risk of mental disorders and DNA methylation changes. The findings suggest that changes in DNA methylation of genes involved in neuronal, neuroendocrine, and immune processes may be a mechanism that promotes the development of mental disorders such as schizophrenia and depression in adult offspring. Such findings are crucial given that undernutrition has risen worldwide, increasing the risk of famine and thus also of negative effects on mental health.
Collapse
Affiliation(s)
- Heike Eichenauer
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland
| | - Ulrike Ehlert
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland.
| |
Collapse
|
16
|
Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C, Zhang W. Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther 2023; 8:309. [PMID: 37644009 PMCID: PMC10465587 DOI: 10.1038/s41392-023-01519-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 08/31/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic, generally episodic and debilitating disease that affects an estimated 300 million people worldwide, but its pathogenesis is poorly understood. The heritability estimate of MDD is 30-40%, suggesting that genetics alone do not account for most of the risk of major depression. Another factor known to associate with MDD involves environmental stressors such as childhood adversity and recent life stress. Recent studies have emerged to show that the biological impact of environmental factors in MDD and other stress-related disorders is mediated by a variety of epigenetic modifications. These epigenetic modification alterations contribute to abnormal neuroendocrine responses, neuroplasticity impairment, neurotransmission and neuroglia dysfunction, which are involved in the pathophysiology of MDD. Furthermore, epigenetic marks have been associated with the diagnosis and treatment of MDD. The evaluation of epigenetic modifications holds promise for further understanding of the heterogeneous etiology and complex phenotypes of MDD, and may identify new therapeutic targets. Here, we review preclinical and clinical epigenetic findings, including DNA methylation, histone modification, noncoding RNA, RNA modification, and chromatin remodeling factor in MDD. In addition, we elaborate on the contribution of these epigenetic mechanisms to the pathological trait variability in depression and discuss how such mechanisms can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Biao Yang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Radiology, Columbia University, New York, NY, 10032, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuang Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Medical Big Data Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
17
|
Sun N, Cui WQ, Min XM, Zhang GM, Liu JZ, Wu HY. A new perspective on hippocampal synaptic plasticity and post-stroke depression. Eur J Neurosci 2023; 58:2961-2984. [PMID: 37518943 DOI: 10.1111/ejn.16093] [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: 04/11/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Post-stroke depression, a common complication after stroke, severely affects the recovery and quality of life of patients with stroke. Owing to its complex mechanisms, post-stroke depression treatment remains highly challenging. Hippocampal synaptic plasticity is one of the key factors leading to post-stroke depression; however, the precise molecular mechanisms remain unclear. Numerous studies have found that neurotrophic factors, protein kinases and neurotransmitters influence depressive behaviour by modulating hippocampal synaptic plasticity. This review further elaborates on the role of hippocampal synaptic plasticity in post-stroke depression by summarizing recent research and analysing possible molecular mechanisms. Evidence for the correlation between hippocampal mechanisms and post-stroke depression helps to better understand the pathological process of post-stroke depression and improve its treatment.
Collapse
Affiliation(s)
- Ning Sun
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Man Min
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jia-Zheng Liu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
18
|
Marinho LSR, Chiarantin GMD, Ikebara JM, Cardoso DS, de Lima-Vasconcellos TH, Higa GSV, Ferraz MSA, De Pasquale R, Takada SH, Papes F, Muotri AR, Kihara AH. The impact of antidepressants on human neurodevelopment: Brain organoids as experimental tools. Semin Cell Dev Biol 2023; 144:67-76. [PMID: 36115764 DOI: 10.1016/j.semcdb.2022.09.007] [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: 07/06/2022] [Revised: 09/10/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022]
Abstract
The use of antidepressants during pregnancy benefits the mother's well-being, but the effects of such substances on neurodevelopment remain poorly understood. Moreover, the consequences of early exposure to antidepressants may not be immediately apparent at birth. In utero exposure to selective serotonin reuptake inhibitors (SSRIs) has been related to developmental abnormalities, including a reduced white matter volume. Several reports have observed an increased incidence of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) after prenatal exposure to SSRIs such as sertraline, the most widely prescribed SSRI. The advent of human-induced pluripotent stem cell (hiPSC) methods and assays now offers appropriate tools to test the consequences of such compounds for neurodevelopment in vitro. In particular, hiPSCs can be used to generate cerebral organoids - self-organized structures that recapitulate the morphology and complex physiology of the developing human brain, overcoming the limitations found in 2D cell culture and experimental animal models for testing drug efficacy and side effects. For example, single-cell RNA sequencing (scRNA-seq) and electrophysiological measurements on organoids can be used to evaluate the impact of antidepressants on the transcriptome and neuronal activity signatures in developing neurons. While the analysis of large-scale transcriptomic data depends on dimensionality reduction methods, electrophysiological recordings rely on temporal data series to discriminate statistical characteristics of neuronal activity, allowing for the rigorous analysis of the effects of antidepressants and other molecules that affect the developing nervous system, especially when applied in combination with relevant human cellular models such as brain organoids.
Collapse
Affiliation(s)
| | | | - Juliane Midori Ikebara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | | | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil; Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | | | - Roberto De Pasquale
- Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; Center for Medicinal Chemistry, University of Campinas, Campinas, SP 13083-875, Brazil; Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, Center for Academic Research and Training in Anthropogeny, Kavli Institute for Brain and Mind, Archealization Center (ArchC), La Jolla, CA 92037, USA.
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil.
| |
Collapse
|
19
|
Corrigan M, O'Rourke A, Moran B, Fletcher J, Harkin A. Inflammation in the pathogenesis of depression: a disorder of neuroimmune origin. Neuronal Signal 2023; 7:NS20220054. [PMID: 37457896 PMCID: PMC10345431 DOI: 10.1042/ns20220054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
There are several hypotheses concerning the underlying pathophysiological mechanisms of major depression, which centre largely around adaptive changes in neuronal transmission and plasticity, neurogenesis, and circuit and regional connectivity. The immune and endocrine systems are commonly implicated in driving these changes. An intricate interaction of stress hormones, innate immune cells and the actions of soluble mediators of immunity within the nervous system is described as being associated with the symptoms of depression. Bridging endocrine and immune processes to neurotransmission and signalling within key cortical and limbic brain circuits are critical to understanding depression as a disorder of neuroimmune origins. Emergent areas of research include a growing recognition of the adaptive immune system, advances in neuroimaging techniques and mechanistic insights gained from transgenic animals. Elucidation of glial-neuronal interactions is providing additional avenues into promising areas of research, the development of clinically relevant disease models and the discovery of novel therapies. This narrative review focuses on molecular and cellular mechanisms that are influenced by inflammation and stress. The aim of this review is to provide an overview of our current understanding of depression as a disorder of neuroimmune origin, focusing on neuroendocrine and neuroimmune dysregulation in depression pathophysiology. Advances in current understanding lie in pursuit of relevant biomarkers, as the potential of biomarker signatures to improve clinical outcomes is yet to be fully realised. Further investigations to expand biomarker panels including integration with neuroimaging, utilising individual symptoms to stratify patients into more homogenous subpopulations and targeting the immune system for new treatment approaches will help to address current unmet clinical need.
Collapse
Affiliation(s)
- Myles Corrigan
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
- Transpharmation Ireland, Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| | - Aoife M. O'Rourke
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity Biosciences Institute, Trinity College, Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences and Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
20
|
Lin PI, Eapen V. New insights into the link between childhood adversity and epigenetic changes. THE LANCET. CHILD & ADOLESCENT HEALTH 2023:S2352-4642(23)00106-2. [PMID: 37327799 DOI: 10.1016/s2352-4642(23)00106-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 06/18/2023]
Affiliation(s)
- Ping-I Lin
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Kensington, NSW 2052, Australia; Mental Health Research Unit, South Western Sydney Local Health District, Liverpool NSW, Australia; Department of Mental Health, School of Medicine, Western Sydney University, Sydney, NSW, Australia.
| | - Valsamma Eapen
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Kensington, NSW 2052, Australia; Academic Unit of Infant, Child, and Adolescent Psychiatry, South Western Sydney Local Health District, Liverpool NSW, Australia
| |
Collapse
|
21
|
Gladkova MG, Leidmaa E, Anderzhanova EA. Epidrugs in the Therapy of Central Nervous System Disorders: A Way to Drive on? Cells 2023; 12:1464. [PMID: 37296584 PMCID: PMC10253154 DOI: 10.3390/cells12111464] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023] Open
Abstract
The polygenic nature of neurological and psychiatric syndromes and the significant impact of environmental factors on the underlying developmental, homeostatic, and neuroplastic mechanisms suggest that an efficient therapy for these disorders should be a complex one. Pharmacological interventions with drugs selectively influencing the epigenetic landscape (epidrugs) allow one to hit multiple targets, therefore, assumably addressing a wide spectrum of genetic and environmental mechanisms of central nervous system (CNS) disorders. The aim of this review is to understand what fundamental pathological mechanisms would be optimal to target with epidrugs in the treatment of neurological or psychiatric complications. To date, the use of histone deacetylases and DNA methyltransferase inhibitors (HDACis and DNMTis) in the clinic is focused on the treatment of neoplasms (mainly of a glial origin) and is based on the cytostatic and cytotoxic actions of these compounds. Preclinical data show that besides this activity, inhibitors of histone deacetylases, DNA methyltransferases, bromodomains, and ten-eleven translocation (TET) proteins impact the expression of neuroimmune inflammation mediators (cytokines and pro-apoptotic factors), neurotrophins (brain-derived neurotropic factor (BDNF) and nerve growth factor (NGF)), ion channels, ionotropic receptors, as well as pathoproteins (β-amyloid, tau protein, and α-synuclein). Based on this profile of activities, epidrugs may be favorable as a treatment for neurodegenerative diseases. For the treatment of neurodevelopmental disorders, drug addiction, as well as anxiety disorders, depression, schizophrenia, and epilepsy, contemporary epidrugs still require further development concerning a tuning of pharmacological effects, reduction in toxicity, and development of efficient treatment protocols. A promising strategy to further clarify the potential targets of epidrugs as therapeutic means to cure neurological and psychiatric syndromes is the profiling of the epigenetic mechanisms, which have evolved upon actions of complex physiological lifestyle factors, such as diet and physical exercise, and which are effective in the management of neurodegenerative diseases and dementia.
Collapse
Affiliation(s)
- Marina G. Gladkova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Este Leidmaa
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 50411 Tartu, Estonia
| | | |
Collapse
|
22
|
Peckham H. Introducing the Neuroplastic Narrative: a non-pathologizing biological foundation for trauma-informed and adverse childhood experience aware approaches. Front Psychiatry 2023; 14:1103718. [PMID: 37283710 PMCID: PMC10239852 DOI: 10.3389/fpsyt.2023.1103718] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/20/2023] [Indexed: 06/08/2023] Open
Abstract
Most people accessing mental health services have adverse childhood experiences (ACEs) and/or histories of complex trauma. In recognition of this, there are calls to move away from medical model approaches and move toward trauma-informed approaches which privilege the impact of life experience over underlying pathology in the etiology of emotional and psychological suffering. Trauma-informed approaches lack a biological narrative linking trauma and adversity to later suffering. In its absence, this suffering is diagnosed and treated as a mental illness. This study articulates the Neuroplastic Narrative, a neuroecological theory that fills this gap, conceptualizing emotional and psychological suffering as the cost of surviving and adapting to the impinging environments of trauma and adversity. The Neuroplastic Narrative privileges lived experience and recognizes that our experiences become embedded in our biology through evolved mechanisms that ultimately act to preserve survival in the service of reproduction. Neuroplasticity refers to the capacity of neural systems to adapt and change. Our many evolved neuroplastic mechanisms including epigenetics, neurogenesis, synaptic plasticity, and white matter plasticity allow us to learn from, and adapt to, past experiences. This learning and adaption in turn allows us to better anticipate and physiologically prepare for future experiences that (nature assumes) are likely to occur, based on past experiences. However, neuroplastic mechanisms cannot discriminate between experiences; they function to embed experience regardless of the quality of that experience, generating vicious or virtuous cycles of psychobiological anticipation, to help us survive or thrive in futures that resemble our privileged or traumatic pasts. The etiology of suffering that arises from this process is not a pathology (a healthy brain is a brain that can adapt to experience) but is the evolutionary cost of surviving traumatizing environments. Misidentifying this suffering as a pathology and responding with diagnosis and medication is not trauma-informed and may cause iatrogenic harm, in part through perpetuating stigma and exacerbating the shame which attends complex trauma and ACEs. As an alternative, this study introduces the Neuroplastic Narrative, which is situated within an evolutionary framework. The Neuroplastic Narrative complements both Life History and Attachment Theory and provides a non-pathologizing, biological foundation for trauma-informed and Adverse Childhood Experience aware approaches.
Collapse
Affiliation(s)
- Haley Peckham
- Centre for Mental Health Nursing, School of Health Sciences, University of Melbourne, Carlton, VIC, Australia
- Department of Psychology, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
23
|
Yang L, Jia FF, Lu XY, Jia CX. Internalizing and externalizing problems mediate the relationship between maltreatment and self-harm among UK adolescents. J Affect Disord 2023; 333:240-248. [PMID: 37084976 DOI: 10.1016/j.jad.2023.04.046] [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: 02/26/2023] [Revised: 03/30/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND Maltreatment is a significant predictive factor for self-harm in adolescents. Internalizing and externalizing problems are both common psychopathological issues in adolescents. This study aimed to look into the link between maltreatment and self-harm in a large sample of adolescents in the UK, as well as the mediating effects that internalizing and externalizing problems play in this link. METHODS Data were pulled from the UK Millennium Cohort Study, and a total of 8894 adolescents were included in this analysis. All variables were assessed by Questionnaires. Path analysis was performed to assess the mediating effects of internalizing and externalizing problems in the link between maltreatment and self-harm. RESULTS 23.4 % of samples reported self-harm incidents during the preceding 12 months. Emotional abuse and physical abuse were significantly related to self-harm, and adolescents who had experienced multiple forms of maltreatment were more prone to self-harm. Mediation analysis revealed that internalizing problems were the primary mediator in the link between emotional abuse and self-harm, with the mediation effect size being 0.29. Internalizing and externalizing problems performed similarly in the link between physical abuse and self-harm, with mediation effect sizes of 0.23 and 0.19, respectively. LIMITATIONS The majority of the data was gathered through self-reporting. CONCLUSIONS Emotional abuse and physical abuse were significant predictors of self-harm, and their links with self-harm were partially mediated via internalizing and externalizing problems. Better supervision of maltreatment, and timely intervention for both internalizing and externalizing problems, are critical in preventing self-harm among adolescents.
Collapse
Affiliation(s)
- Li Yang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fei-Fei Jia
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Xin-Yi Lu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Cun-Xian Jia
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
24
|
Teo CH, Wong ACH, Sivakumaran RN, Parhar I, Soga T. Gender Differences in Cortisol and Cortisol Receptors in Depression: A Narrative Review. Int J Mol Sci 2023; 24:ijms24087129. [PMID: 37108291 PMCID: PMC10138698 DOI: 10.3390/ijms24087129] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Stress is known to have a significant impact on mental health. While gender differences can be found in stress response and mental disorders, there are limited studies on the neuronal mechanisms of gender differences in mental health. Here, we discuss gender and cortisol in depression as presented by recent clinical studies, as well as gender differences in the role of glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs) in stress-associated mental disorders. When examining clinical studies drawn from PubMed/MEDLINE (National Library of Medicine) and EMBASE, salivary cortisol generally showed no gender correlation. However, young males were reported to show heightened cortisol reactivity compared to females of similar age in depression. Pubertal hormones, age, early life stressors, and types of bio-samples for cortisol measurement affected the recorded cortisol levels. The role of GRs and MRs in the HPA axis could be different between males and females during depression, with increased HPA activity and upregulated MR expression in male mice, while the inverse happened in female mice. The functional heterogeneity and imbalance of GRs and MRs in the brain may explain gender differences in mental disorders. This knowledge and understanding will support the development of gender-specific diagnostic markers involving GRs and MRs in depression.
Collapse
Affiliation(s)
- Chuin Hau Teo
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Ally Chai Hui Wong
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Rooba Nair Sivakumaran
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Ishwar Parhar
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Kuala Lumpur 47500, Selangor, Malaysia
| |
Collapse
|
25
|
Overnight Corticosterone and Gene Expression in Mouse Hippocampus: Time Course during Resting Period. Int J Mol Sci 2023; 24:ijms24032828. [PMID: 36769150 PMCID: PMC9917930 DOI: 10.3390/ijms24032828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The aim of the experiment was to test the effect of an elevated level of glucocorticoids on the mouse hippocampal transcriptome after 12 h of treatment with corticosterone that was administered during an active phase of the circadian cycle. Additionally, we also tested the circadian changes in gene expression and the decay time of transcriptomic response to corticosterone. Gene expression was analyzed using microarrays. Obtained results show that transcriptomic responses to glucocorticoids are heterogeneous in terms of the decay time with some genes displaying persistent changes in expression during 9 h of rest. We have also found a considerable overlap between genes regulated by corticosterone and genes implicated previously in stress response. The examples of such genes are Acer2, Agt, Apod, Aqp4, Etnppl, Fabp7, Fam107a, Fjx1, Fmo2, Galnt15, Gjc2, Heph, Hes5, Htra1, Jdp2, Kif5a, Lfng, Lrg1, Mgp, Mt1, Pglyrp1, Pla2g3, Plin4, Pllp, Ptgds, Ptn, Slc2a1, Slco1c1, Sult1a1, Thbd and Txnip. This indicates that the applied model is a useful tool for the investigation of mechanisms underlying the stress response.
Collapse
|
26
|
Epigenetic differences in stress response gene FKBP5 among children with abusive vs accidental injuries. Pediatr Res 2023:10.1038/s41390-022-02441-w. [PMID: 36624283 DOI: 10.1038/s41390-022-02441-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/08/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Survivors of child abuse experience high rates of adverse physical and mental health outcomes. Epigenetic alterations in the stress response system, the FKBP5 gene specifically, have been implicated as one mechanism that may link abuse to lifelong health issues. Prior studies primarily included older individuals with a remote history of maltreatment; our objective was to test for differential methylation of FKBP5 in children with abusive vs accidental injuries at the time of diagnosis. METHODS We conducted a cross-sectional pilot study of acutely injured children <4 years old at two children's hospitals (n = 82). Research personnel collected injury histories, buccal swabs (n = 65), and blood samples (n = 25) to measure DNA methylation. An expert panel classified the injuries as abusive, accidental, or indeterminate. RESULTS Children with abusive as compared to accidental injuries had lower methylation of the FKBP5 promoter in buccal and blood cells, even after controlling for injury severity, socioeconomic status, and psychosocial risk factors. CONCLUSION These findings suggest that epigenetic variation in FKBP5 may occur at the earliest indication of abuse and may be associated with delayed resolution of the HPA axis stress response. Additional testing for epigenetic differences in larger sample sizes is needed to further verify these findings. IMPACT Children (<4 years old) with abusive compared to accidental injuries showed lower methylation of the FKBP5 promoter in buccal and blood cells at the time of initial diagnosis even after controlling for injury severity, socioeconomic status, and psychosocial risk factors. Early childhood physical abuse may impact the epigenetic regulation of the stress response system, including demethylation within promoters and enhancers of the FKBP5 gene, even at the earliest indication of abuse. The findings are important because unmitigated stress is associated with adverse health outcomes throughout the life-course.
Collapse
|
27
|
Comtois-Cabana M, Barr E, Provençal N, Ouellet-Morin I. Association between child maltreatment and depressive symptoms in emerging adulthood: The mediating and moderating roles of DNA methylation. PLoS One 2023; 18:e0280203. [PMID: 36634080 PMCID: PMC9836296 DOI: 10.1371/journal.pone.0280203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Prospective studies suggest that child maltreatment substantially increases the risk for depression in adulthood. However, the mechanisms underlying this association require further elucidation. In recent years, DNA methylation has emerged as a potential mechanism by which maltreatment experiences (a) could partly explain the emergence or aggravation of depressive symptoms (i.e., mediation) and/or (b) could increase (or decrease) the risk for depressive symptoms (i.e., moderation). The present study tested whether the methylation levels of nine candidate genes mediated and/or moderated the association between maltreatment experiences in childhood and depressive symptoms in emerging adulthood. The sample comprised 156 men aged between 18 and 35 years. Maltreatment experiences and depressive symptoms were assessed retrospectively using self-reported questionnaires. Methylation levels of nine candidate genes (COMT, FKBP5, IL6, IL10, MAOA, NR3C1, OXTR, SLC6A3 and SLC6A4), previously reported to be sensitive to early-life stress, were quantified from saliva samples. Maltreatment experiences in childhood were significantly associated with depressive symptoms in emerging adulthood. Both maltreatment experiences and depressive symptoms were associated with the methylation levels of two genomic sites, which cumulatively, but not individually, explained 16% of the association between maltreatment experiences in childhood and depressive symptoms in emerging adulthood. Moreover, maltreatment experiences in childhood interacted with the methylation levels of fourteen genomic sites, which cumulatively, but not individually, modulated the level of depressive symptoms in young male adults who were maltreated as children. However, none of these effects survived multiple testing correction. These findings bring attention to the cumulative effects of DNA methylation measured in several candidate genes on the risk of reporting depressive symptoms following maltreatment experiences in childhood. Nonetheless, future studies need to clarify the robustness of these putative cumulative effects in larger samples and longitudinal cohorts.
Collapse
Affiliation(s)
- Maude Comtois-Cabana
- Department of Psychology, University of Montreal, Montreal, Quebec, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Emily Barr
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Nadine Provençal
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Isabelle Ouellet-Morin
- School of Criminology, University of Montreal, Montreal, Quebec, Canada
- Research Center of the Montreal Mental Health University Institute, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
28
|
Kanes SJ, Dennie L, Perera P. Targeting the Arginine Vasopressin V 1b Receptor System and Stress Response in Depression and Other Neuropsychiatric Disorders. Neuropsychiatr Dis Treat 2023; 19:811-828. [PMID: 37077711 PMCID: PMC10106826 DOI: 10.2147/ndt.s402831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
A healthy stress response is critical for good mental and overall health and promotes neuronal growth and adaptation, but the intricately balanced biological mechanisms that facilitate a stress response can also result in predisposition to disease when that equilibrium is disrupted. The hypothalamic-pituitary-adrenal (HPA) axis neuroendocrine system plays a critical role in the body's response and adaptation to stress, and vasopressinergic regulation of the HPA axis is critical to maintaining system responsiveness during chronic stress. However, exposure to repeated or excessive physical or emotional stress or trauma can shift the body's stress response equilibrium to a "new normal" underpinned by enduring changes in HPA axis function. Exposure to early life stress due to adverse childhood experiences can also lead to lasting neurobiological changes, including in HPA axis function. HPA axis impairment in patients with depression is considered among the most reliable findings in biological psychiatry, and chronic stress has been shown to play a major role in the pathogenesis and onset of depression and other neuropsychiatric disorders. Modulating HPA axis activity, for example via targeted antagonism of the vasopressin V1b receptor, is a promising approach for patients with depression and other neuropsychiatric disorders associated with HPA axis impairment. Despite favorable preclinical indications in animal models, demonstration of clinical efficacy for the treatment of depressive disorders by targeting HPA axis dysfunction has been challenging, possibly due to the heterogeneity and syndromal nature of depressive disorders. Measures of HPA axis function, such as elevated cortisol levels, may be useful biomarkers for identifying patients who may benefit from treatments that modulate HPA axis activity. Utilizing clinical biomarkers to identify subsets of patients with impaired HPA axis function who may benefit is a promising next step in fine-tuning HPA axis activity via targeted antagonism of the V1b receptor.
Collapse
Affiliation(s)
- Stephen J Kanes
- EmbarkNeuro, Oakland, CA, USA
- Correspondence: Stephen J Kanes, EmbarkNeuro, Inc, 1111 Broadway, Suite 1300, Oakland, CA, 94607, USA, Tel +1 610 757 7821, Email
| | | | | |
Collapse
|
29
|
Schrott R, Song A, Ladd-Acosta C. Epigenetics as a Biomarker for Early-Life Environmental Exposure. Curr Environ Health Rep 2022; 9:604-624. [PMID: 35907133 DOI: 10.1007/s40572-022-00373-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW There is interest in evaluating the developmental origins of health and disease (DOHaD) which emphasizes the role of prenatal and early-life environments on non-communicable health outcomes throughout the life course. The ability to rigorously assess and identify early-life risk factors for later health outcomes, including those with childhood onset, in large population samples is often limited due to measurement challenges such as impractical costs associated with prospective studies with a long follow-up duration, short half-lives for some environmental toxicants, and lack of biomarkers that capture inter-individual differences in biologic response to external environments. RECENT FINDINGS Epigenomic patterns, and DNA methylation in particular, have emerged as a potential objective biomarker to address some of these study design and exposure measurement challenges. In this article, we summarize the literature to date on epigenetic changes associated with specific prenatal and early-life exposure domains as well as exposure mixtures in human observational studies and their biomarker potential. Additionally, we highlight evidence for other types of epigenetic patterns to serve as exposure biomarkers. Evidence strongly supports epigenomic biomarkers of exposure that are detectable across the lifespan and across a range of exposure domains. Current and future areas of research in this field seek to expand these lines of evidence to other environmental exposures, to determine their specificity, and to develop predictive algorithms and methylation scores that can be used to evaluate early-life risk factors for health outcomes across the life span.
Collapse
Affiliation(s)
- Rose Schrott
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ashley Song
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
30
|
Notomi R, Sasaki S, Taniguchi Y. Recognition of 5-methyl-CG and CG base pairs in duplex DNA with high stability using antiparallel-type triplex-forming oligonucleotides with 2-guanidinoethyl-2'-deoxynebularine. Nucleic Acids Res 2022; 50:12071-12081. [PMID: 36454012 PMCID: PMC9757063 DOI: 10.1093/nar/gkac1110] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/26/2022] [Accepted: 11/05/2022] [Indexed: 12/03/2022] Open
Abstract
The formation of triplex DNA is a site-specific recognition method that directly targets duplex DNA. However, triplex DNA formation is generally formed for the GC and AT base pairs of duplex DNA, and there are no natural nucleotides that recognize the CG and TA base pairs, or even the 5-methyl-CG (5mCG) base pair. Moreover, duplex DNA, including 5mCG base pairs, epigenetically regulates gene expression in vivo, and thus targeting strategies are of biological importance. Therefore, the development of triplex-forming oligonucleotides (TFOs) with artificial nucleosides that selectively recognize these base pairs with high affinity is needed. We recently reported that 2'-deoxy-2-aminonebularine derivatives exhibited the ability to recognize 5mCG and CG base pairs in triplex formation; however, this ability was dependent on sequences. Therefore, we designed and synthesized new nucleoside derivatives based on the 2'-deoxy-nebularine (dN) skeleton to shorten the linker length connecting to the hydrogen-bonding unit in formation of the antiparallel motif triplex. We successfully demonstrated that TFOs with 2-guanidinoethyl-2'-deoxynebularine (guanidino-dN) recognized 5mCG and CG base pairs with very high affinity in all four DNA sequences with different adjacent nucleobases of guanidino-dN as well as in the promoter sequences of human genes containing 5mCG base pairs with a high DNA methylation frequency.
Collapse
Affiliation(s)
- Ryotaro Notomi
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shigeki Sasaki
- Graduate School of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch Machi, Sasebo city, Nagasaki 859-3298, Japan
| | - Yosuke Taniguchi
- To whom correspondence should be addressed. Tel: +81 92 642 6569; Fax: +81 92 642 6876;
| |
Collapse
|
31
|
Paoli C, Misztak P, Mazzini G, Musazzi L. DNA Methylation in Depression and Depressive-Like Phenotype: Biomarker or Target of Pharmacological Intervention? Curr Neuropharmacol 2022; 20:2267-2291. [PMID: 35105292 PMCID: PMC9890294 DOI: 10.2174/1570159x20666220201084536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 12/29/2022] Open
Abstract
Major depressive disorder (MDD) is a debilitating psychiatric disorder, the third leading global cause of disability. Regarding aetiopathogenetic mechanisms involved in the onset of depressive disorders, the interaction between genetic vulnerability traits and environmental factors is believed to play a major role. Although much is still to be elucidated about the mechanisms through which the environment can interact with genetic background shaping the disease risk, there is a general agreement about a key role of epigenetic marking. In this narrative review, we focused on the association between changes in DNA methylation patterns and MDD or depressive-like phenotype in animal models, as well as mechanisms of response to antidepressant drugs. We discussed studies presenting DNA methylation changes at specific genes of interest and profiling analyses in both patients and animal models of depression. Overall, we collected evidence showing that DNA methylation could not only be considered as a promising epigenetic biomarker of pathology but could also help in predicting antidepressant treatment efficacy. Finally, we discussed the hypothesis that specific changes in DNA methylation signature could play a role in aetiopathogenetic processes as well as in the induction of antidepressant effect.
Collapse
Affiliation(s)
- Caterina Paoli
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- School of Pharmacy, Pharmacy Unit, University of Camerino, 62032 Camerino, Italy
| | - Paulina Misztak
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giulia Mazzini
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Laura Musazzi
- Department of Medicine and Surgery, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
32
|
Šalamon Arčan I, Kouter K, Videtič Paska A. Depressive disorder and antidepressants from an epigenetic point of view. World J Psychiatry 2022; 12:1150-1168. [PMID: 36186508 PMCID: PMC9521527 DOI: 10.5498/wjp.v12.i9.1150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/27/2022] [Accepted: 08/05/2022] [Indexed: 02/05/2023] Open
Abstract
Depressive disorder is a complex, heterogeneous disease that affects approximately 280 million people worldwide. Environmental, genetic, and neurobiological factors contribute to the depressive state. Since the nervous system is susceptible to shifts in activity of epigenetic modifiers, these allow for significant plasticity and response to rapid changes in the environment. Among the most studied epigenetic modifications in depressive disorder is DNA methylation, with findings centered on the brain-derived neurotrophic factor gene, the glucocorticoid receptor gene, and the serotonin transporter gene. In order to identify biomarkers that would be useful in clinical settings, for diagnosis and for treatment response, further research on antidepressants and alterations they cause in the epigenetic landscape throughout the genome is needed. Studies on cornerstone antidepressants, such as selective serotonin reuptake inhibitors, selective serotonin and norepinephrine reuptake inhibitors, norepinephrine, and dopamine reuptake inhibitors and their effects on depressive disorder are available, but systematic conclusions on their effects are still hard to draw due to the highly heterogeneous nature of the studies. In addition, two novel drugs, ketamine and esketamine, are being investigated particularly in association with treatment of resistant depression, which is one of the hot topics of contemporary research and the field of precision psychiatry.
Collapse
Affiliation(s)
- Iris Šalamon Arčan
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| | - Katarina Kouter
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| | - Alja Videtič Paska
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana SI-1000, Slovenia
| |
Collapse
|
33
|
Cheng Z, Su J, Zhang K, Jiang H, Li B. Epigenetic Mechanism of Early Life Stress-Induced Depression: Focus on the Neurotransmitter Systems. Front Cell Dev Biol 2022; 10:929732. [PMID: 35865627 PMCID: PMC9294154 DOI: 10.3389/fcell.2022.929732] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Depression has an alarmingly high prevalence worldwide. A growing body of evidence indicates that environmental factors significantly affect the neural development and function of the central nervous system and then induce psychiatric disorders. Early life stress (ELS) affects brain development and has been identified as a major cause of depression. It could promote susceptibility to stress in adulthood. Recent studies have found that ELS induces epigenetic changes that subsequently affect transcriptional rates of differentially expressed genes. The epigenetic modifications involved in ELS include histone modifications, DNA methylation, and non-coding RNA. Understanding of these genetic modifications may identify mechanisms that may lead to new interventions for the treatment of depression. Many reports indicate that different types of ELS induce epigenetic modifications of genes involved in the neurotransmitter systems, such as the dopaminergic system, the serotonergic system, the gamma-aminobutyric acid (GABA)-ergic system, and the glutamatergic system, which further regulate gene expression and ultimately induce depression-like behaviors. In this article, we review the effects of epigenetic modifications on the neurotransmitter systems in depression-like outcomes produced by different types of ELS in recent years, aiming to provide new therapeutic targets for patients who suffer from depression.
Collapse
Affiliation(s)
- Ziqian Cheng
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Jingyun Su
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
| | - Kai Zhang
- Central Laboratory, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Huiyi Jiang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Huiyi Jiang, ; Bingjin Li,
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, Second Hospital of Jilin University, Changchun, China
- Engineering Lab on Screening of Antidepressant Drugs, Jilin Province Development and Reform Commission, Changchun, China
- *Correspondence: Huiyi Jiang, ; Bingjin Li,
| |
Collapse
|
34
|
Alameda L, Trotta G, Quigley H, Rodriguez V, Gadelrab R, Dwir D, Dempster E, Wong CCY, Forti MD. Can epigenetics shine a light on the biological pathways underlying major mental disorders? Psychol Med 2022; 52:1645-1665. [PMID: 35193719 PMCID: PMC9280283 DOI: 10.1017/s0033291721005559] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/30/2021] [Accepted: 12/29/2021] [Indexed: 12/27/2022]
Abstract
A significant proportion of the global burden of disease can be attributed to mental illness. Despite important advances in identifying risk factors for mental health conditions, the biological processing underlying causal pathways to disease onset remain poorly understood. This represents a limitation to implement effective prevention and the development of novel pharmacological treatments. Epigenetic mechanisms have emerged as mediators of environmental and genetic risk factors which might play a role in disease onset, including childhood adversity (CA) and cannabis use (CU). Particularly, human research exploring DNA methylation has provided new and promising insights into the role of biological pathways implicated in the aetio-pathogenesis of psychiatric conditions, including: monoaminergic (Serotonin and Dopamine), GABAergic, glutamatergic, neurogenesis, inflammatory and immune response and oxidative stress. While these epigenetic changes have been often studied as disease-specific, similarly to the investigation of environmental risk factors, they are often transdiagnostic. Therefore, we aim to review the existing literature on DNA methylation from human studies of psychiatric diseases (i) to identify epigenetic modifications mapping onto biological pathways either transdiagnostically or specifically related to psychiatric diseases such as Eating Disorders, Post-traumatic Stress Disorder, Bipolar and Psychotic Disorder, Depression, Autism Spectrum Disorder and Anxiety Disorder, and (ii) to investigate a convergence between some of these epigenetic modifications and the exposure to known risk factors for psychiatric disorders such as CA and CU, as well as to other epigenetic confounders in psychiatry research.
Collapse
Affiliation(s)
- Luis Alameda
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Departamento de Psiquiatría, Centro Investigación Biomedica en Red de Salud Mental (CIBERSAM), Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Sevilla, Spain
| | - Giulia Trotta
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
| | - Harriet Quigley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Victoria Rodriguez
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Romayne Gadelrab
- Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniella Dwir
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Emma Dempster
- University of Exeter Medical School, University of Exeter, Barrack Road, Exeter, UK
| | - Chloe C. Y. Wong
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
35
|
Carpenter JR, Jablonski KA, Koncinsky J, Varner MW, Gyamfi-Bannerman C, Joss-Moore LA. Antenatal Steroids and Cord Blood T-cell Glucocorticoid Receptor DNA Methylation and Exon 1 Splicing. Reprod Sci 2022; 29:1513-1523. [PMID: 35146694 PMCID: PMC9010373 DOI: 10.1007/s43032-022-00859-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 01/18/2022] [Indexed: 02/03/2023]
Abstract
Antenatal administration of glucocorticoids such as betamethasone (BMZ) during the late preterm period improves neonatal respiratory outcomes. However, glucocorticoids may elicit programming effects on immune function and gene regulation. Here, we test the hypothesis that exposure to antenatal BMZ alters cord blood immune cell composition in association with altered DNA methylation and alternatively expressed Exon 1 transcripts of the glucocorticoid receptor (GR) gene in cord blood CD4+ T-cells. Cord blood was collected from 51 subjects in the Antenatal Late Preterm Steroids Trial: 27 BMZ, 24 placebo. Proportions of leukocytes were compared between BMZ and placebo. In CD4+ T-cells, methylation at CpG sites in the GR promoter regions and expression of GR mRNA exon 1 variants were compared between BMZ and placebo. BMZ was associated with an increase in granulocytes (51.6% vs. 44.7% p = 0.03) and a decrease in lymphocytes (36.8% vs. 43.0% p = 0.04) as a percent of the leukocyte population vs. placebo. Neither GR methylation nor exon 1 transcript levels differed between groups. BMZ is associated with altered cord blood leukocyte proportions, although no associated alterations in GR methylation were observed.
Collapse
Affiliation(s)
| | - Kathleen A. Jablonski
- Milken School of Public Health, Biostatistics Center, George Washington University, Washington, D.C, USA
| | | | - Michael W. Varner
- Obstetrics & Gynecology, University of Utah, Salt Lake City, Utah, USA
| | | | - Lisa A. Joss-Moore
- Pediatrics, University of Utah, Salt Lake City, Utah, USA,Corresponding author: Lisa Joss-Moore, Ph.D., University of Utah, Department of Pediatrics, 295 Chipeta Way, Salt Lake City, Utah, 84108, USA, Ph: 1-801-213-3494,
| | | |
Collapse
|
36
|
Cuomo M, Florio E, Della Monica R, Costabile D, Buonaiuto M, Di Risi T, De Riso G, Sarnataro A, Cocozza S, Visconti R, Chiariotti L. Epigenetic remodelling of Fxyd1 promoters in developing heart and brain tissues. Sci Rep 2022; 12:6471. [PMID: 35440736 PMCID: PMC9018693 DOI: 10.1038/s41598-022-10365-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/04/2022] [Indexed: 11/24/2022] Open
Abstract
FXYD1 is a key protein controlling ion channel transport. FXYD1 exerts its function by regulating Na+/K+-ATPase activity, mainly in brain and cardiac tissues. Alterations of the expression level of the FXYD1 protein cause diastolic dysfunction and arrhythmias in heart and decreased neuronal dendritic tree and spine formation in brain. Moreover, FXYD1, a target of MeCP2, plays a crucial role in the pathogenesis of the Rett syndrome, a neurodevelopmental disorder. Thus, the amount of FXYD1 must be strictly controlled in a tissue specific manner and, likely, during development. Epigenetic modifications, particularly DNA methylation, represent the major candidate mechanism that may regulate Fxyd1 expression. In the present study, we performed a comprehensive DNA methylation analysis and mRNA expression level measurement of the two Fxyd1 transcripts, Fxyd1a and Fxyd1b, in brain and heart tissues during mouse development. We found that DNA methylation at Fxyd1a increased during brain development and decreased during heart development along with coherent changes in mRNA expression levels. We also applied ultra-deep methylation analysis to detect cell to cell methylation differences and to identify possible distinct methylation profile (epialleles) distribution between heart and brain and in different developmental stages. Our data indicate that the expression of Fxyd1 transcript isoforms inversely correlates with DNA methylation in developing brain and cardiac tissues suggesting the existence of a temporal-specific epigenetic program. Moreover, we identified a clear remodeling of epiallele profiles which were distinctive for single developmental stage both in brain and heart tissues.
Collapse
Affiliation(s)
- Mariella Cuomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy. .,CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy.
| | - Ermanno Florio
- Department of Medicine, University of California, San Diego UCSD, Gilman Dr, La Jolla, CA, 95000, USA
| | - Rosa Della Monica
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy.,CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy
| | - Davide Costabile
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy.,SEMM-European School of Molecular Medicine, University of Naples, "Federico II", 80131, Naples, Italy
| | - Michela Buonaiuto
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy.,CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy
| | - Teodolinda Di Risi
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy.,Department of Public Health, University of Naples "Federico II", Via S. Pansini, 5, 80131, Naples, Italy
| | - Giulia De Riso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy
| | - Antonella Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy
| | - Sergio Cocozza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy
| | - Roberta Visconti
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy.,Institute of Experimental Endocrinology and Oncology, Italian National Council of Research, Via S. Pansini 5, 80131, Naples, Italy
| | - Lorenzo Chiariotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131, Naples, Italy. .,CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80145, Naples, Italy. .,SEMM-European School of Molecular Medicine, University of Naples, "Federico II", 80131, Naples, Italy.
| |
Collapse
|
37
|
Zhang Y, Liu C. Evaluating the challenges and reproducibility of studies investigating DNA methylation signatures of psychological stress. Epigenomics 2022; 14:405-421. [PMID: 35170363 PMCID: PMC8978984 DOI: 10.2217/epi-2021-0190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Psychological stress can increase the risk of a wide range of negative health outcomes. Studies have been completed to determine if DNA methylation changes occur in the human brain because of stress and are associated with long-term effects and disease, but results have been inconsistent. Human candidate gene studies (150) and epigenome-wide association studies (67) were systematically evaluated to assess how DNA methylation is impacted by stress during the prenatal period, early childhood and adulthood. The association between DNA methylation of NR3C1 exon 1F and child maltreatment and early life adversity was well demonstrated, but other genes did not exhibit a clear association. The reproducibility of individual CpG sites in epigenome-wide association studies was also poor. However, biological pathways, including stress response, brain development and immunity, have been consistently identified across different stressors throughout the life span. Future studies would benefit from the increased sample size, longitudinal design, standardized methodology, optimal quality control, and improved statistical procedures.
Collapse
Affiliation(s)
- Yun Zhang
- Medical Department, Northwest Minzu University, Lanzhou, Gansu, 730000, China
- Key Laboratory of Environmental Ecology and Population Health in Northwest Minority Areas, Northwest Minzu University, Lanzhou, Gansu, 730000, China
| | - Chunyu Liu
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
38
|
Wadji DL, Tandon T, Ketcha Wanda GJM, Wicky C, Dentz A, Hasler G, Morina N, Martin-Soelch C. Child maltreatment and NR3C1 exon 1 F methylation, link with deregulated hypothalamus-pituitary-adrenal axis and psychopathology: A systematic review. CHILD ABUSE & NEGLECT 2021; 122:105304. [PMID: 34488052 DOI: 10.1016/j.chiabu.2021.105304] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 06/10/2021] [Accepted: 08/26/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Epigenetics offers one promising method for assessing the psychobiological response to stressful experiences during childhood. In particular, deoxyribonucleic acid (DNA) methylation has been associated with an altered hypothalamus-pituitary-adrenal (HPA) axis and the onset of mental disorders. Equally, there are promising leads regarding the association between the methylation of the glucocorticoid receptor gene (NR3C1-1F) and child maltreatment and its link with HPA axis and psychopathology. OBJECTIVE The current study aimed to assess the evidence of a link among child maltreatment, NR3C1-1F methylation, HPA axis deregulation, and symptoms of psychopathology. METHODS We followed the Prisma guidelines and identified 11 articles that met our inclusion criteria. RESULTS We found that eight studies (72.72%) reported increased NR3C1-1F methylation associated with child maltreatment, specifically physical abuse, emotional abuse, sexual abuse, neglect, and exposure to intimate partner violence, while three studies (27.27%) found no significant association. Furthermore, a minority of studies (36.36%) provided additional measures of symptoms of psychopathology or cortisol in order to examine the link among NR3C1-1F methylation, HPA axis deregulation, and psychopathology in a situation of child maltreatment. These results suggest that NR3C1-1F hypermethylation is positively associated with higher HPA axis activity, i.e. increased production of cortisol, as well as symptoms of psychopathology, including emotional lability-negativity, externalizing behavior symptoms, and depressive symptoms. CONCLUSION NR3C1-1F methylation could be one mechanism that links altered HPA axis activity with the development of psychopathology.
Collapse
Affiliation(s)
- D L Wadji
- I-Reach Lab, Unit of Clinical and Health Psychology, University of Fribourg, Switzerland.
| | - T Tandon
- I-Reach Lab, Unit of Clinical and Health Psychology, University of Fribourg, Switzerland
| | - G J M Ketcha Wanda
- Clinical psychology Lab, Department of Psychology, University of Yaoundé I, Cameroon
| | - C Wicky
- Department of Biology, University of Fribourg, Switzerland
| | - A Dentz
- I-Reach Lab, Unit of Clinical and Health Psychology, University of Fribourg, Switzerland
| | - G Hasler
- Department of Psychiatric, University of Fribourg, Fribourg, Switzerland
| | - N Morina
- Department of Consultant-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Switzerland
| | - C Martin-Soelch
- I-Reach Lab, Unit of Clinical and Health Psychology, University of Fribourg, Switzerland
| |
Collapse
|
39
|
Donofry SD, Stillman CM, Hanson JL, Sheridan M, Sun S, Loucks EB, Erickson KI. Promoting brain health through physical activity among adults exposed to early life adversity: Potential mechanisms and theoretical framework. Neurosci Biobehav Rev 2021; 131:688-703. [PMID: 34624365 PMCID: PMC8642290 DOI: 10.1016/j.neubiorev.2021.09.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/13/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022]
Abstract
Adverse childhood experiences such as abuse, neglect, and poverty, profoundly alter neurobehavioral development in a manner that negatively impacts health across the lifespan. Adults who have been exposed to such adversities exhibit premature and more severe age-related declines in brain health. Unfortunately, it remains unclear whether the negative effects of early life adversity (ELA) on brain health can be remediated through intervention in adulthood. Physical activity may represent a low-cost behavioral approach to address the long-term consequences of ELA on brain health. However, there has been limited research examining the impact of physical activity on brain health among adults with a history of ELA. Accordingly, the purpose of this review is to (1) review the influence of ELA on brain health in adulthood and (2) highlight evidence for the role of neurotrophic factors, hypothalamic-adrenal-pituitary axis regulation, inflammatory processes, and epigenetic modifications in mediating the effects of both ELA and physical activity on brain health outcomes in adulthood. We then propose a theoretical framework to guide future research in this area.
Collapse
Affiliation(s)
- Shannon D Donofry
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Psychiatric and Behavioral Health Institute, Allegheny Health Network Pittsburgh, PA, United States.
| | - Chelsea M Stillman
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jamie L Hanson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Learning Research and Development Center, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Margaret Sheridan
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Shufang Sun
- Department of Behavioral and Social Sciences, Brown University School of Public Health, Providence, RI, United States; Mindfulness Center, Brown University, Providence, RI, United States
| | - Eric B Loucks
- Department of Behavioral and Social Sciences, Brown University School of Public Health, Providence, RI, United States; Mindfulness Center, Brown University, Providence, RI, United States; Department of Epidemiology, Brown University School of Public Health, Providence, RI, United States
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States; Center for the Neural Basis of Cognition, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Murdoch University, College of Science, Health, Engineering, and Education, Perth, Western Australia, Australia; PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Department of Physical Education and Sports, Faculty of Sport Sciences, University of Granada, Granada, Spain
| |
Collapse
|
40
|
Epigenetic signature of chronic low back pain in human T cells. Pain Rep 2021; 6:e960. [PMID: 34746619 PMCID: PMC8568391 DOI: 10.1097/pr9.0000000000000960] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/21/2021] [Accepted: 08/05/2021] [Indexed: 12/19/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. This study reveals sex-specific DNA methylation signatures in human T cells that discriminate chronic low back pain participants from healthy controls. Objective: Determine if chronic low back pain (LBP) is associated with DNA methylation signatures in human T cells that will reveal novel mechanisms and potential therapeutic targets and explore the feasibility of epigenetic diagnostic markers for pain-related pathophysiology. Methods: Genome-wide DNA methylation analysis of 850,000 CpG sites in women and men with chronic LBP and pain-free controls was performed. T cells were isolated (discovery cohort, n = 32) and used to identify differentially methylated CpG sites, and gene ontologies and molecular pathways were identified. A polygenic DNA methylation score for LBP was generated in both women and men. Validation was performed in an independent cohort (validation cohort, n = 63) of chronic LBP and healthy controls. Results: Analysis with the discovery cohort revealed a total of 2,496 and 419 differentially methylated CpGs in women and men, respectively. In women, most of these sites were hypomethylated and enriched in genes with functions in the extracellular matrix, in the immune system (ie, cytokines), or in epigenetic processes. In men, a unique chronic LBP DNA methylation signature was identified characterized by significant enrichment for genes from the major histocompatibility complex. Sex-specific polygenic DNA methylation scores were generated to estimate the pain status of each individual and confirmed in the validation cohort using pyrosequencing. Conclusion: This study reveals sex-specific DNA methylation signatures in human T cells that discriminates chronic LBP participants from healthy controls.
Collapse
|
41
|
Chatzittofis A, Boström ADE, Ciuculete DM, Öberg KG, Arver S, Schiöth HB, Jokinen J. HPA axis dysregulation is associated with differential methylation of CpG-sites in related genes. Sci Rep 2021; 11:20134. [PMID: 34635736 PMCID: PMC8505644 DOI: 10.1038/s41598-021-99714-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/27/2021] [Indexed: 11/01/2022] Open
Abstract
DNA methylation shifts in Hypothalamic-pituitary-adrenal (HPA) axis related genes is reported in psychiatric disorders including hypersexual disorder. This study, comprising 20 dexamethasone suppression test (DST) non-suppressors and 73 controls, examined the association between the HPA axis dysregulation, shifts in DNA methylation of HPA axis related genes and importantly, gene expression. Individuals with cortisol level ≥ 138 nmol/l, after the low dose (0.5 mg) dexamethasone suppression test (DST) were classified as non-suppressors. Genome-wide methylation pattern, measured in whole blood using the EPIC BeadChip, investigated CpG sites located within 2000 bp of the transcriptional start site of key HPA axis genes, i.e.: CRH, CRHBP, CRHR-1, CRHR-2, FKBP5 and NR3C1. Regression models including DNA methylation M-values and the binary outcome (DST non-suppression status) were performed. Gene transcripts with an abundance of differentially methylated CpG sites were identified with binomial tests. Pearson correlations and robust linear regressions were performed between CpG methylation and gene expression in two independent cohorts. Six of 76 CpG sites were significantly hypermethylated in DST non-suppressors (nominal P < 0.05), associated with genes CRH, CRHR1, CRHR2, FKBP5 and NR3C1. NR3C1 transcript AJ877169 showed statistically significant abundance of probes differentially methylated by DST non-suppression status and correlated with DST cortisol levels. Further, methylation levels of cg07733851 and cg27122725 were positively correlated with gene expression levels of the NR3C1 gene. Methylation levels of cg08636224 (FKBP5) correlated with baseline cortisol and gene expression. Our findings revealed that DNA methylation shifts are involved in the altered mechanism of the HPA axis suggesting that new epigenetic targets should be considered behind psychiatric disorders.
Collapse
Affiliation(s)
- Andreas Chatzittofis
- Medical School, University of Cyprus, 1678, Nicosia, Cyprus. .,Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden.
| | - Adrian Desai E Boström
- Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden.,Neuropaediatric Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | | | - Katarina Görts Öberg
- Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Stefan Arver
- Department of Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.,Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Jussi Jokinen
- Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden.,Department of Clinical Neuroscience/Psychiatry, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Abstract
Epigenetic mechanisms such as DNA methylation (DNAm) have been associated with stress responses and increased vulnerability to depression. Abnormal DNAm is observed in stressed animals and depressed individuals. Antidepressant treatment modulates DNAm levels and regulates gene expression in diverse tissues, including the brain and the blood. Therefore, DNAm could be a potential therapeutic target in depression. Here, we reviewed the current knowledge about the involvement of DNAm in the behavioural and molecular changes associated with stress exposure and depression. We also evaluated the possible use of DNAm changes as biomarkers of depression. Finally, we discussed current knowledge limitations and future perspectives.
Collapse
|
43
|
McQuaid RJ. Transdiagnostic biomarker approaches to mental health disorders: Consideration of symptom complexity, comorbidity and context. Brain Behav Immun Health 2021; 16:100303. [PMID: 34589795 PMCID: PMC8474161 DOI: 10.1016/j.bbih.2021.100303] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/15/2021] [Accepted: 07/24/2021] [Indexed: 12/28/2022] Open
Abstract
Depression is a multifaceted disorder characterized by heterogeneous symptom profiles and high rates of comorbidity with other commonly occurring mental illnesses. Considering the burden of mental health disorders and the lack of efficacy of available treatments, there is a need for biomarkers to predict tailored or personalized treatments. However, identifying reliable biomarkers for complex mental illnesses, such as depression, anxiety and PTSD, has been challenging, likely owing to the heterogeneity, comorbidity and differences in experiences and histories of individuals. For these reasons, taking a transdiagnostic approach, which identifies biomarkers that map onto shared symptoms/constructs across disorders could be most effective for informing personalized or precision medicine approaches in psychiatry. Transdiagnostic features of anxiety, depression and anhedonia have been examined in relation to brain activity and connectivity patterns. Neuroendocrine and inflammatory markers, which are altered in depression and other comorbid illness, such as post-traumatic stress disorder (PTSD), might be useful in differentiating transdiagnostic symptom profiles as well as treatment responses. Ultimately, biomarker research that looks beyond diagnostic categories and embraces the complexity of individuals' lives and experiences might be more effective in moving towards precision medicine in psychiatry.
Collapse
Affiliation(s)
- Robyn J. McQuaid
- Carleton University, Department of Neuroscience, Ottawa, ON, Canada
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| |
Collapse
|
44
|
Motavalli R, Majidi T, Pourlak T, Abediazar S, Shoja MM, Zununi Vahed S, Etemadi J. The clinical significance of the glucocorticoid receptors: Genetics and epigenetics. J Steroid Biochem Mol Biol 2021; 213:105952. [PMID: 34274458 DOI: 10.1016/j.jsbmb.2021.105952] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/04/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022]
Abstract
The impacts of glucocorticoids (GCs) are mainly mediated by a nuclear receptor (GR) existing in almost every tissue. The GR regulates a wide range of physiological functions, including inflammation, cell metabolism, and differentiation playing a major role in cellular responses to GCs and stress. Therefore, the dysregulation or disruption of GR can cause deficiencies in the adaptation to stress and the preservation of homeostasis. The number of GR polymorphisms associated with different diseases has been mounting per year. Tackling these clinical complications obliges a comprehensive understanding of the molecular network action of GCs at the level of the GR structure and its signaling pathways. Beyond genetic variation in the GR gene, epigenetic changes can enhance our understanding of causal factors involved in the development of diseases and identifying biomarkers. In this review, we highlight the relationships of GC receptor gene polymorphisms and epigenetics with different diseases.
Collapse
Affiliation(s)
- Roza Motavalli
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taraneh Majidi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tala Pourlak
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sima Abediazar
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali M Shoja
- Clinical Academy of Teaching and Learning, Ross University School of Medicine, Miramar, FL, USA
| | | | - Jalal Etemadi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
45
|
DNA methylation of the glucocorticoid receptor gene predicts substance use in adolescence: longitudinal data from over 1000 young individuals. Transl Psychiatry 2021; 11:477. [PMID: 34526487 PMCID: PMC8443651 DOI: 10.1038/s41398-021-01601-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 02/01/2023] Open
Abstract
Early life stress has been linked to increased methylation of the Nuclear Receptor Subfamily 3 Group C Member 1 (NR3C1) gene, which codes for the glucocorticoid receptor. Moreover, early life stress has been associated with substance use initiation at a younger age, a risk factor for developing substance use disorders. However, no studies to date have investigated whether NR3C1 methylation can predict substance use in young individuals. This study included adolescents 13-14 years of age that reported no history of substance use at baseline, (N = 1041; males = 46%). Participants contributed saliva DNA samples and were followed in middle adolescence as part of KUPOL, a prospective cohort study of 7th-grade students in Sweden. Outcome variables were self-reports of (i) recent use, (ii) lifetime use, and (iii) use duration of (a) alcohol, (b) tobacco products, (c) cannabis, or (d) any substance. Outcomes were measured annually for three consecutive years. The predictor variable was DNA methylation at the exon 1 F locus of NR3C1. Risk and rate ratios were calculated as measures of association, with or without adjustment for internalizing symptoms and parental psychiatric disorders. For a subset of individuals (N = 320), there were also morning and afternoon salivary cortisol measurements available that were analyzed in relation to NR3C1 methylation levels. Baseline NR3C1 hypermethylation associated with future self-reports of recent use and use duration of any substance, before and after adjustment for potential confounders. The overall estimates were attenuated when considering lifetime use. Sex-stratified analyses revealed the strongest association for cigarette use in males. Cortisol analyses revealed associations between NR3C1 methylation and morning cortisol levels. Findings from this study suggest that saliva NR3C1 hypermethylation can predict substance use in middle adolescence. Additional longitudinal studies are warranted to confirm these findings.
Collapse
|
46
|
Roddy D, Kelly JR, Farrell C, Doolin K, Roman E, Nasa A, Frodl T, Harkin A, O'Mara S, O'Hanlon E, O'Keane V. Amygdala substructure volumes in Major Depressive Disorder. NEUROIMAGE-CLINICAL 2021; 31:102781. [PMID: 34384996 PMCID: PMC8361319 DOI: 10.1016/j.nicl.2021.102781] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/02/2021] [Accepted: 08/01/2021] [Indexed: 11/27/2022]
Abstract
The role of the amygdala in the experience of emotional states and stress is well established. Connections from the amygdala to the hypothalamus activate the hypothalamic-pituitaryadrenal (HPA) axis and the cortisol response. Previous studies have failed to find consistent whole amygdala volume changes in Major Depressive Disorder (MDD), but differences may exist at the smaller substructural level of the amygdala nuclei. High-resolution T1 and T2-weighted-fluid-attenuated inversion recovery MRIs were compared between 80 patients with MDD and 83 healthy controls (HC) using the automated amygdala substructure module in FreeSurfer 6.0. Volumetric assessments were performed for individual nuclei and three anatomico-functional composite groups of nuclei. Salivary cortisol awakening response (CAR), as a measure of HPA responsivity, was measured in a subset of patients. The right medial nucleus volume was larger in MDD compared to HC (p = 0.002). Increased right-left volume ratios were found in MDD for the whole amygdala (p = 0.004), the laterobasal composite (p = 0.009) and in the central (p = 0.003) and medial (p = 0.014) nuclei. The CAR was not significantly different between MDD and HC. Within the MDD group the left corticoamygdaloid transition area was inversely correlated with the CAR, as measured by area under the curve (AUCg) (p ≤ 0.0001). In conclusion, our study found larger right medial nuclei volumes in MDD compared to HC and relatively increased right compared to left whole and substructure volume ratios in MDD. The results suggest that amygdala substructure volumes may be involved in the pathophysiology of depression.
Collapse
Affiliation(s)
- Darren Roddy
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - John R Kelly
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland.
| | - Chloë Farrell
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Kelly Doolin
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Elena Roman
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Anurag Nasa
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas Frodl
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry and Psychotherapy, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Andrew Harkin
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Shane O'Mara
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| | - Erik O'Hanlon
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland; Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Veronica O'Keane
- Trinity College Institute of Neuroscience, Lloyd Building, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
47
|
Gu X, Ke S, Wang Q, Zhuang T, Xia C, Xu Y, Yang L, Zhou M. Energy metabolism in major depressive disorder: Recent advances from omics technologies and imaging. Biomed Pharmacother 2021; 141:111869. [PMID: 34225015 DOI: 10.1016/j.biopha.2021.111869] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/06/2021] [Accepted: 06/28/2021] [Indexed: 02/08/2023] Open
Abstract
Major depressive disorder (MDD) is a serious psychiatric disorder that associated with high rate of disability and increasing suicide rate, and the pathogenesis is still unclear. Many researches showed that the energy metabolism of patients with depression is impaired, which may be the direction of depression treatment. In this review, we focus on the "omics" technologies such as genomics, proteomics, transcriptomics and metabolomics, as well as imaging, and the progress on energy metabolism of MDD. These findings indicate that abnormal energy metabolism is one of the important mechanisms for the occurrence and development of depression. Although the research on various mechanisms of depression is still ongoing, the rapid development of new technologies and the joint use of various technologies will help to clarify the pathogenesis of depression and explore efficient diagnosis and treatment methods.
Collapse
Affiliation(s)
- Xinyi Gu
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuang Ke
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qixue Wang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tongxi Zhuang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chenyi Xia
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying Xu
- Department of Physiology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Yang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingmei Zhou
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
48
|
Womersley JS, Nothling J, Toikumo S, Malan-Müller S, van den Heuvel LL, McGregor NW, Seedat S, Hemmings SMJ. Childhood trauma, the stress response and metabolic syndrome: A focus on DNA methylation. Eur J Neurosci 2021; 55:2253-2296. [PMID: 34169602 DOI: 10.1111/ejn.15370] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/13/2021] [Accepted: 06/12/2021] [Indexed: 12/12/2022]
Abstract
Childhood trauma (CT) is well established as a potent risk factor for the development of mental disorders. However, the potential of adverse early experiences to exert chronic and profound effects on physical health, including aberrant metabolic phenotypes, has only been more recently explored. Among these consequences is metabolic syndrome (MetS), which is characterised by at least three of five related cardiometabolic traits: hypertension, insulin resistance/hyperglycaemia, raised triglycerides, low high-density lipoprotein and central obesity. The deleterious effects of CT on health outcomes may be partially attributable to dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which coordinates the response to stress, and the consequent fostering of a pro-inflammatory environment. Epigenetic tags, such as DNA methylation, which are sensitive to environmental influences provide a means whereby the effects of CT can be biologically embedded and persist into adulthood to affect health and well-being. The methylome regulates the transcription of genes involved in the stress response, metabolism and inflammation. This narrative review examines the evidence for DNA methylation in CT and MetS in order to identify shared neuroendocrine and immune correlates that may mediate the increased risk of MetS following CT exposure. Our review specifically highlights differential methylation of FKBP5, the gene that encodes FK506-binding protein 51 and has pleiotropic effects on stress responding, inflammation and energy metabolism, as a central candidate to understand the molecular aetiology underlying CT-associated MetS risk.
Collapse
Affiliation(s)
- Jacqueline S Womersley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jani Nothling
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,Gender and Health Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Sylvanus Toikumo
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stefanie Malan-Müller
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nathaniel W McGregor
- Systems Genetics Working Group, Department of Genetics, Faculty of Agriculture, Stellenbosch University, Stellenbosch, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sîan M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.,South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
49
|
Stress Modifies the Expression of Glucocorticoid-Responsive Genes by Acting at Epigenetic Levels in the Rat Prefrontal Cortex: Modulatory Activity of Lurasidone. Int J Mol Sci 2021; 22:ijms22126197. [PMID: 34201279 PMCID: PMC8228132 DOI: 10.3390/ijms22126197] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 01/09/2023] Open
Abstract
Epigenetics is one of the mechanisms by which environmental factors can alter brain function and may contribute to central nervous system disorders. Alterations of DNA methylation and miRNA expression can induce long-lasting changes in neurobiological processes. Hence, we investigated the effect of chronic stress, by employing the chronic mild stress (CMS) and the chronic restraint stress protocol, in adult male rats, on the glucocorticoid receptor (GR) function. We focused on DNA methylation specifically in the proximity of the glucocorticoid responsive element (GRE) of the GR responsive genes Gadd45β, Sgk1, and Gilz and on selected miRNA targeting these genes. Moreover, we assessed the role of the antipsychotic lurasidone in modulating these alterations. Chronic stress downregulated Gadd45β and Gilz gene expression and lurasidone normalized the Gadd45β modification. At the epigenetic level, CMS induced hypermethylation of the GRE of Gadd45β gene, an effect prevented by lurasidone treatment. These stress-induced alterations were still present even after a period of rest from stress, indicating the enduring nature of such changes. However, the contribution of miRNA to the alterations in gene expression was moderate in our experimental conditions. Our results demonstrated that chronic stress mainly affects Gadd45β expression and methylation, effects that are prolonged over time, suggesting that stress leads to changes in DNA methylation that last also after the cessation of stress procedure, and that lurasidone is a modifier of such mechanisms.
Collapse
|
50
|
Guo J, Wang J, Sun W, Liu X. The advances of post-stroke depression: 2021 update. J Neurol 2021; 269:1236-1249. [PMID: 34052887 DOI: 10.1007/s00415-021-10597-4] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022]
Abstract
Post-stroke depression (PSD) is one of common and serious sequelae of stroke. Approximately, one in three stroke survivors suffered from depression after stroke. It heavily affected functional rehabilitation, which leaded to poor quality of life. What is worse, it is strongly associated with high mortality. In this review, we aimed to derive a comprehensive and integrated understanding of PSD according to recently published papers and previous classic articles. Based on the considerable number of studies, we found that within 2 years incidence of PSD has a range from 11 to 41%. Many factors contribute to the occurrence of PSD, including the history of depression, stroke severity, lesion location, and so on. Currently, the diagnosis of PSD is mainly based on the DSM guidelines and combined with various depression scales. Unfortunately, we lack a unified mechanism to explain PSD which mechanisms now involve dysregulation of hypothalamic-pituitary-adrenal (HPA) axis, increased inflammatory factors, decreased levels of monoamines, glutamate-mediated excitotoxicity, and abnormal neurotrophic response. At present, both pharmacotherapy and psychological therapies are employed in treating PSD. Although great advance has been made by researchers, there are still a lot of issues need to be addressed. Especially, the mechanism of PSD is not completely clear.
Collapse
Affiliation(s)
- Jianglong Guo
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, People's Republic of China
| | - Jinjing Wang
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wen Sun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, People's Republic of China
| | - Xinfeng Liu
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, People's Republic of China.
- Department of Neurology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|