1
|
Tonna M, Borrelli DF, Marchesi C, Gerra MC, Dallabona C. Childhood obsessive-compulsive disorder, epigenetics, and heterochrony: An evolutionary and developmental approach. Dev Psychopathol 2025:1-15. [PMID: 40099440 DOI: 10.1017/s0954579425000124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Childhood obsessive-compulsive disorder (OCD) stems from a bunch of restricted and repetitive behaviors, which are part of normal behavioral repertoire up to the age of 7. The persistence of compulsive-like behaviors after that age is often associated with unique comorbidity patterns, which are age-at-onset dependent and reflect different developmental stages. In particular, OCD synchronically co-occurs with a broad constellation of neurodevelopmental disorders, whereas diachronically it is related to an increased risk of major adult psychoses. Moreover, OCD is associated with trait-like sensory phenomena, suggesting a common disrupted sensorimotor grounding.The present study is aimed at exploring the hypothesis that this specific temporal and comorbidity OCD profile may be due to a developmental heterochronic mechanism of delay in attenuation of ontogenetically early behavioral patterns. The developmental shift of highly evolutionarily conserved behavioral phenotypes might be regulated by epigenetic changes induced by different conditions of sensory unbalance. This evolutionary and developmental model allows capturing childhood OCD in light of the ultimate causes of ritual behavior throughout phylogeny, namely its "homeostatic" function over conditions of unpredictability. Moreover, it may have important clinical implications, as OCD symptoms could represent putative biomarkers of early divergent developmental trajectories, with a pathoplastic effect on course and outcome.
Collapse
Affiliation(s)
- Matteo Tonna
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Davide Fausto Borrelli
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Piacenza, Italy
| | - Carlo Marchesi
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Maria Carla Gerra
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| |
Collapse
|
2
|
Aljabali AAA, Alkaraki AK, Gammoh O, Tambuwala MM, Mishra V, Mishra Y, Hassan SS, El-Tanani M. Deciphering Depression: Epigenetic Mechanisms and Treatment Strategies. BIOLOGY 2024; 13:638. [PMID: 39194576 DOI: 10.3390/biology13080638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Depression, a significant mental health disorder, is under intense research scrutiny to uncover its molecular foundations. Epigenetics, which focuses on controlling gene expression without altering DNA sequences, offers promising avenues for innovative treatment. This review explores the pivotal role of epigenetics in depression, emphasizing two key aspects: (I) identifying epigenetic targets for new antidepressants and (II) using personalized medicine based on distinct epigenetic profiles, highlighting potential epigenetic focal points such as DNA methylation, histone structure alterations, and non-coding RNA molecules such as miRNAs. Variations in DNA methylation in individuals with depression provide opportunities to target genes that are associated with neuroplasticity and synaptic activity. Aberrant histone acetylation may indicate that antidepressant strategies involve enzyme modifications. Modulating miRNA levels can reshape depression-linked gene expression. The second section discusses personalized medicine based on epigenetic profiles. Analyzing these patterns could identify biomarkers associated with treatment response and susceptibility to depression, facilitating tailored treatments and proactive mental health care. Addressing ethical concerns regarding epigenetic information, such as privacy and stigmatization, is crucial in understanding the biological basis of depression. Therefore, researchers must consider these issues when examining the role of epigenetics in mental health disorders. The importance of epigenetics in depression is a critical aspect of modern medical research. These findings hold great potential for novel antidepressant medications and personalized treatments, which would significantly improve patient outcomes, and transform psychiatry. As research progresses, it is expected to uncover more complex aspects of epigenetic processes associated with depression, enhance our comprehension, and increase the effectiveness of therapies.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan
| | - Almuthanna K Alkaraki
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan
| | - Murtaza M Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur 721140, West Bengal, India
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates
| |
Collapse
|
3
|
Winter JJ, Rodríguez-Acevedo KL, Dittrich M, Heller EA. Early life adversity: Epigenetic regulation underlying drug addiction susceptibility. Mol Cell Neurosci 2023; 125:103825. [PMID: 36842544 PMCID: PMC10247461 DOI: 10.1016/j.mcn.2023.103825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/28/2023] Open
Abstract
Drug addiction is a leading cause of disability worldwide, with more than 70,000 Americans dying from drug overdose in 2019 alone. While only a small percentage of chronic drug users escalate to drug addiction, little is understood on the precise mechanisms of this susceptibility. Early life adversity is causally relevant to adult psychiatric disease and may contribute to the risk of addiction. Here we review recent pre-clinical evidence showing that early life exposure to stress and/or drugs regulates changes in behavior, gene expression, and the epigenome that persist into adulthood. We summarize the major findings and gaps in the preclinical literature, highlighting studies that demonstrate the often profound differences between female and male subjects.
Collapse
Affiliation(s)
| | | | - Mia Dittrich
- University of Pennsylvania, Philadelphia, PA 19106, USA
| | | |
Collapse
|
4
|
Kruyer A, Kalivas PW, Scofield MD. Astrocyte regulation of synaptic signaling in psychiatric disorders. Neuropsychopharmacology 2023; 48:21-36. [PMID: 35577914 PMCID: PMC9700696 DOI: 10.1038/s41386-022-01338-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 02/07/2023]
Abstract
Over the last 15 years, the field of neuroscience has evolved toward recognizing the critical role of astroglia in shaping neuronal synaptic activity and along with the pre- and postsynapse is now considered an equal partner in tripartite synaptic transmission and plasticity. The relative youth of this recognition and a corresponding deficit in reagents and technologies for quantifying and manipulating astroglia relative to neurons continues to hamper advances in understanding tripartite synaptic physiology. Nonetheless, substantial advances have been made and are reviewed herein. We review the role of astroglia in synaptic function and regulation of behavior with an eye on how tripartite synapses figure into brain pathologies underlying behavioral impairments in psychiatric disorders, both from the perspective of measures in postmortem human brains and more subtle influences on tripartite synaptic regulation of behavior in animal models of psychiatric symptoms. Our goal is to provide the reader a well-referenced state-of-the-art understanding of current knowledge and predict what we may discover with deeper investigation of tripartite synapses using reagents and technologies not yet available.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA.
- Department of Anesthesia & Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
5
|
Lewis CR, Tafur J, Spencer S, Green JM, Harrison C, Kelmendi B, Rabin DM, Yehuda R, Yazar-Klosinski B, Cahn BR. Pilot study suggests DNA methylation of the glucocorticoid receptor gene (NR3C1) is associated with MDMA-assisted therapy treatment response for severe PTSD. Front Psychiatry 2023; 14:959590. [PMID: 36815187 PMCID: PMC9939628 DOI: 10.3389/fpsyt.2023.959590] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Background Previous research has demonstrated that epigenetic changes in specific hypothalamic-pituitary-adrenal (HPA) genes may predict successful psychotherapy in post-traumatic stress disorder (PTSD). A recent Phase 3 clinical trial reported high efficacy of 3,4-methylenedioxymethamphetamine (MDMA)-assisted therapy for treating patients with severe PTSD compared to a therapy with placebo group (NCT03537014). This raises important questions regarding potential mechanisms of MDMA-assisted therapy. In the present study, we examined epigenetic changes in three key HPA axis genes before and after MDMA and placebo with therapy. As a pilot sub-study to the parent clinical trial, we assessed potential HPA epigenetic predictors for treatment response with genomic DNA derived from saliva (MDMA, n = 16; placebo, n = 7). Methylation levels at all 259 CpG sites annotated to three HPA genes (CRHR1, FKBP5, and NR3C1) were assessed in relation to treatment response as measured by the Clinician-Administered PTSD Scale (CAPS-5; Total Severity Score). Second, group (MDMA vs. placebo) differences in methylation change were assessed for sites that predicted treatment response. Results Methylation change across groups significantly predicted symptom reduction on 37 of 259 CpG sites tested, with two sites surviving false discovery rate (FDR) correction. Further, the MDMA-treatment group showed more methylation change compared to placebo on one site of the NR3C1 gene. Conclusion The findings of this study suggest that therapy-related PTSD symptom improvements may be related to DNA methylation changes in HPA genes and such changes may be greater in those receiving MDMA-assisted therapy. These findings can be used to generate hypothesis driven analyses for future studies with larger cohorts.
Collapse
Affiliation(s)
- Candace R. Lewis
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
- *Correspondence: Candace R. Lewis,
| | | | - Sophie Spencer
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Joseph M. Green
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Benjamin Kelmendi
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States
| | | | - Rachel Yehuda
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Psychiatry, James J. Peters VA Medical Center, Bronx, NY, United States
| | | | - Baruch Rael Cahn
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, United States
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA, United States
- Baruch Rael Cahn,
| |
Collapse
|
6
|
Dissecting early life stress-induced adolescent depression through epigenomic approach. Mol Psychiatry 2023; 28:141-153. [PMID: 36517640 PMCID: PMC9812796 DOI: 10.1038/s41380-022-01907-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022]
Abstract
Early life stress (ELS), such as abuse and neglect during childhood, can lead to psychiatric disorders in later life. Previous studies have suggested that ELS can cause profound changes in gene expression through epigenetic mechanisms, which can lead to psychiatric disorders in adulthood; however, studies on epigenetic modifications associated with ELS and psychiatric disorders in adolescents are limited. Moreover, how these epigenetic modifications can lead to psychiatric disorders in adolescents is not fully understood. Commonly, DNA methylation, histone modification, and the regulation of noncoding RNAs have been attributed to the reprogramming of epigenetic profiling associated with ELS. Although only a few studies have attempted to examine epigenetic modifications in adolescents with ELS, existing evidence suggests that there are commonalities and differences in epigenetic profiling between adolescents and adults. In addition, epigenetic modifications are sex-dependent and are influenced by the type of ELS. In this review, we have critically evaluated the current evidence on epigenetic modifications in adolescents with ELS, particularly DNA methylation and the expression of microRNAs in both preclinical models and humans. We have also clarified the impact of ELS on psychiatric disorders in adolescents to predict the development of neuropsychiatric disorders and to prevent and recover these disorders through personalized medicine.
Collapse
|
7
|
Cell-type-specific epigenetic effects of early life stress on the brain. Transl Psychiatry 2022; 12:326. [PMID: 35948532 PMCID: PMC9365848 DOI: 10.1038/s41398-022-02076-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
Early life stress (ELS) induces long-term phenotypic adaptations that contribute to increased vulnerability to a host of neuropsychiatric disorders. Epigenetic mechanisms, including DNA methylation, histone modifications and non-coding RNA, are a proposed link between environmental stressors, alterations in gene expression, and phenotypes. Epigenetic modifications play a primary role in shaping functional differences between cell types and can be modified by environmental perturbations, especially in early development. Together with contributions from genetic variation, epigenetic mechanisms orchestrate patterns of gene expression within specific cell types that contribute to phenotypic variation between individuals. To date, many studies have provided insights into epigenetic changes resulting from ELS. However, most of these studies have examined heterogenous brain tissue, despite evidence of cell-type-specific epigenetic modifications in phenotypes associated with ELS. In this review, we focus on rodent and human studies that have examined epigenetic modifications induced by ELS in select cell types isolated from the brain or associated with genes that have cell-type-restricted expression in neurons, microglia, astrocytes, and oligodendrocytes. Although significant challenges remain, future studies using these approaches can enable important mechanistic insight into the role of epigenetic variation in the effects of ELS on brain function.
Collapse
|
8
|
Cahill S, Chandola T, Hager R. Genetic Variants Associated With Resilience in Human and Animal Studies. Front Psychiatry 2022; 13:840120. [PMID: 35669264 PMCID: PMC9163442 DOI: 10.3389/fpsyt.2022.840120] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Resilience is broadly defined as the ability to maintain or regain functioning in the face of adversity and is influenced by both environmental and genetic factors. The identification of specific genetic factors and their biological pathways underpinning resilient functioning can help in the identification of common key factors, but heterogeneities in the operationalisation of resilience have hampered advances. We conducted a systematic review of genetic variants associated with resilience to enable the identification of general resilience mechanisms. We adopted broad inclusion criteria for the definition of resilience to capture both human and animal model studies, which use a wide range of resilience definitions and measure very different outcomes. Analyzing 158 studies, we found 71 candidate genes associated with resilience. OPRM1 (Opioid receptor mu 1), NPY (neuropeptide Y), CACNA1C (calcium voltage-gated channel subunit alpha1 C), DCC (deleted in colorectal carcinoma), and FKBP5 (FKBP prolyl isomerase 5) had both animal and human variants associated with resilience, supporting the idea of shared biological pathways. Further, for OPRM1, OXTR (oxytocin receptor), CRHR1 (corticotropin-releasing hormone receptor 1), COMT (catechol-O-methyltransferase), BDNF (brain-derived neurotrophic factor), APOE (apolipoprotein E), and SLC6A4 (solute carrier family 6 member 4), the same allele was associated with resilience across divergent resilience definitions, which suggests these genes may therefore provide a starting point for further research examining commonality in resilience pathways.
Collapse
Affiliation(s)
- Stephanie Cahill
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
| | - Tarani Chandola
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
- Methods Hub, Department of Sociology, Faculty of Social Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Reinmar Hager
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Luo L, van der Zande LE, van Marwijk MA, Knol EF, Rodenburg TB, Bolhuis JE, Parois SP. Impact of Enrichment and Repeated Mixing on Resilience in Pigs. Front Vet Sci 2022; 9:829060. [PMID: 35400108 PMCID: PMC8988148 DOI: 10.3389/fvets.2022.829060] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 02/24/2022] [Indexed: 12/20/2022] Open
Abstract
Resilience, the capacity of animals to be minimally affected by a disturbance or to rapidly bounce back to the state before the challenge, may be improved by enrichment, but negatively impacted by a high allostatic load from stressful management procedures in pigs. We investigated the combined effects of diverging environmental conditions from weaning and repeated mixing to create high allostatic load on resilience of pigs. Pigs were either exposed to barren housing conditions (B) from weaning onwards or provided with sawdust, extra toys, regular access to a “play arena” and daily positive human contact (E). Half of the pigs were exposed to repeated mixing (RM) and the other half to one mixing only at weaning (minimal mixing, MM). To assess their resilience, the response to and recovery from a lipopolysaccharide (LPS) sickness challenge and a Frustration challenge were studied. In addition, potential long-term resilience indicators, i.e. natural antibodies, hair cortisol and growth were measured. Some indications of more favorable responses to the challenges in E pigs were found, such as lower serum reactive oxygen metabolite (dROM) concentrations and a smaller area under the curve of dROM after LPS injection. In the Frustration challenge, E pigs showed less standing alert, escape behaviors and other negative behaviors, a tendency for a smaller area under the curve of salivary cortisol and a lower plasma cortisol level at 1 h after the challenge. Aggression did not decrease over mixings in RM pigs and was higher in B pigs than in E pigs. Repeated mixing did not seem to reduce resilience. Contrary to expectations, RM pigs showed a higher relative growth than MM pigs during the experiment, especially in the week of the challenges. Barren RM pigs showed a lower plasma cortisol concentration than barren MM pigs after the LPS challenge, which may suggest that those RM pigs responded less detrimentally than MM pigs. Enriched RM pigs showed a higher level of IgM antibodies binding keyhole limpet hemocyanin (KLH) than enriched MM and barren RM pigs, and RM pigs showed a sharper decline in IgG antibodies binding Bovine Serum Albumin (PC-BSA) over time than MM pigs. Hair cortisol concentrations were not affected by enrichment or mixing. To conclude, enrichment did not enhance the speed of recovery from challenges in pigs, although there were indications of reduced stress. Repeated as opposed to single mixing did not seem to aggravate the negative effects of barren housing on resilience and for some parameters even seemed to reduce the negative effects of barren housing.
Collapse
Affiliation(s)
- Lu Luo
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Lisette E. van der Zande
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | - Manon A. van Marwijk
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
| | | | - T. Bas Rodenburg
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
- Animals in Science and Society, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - J. Elizabeth Bolhuis
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
- *Correspondence: J. Elizabeth Bolhuis
| | - Severine P. Parois
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University & Research, Wageningen, Netherlands
- PEGASE, INRAE, Institut Agro, Saint-Gilles, France
- Epidemiology Health and Welfare Research Unit, Ploufragan-Plouzané-Niort Laboratory, French Agency for Food, Environmental and Occupational Health and Safety (ANSES), Ploufragan, France
| |
Collapse
|
10
|
Joo YY, Moon SY, Wang HH, Kim H, Lee EJ, Kim JH, Posner J, Ahn WY, Choi I, Kim JW, Cha J. Association of Genome-Wide Polygenic Scores for Multiple Psychiatric and Common Traits in Preadolescent Youths at Risk of Suicide. JAMA Netw Open 2022; 5:e2148585. [PMID: 35188556 PMCID: PMC8861848 DOI: 10.1001/jamanetworkopen.2021.48585] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
IMPORTANCE Suicide is the second leading cause of death among youths worldwide, but no available means exist to identify the risk of suicide in this population. OBJECTIVE To assess whether genome-wide polygenic scores for psychiatric and common traits are associated with the risk of suicide among preadolescent children and to investigate whether and to what extent the interaction between early life stress (a major environmental risk factor) and polygenic factors is associated with suicidal thoughts and behaviors among youths. DESIGN, SETTING, AND PARTICIPANTS This cohort study analyzed the genotype-phenotype data of 11 869 preadolescent children aged 9 to 10 years from the Adolescent Brain and Cognitive Development study. Data were collected from September 1, 2016, to October 21, 2018, and analyzed from August 1, 2020, to January 3, 2021. Using machine learning approaches, genome-wide polygenic scores of 24 complex traits were estimated to investigate their phenome-wide associations and utility for assessing risk of suicidal thoughts and behaviors (suicidal ideation [active, passive, and overall] and suicide attempt). MAIN OUTCOMES AND MEASURES Genome-wide polygenic scores were used to measure 24 traits, including psychiatric disorders, cognitive capacity, and personality and psychological characteristics. The Child Behavior Checklist was used to measure early life stress, and the Family Environment Scale was used to assess family environment. Suicidal ideation and suicide attempts were derived from the computerized version of the Kiddie Schedule for Affective Disorders and Schizophrenia. RESULTS Among 11 869 preadolescent children in the US, complete data for phenotypic outcomes, genotypes, and covariates were available for 7140 participants in the multiethnic cohort (mean [SD] age, 9.9 [0.6] years; 3588 girls [50.3%]), including 925 participants with suicidal ideation and 63 participants with suicide attempts. Among those 7140 participants, 729 had African ancestry (self-reported race or ethnicity: 569 Black, 71 Hispanic, and 89 other), 276 had admixed American ancestry (self-reported race or ethnicity: 265 Hispanic, 3 White, and 8 other), 150 had East Asian ancestry (self-reported race or ethnicity: 67 Asian, 18 Hispanic, and 65 other), 5718 had European ancestry (self-reported race or ethnicity: 7 Asian, 39 Black, 1142 Hispanic, 3934 White, and 596 other), and 267 had other ancestries (self-reported race or ethnicity: 70 Asian, 13 Black, 126 Hispanic, 48 White, and 10 other). Three genome-wide polygenic scores were significantly associated (false discovery rate P < .05) with suicidal thoughts and behaviors among all participants: attention-deficit/hyperactivity disorder (odds ratio [OR], 1.12; 95% CI, 1.05-1.21; P = .001), schizophrenia (OR, 1.50; 95% CI, 1.17-1.93; P = .002), and general happiness (OR, 0.89; 95% CI, 0.83-0.96; P = .002). In the analysis including only children with European ancestry, 3 additional genome-wide polygenic scores with false discovery rate significance were associated with suicidal thoughts and behaviors: autism spectrum disorder (OR, 1.18; 95% CI, 1.06-1.31; P = .002), major depressive disorder (OR, 1.12; 95% CI, 1.04-1.21; P = .003), and posttraumatic stress disorder (OR, 1.12; 95% CI, 1.04-1.21; P = .004). A significant interaction between genome-wide polygenic scores and environment was found, with genetic risk factors for autism spectrum disorder and the level of early life stress associated with increases in the risk of overall suicidal ideation and overall suicidal thoughts and behaviors (OR, 1.20; 95% CI, 1.07-1.35; P = .002). A machine learning model using multitrait genome-wide polygenic scores and additional self-reported questionnaire data (Child Behavior Checklist and Family Environment Scale) produced a moderately accurate estimate of overall suicidal thoughts and behaviors (area under the receiver operating characteristic curve [AUROC], 0.77; 95% CI, 0.73-0.81; accuracy, 0.67) and suicidal ideation (AUROC, 0.76; 95% CI, 0.72-0.80; accuracy, 0.66) among children with European ancestry only. Among all children in the multiethnic cohort, the integrated model also outperformed the baseline model in estimating the risk of overall suicidal thoughts and behaviors (AUROC, 0.71; 95% CI, 0.67-0.75; accuracy, 0.68) and suicidal ideation (AUROC, 0.75; 95% CI, 0.71-0.78; accuracy, 0.67). CONCLUSIONS AND RELEVANCE In this cohort study of preadolescent youths in the US, higher genome-wide polygenic scores for psychiatric disorders, such as attention-deficit/hyperactivity disorder, autism spectrum disorder, posttraumatic stress disorder, and schizophrenia, were significantly associated with a greater risk of suicidal ideation and suicide attempt. The findings and quantitative models from this study may help to identify children with a high risk of suicide, potentially assisting with early screening, intervention, and prevention.
Collapse
Affiliation(s)
- Yoonjung Yoonie Joo
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
- Institute of Data Science, Korea University, Seoul, Republic of Korea
| | - Seo-Yoon Moon
- College of Liberal Studies, Seoul National University, Seoul, Republic of Korea
| | - Hee-Hwan Wang
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyeonjin Kim
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
| | - Eun-Ji Lee
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
| | - Jong Hun Kim
- Department of Neurology, Dementia Center, Stroke Center, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Jonathan Posner
- Department of Psychiatry, Columbia University Medical Center, New York, New York
| | - Woo-Young Ahn
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
| | - Incheol Choi
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
| | - Jae-Won Kim
- Department of Child and Adolescent Psychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jiook Cha
- Department of Psychology, Seoul National University, Seoul, Republic of Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Republic of Korea
- Artificial Intelligence Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Gerra ML, Gerra MC, Tadonio L, Pellegrini P, Marchesi C, Mattfeld E, Gerra G, Ossola P. Early parent-child interactions and substance use disorder: An attachment perspective on a biopsychosocial entanglement. Neurosci Biobehav Rev 2021; 131:560-580. [PMID: 34606823 DOI: 10.1016/j.neubiorev.2021.09.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022]
Abstract
This review aims to elucidate environmental and genetic factors, as well as their epigenetic and neuroendocrine moderators, that may underlie the association between early childhood experiences and Substance Use Disorders (SUD), through the lens of parental attachment. Here we review those attachment-related studies that examined the monoaminergic systems, the hypothalamic pituitary adrenal stress response system, the oxytoninergic system, and the endogenous opioid system from a genetic, epigenetic, and neuroendocrine perspective. Overall, the selected studies point to a moderating effect of insecure attachment between genetic vulnerability and SUD, reasonably through epigenetic modifications. Preliminary evidence suggests that vulnerability to SUDs is related with hypo-methylation (e.g. hyper-expression) of high-risk polymorphisms on the monoaminergic and hypothalamic pituitary adrenal system and hyper-methylation (e.g. hypo-expressions) of protective polymorphisms on the opioid and oxytocin system. These epigenetic modifications may induce a cascade of neuroendocrine changes contributing to the subclinical and behavioural manifestations that precede the clinical onset of SUD. Protective and supportive parenting could hence represent a key therapeutic target to prevent addiction and moderate insecure attachment.
Collapse
Affiliation(s)
| | - Maria Carla Gerra
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| | | | | | - Carlo Marchesi
- Psychiatry Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Elizabeth Mattfeld
- Drug Prevention and Health Branch, Prevention Treatment and Rehabilitation Section, United Nations Office on Drugs and Crime, Vienna, Austria.
| | - Gilberto Gerra
- Department of Mental Health, AUSL of Parma, Parma, Italy.
| | - Paolo Ossola
- Center for Neuroplasticity and Pain (CNAP), SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
12
|
Li S, Wang W, Zhang D, Li W, Lund J, Kruse T, Mengel-From J, Christensen K, Tan Q. Differential regulation of the DNA methylome in adults born during the Great Chinese Famine in 1959-1961. Genomics 2021; 113:3907-3918. [PMID: 34600028 DOI: 10.1016/j.ygeno.2021.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/24/2021] [Accepted: 09/25/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Extensive epidemiological studies have established the association between exposure to early-life adversity and health status and diseases in adults. Epigenetic regulation is considered as a key mediator for this phenomenon but analysis on humans is sparse. The Great Chinese Famine lasting from 1958 to 1961 is a natural string of disasters offering a precious opportunity for elucidating the underlying epigenetic mechanism of the long-term effect of early adversity. METHODS Using a high-throughput array platform for DNA methylome profiling, we conducted a case-control epigenome-wide association study on early-life exposure to Chinese famine in 79 adults born during 1959-1961 and compared to 105 unexposed subjects born 1963-1964. RESULTS The single CpG site analysis of whole epigenome revealed a predominant pattern of decreased DNA methylation levels associated with fetal exposure to famine. Four CpG sites were detected with p < 1e-06 (linked to EHMT1, CNR1, UBXN7 and ESM1 genes), 16 CpGs detected with 1e-06 < p < 1e-05 and 157 CpGs with 1e-05 < p < 1e-04, with a predominant pattern of hypomethylation. Functional annotation to genes and their enriched biological pathways mainly involved neurodevelopment, neuropsychological disorders and metabolism. Multiple sites analysis detected two top-rank differentially methylated regions harboring RNF39 on chromosome 6 and PTPRN2 on chromosome 7, both showing epigenetic association with stress-related conditions. CONCLUSION Early-life exposure to famine could mediate DNA methylation regulations that persist into adulthood with broad impacts in the activities of genes and biological pathways. Results from this study provide new clues to the epigenetic embedding of early-life adversity and its impacts on adult health.
Collapse
Affiliation(s)
- Shuxia Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Weijing Wang
- Qingdao University School of Public Health, Qingdao, China
| | - Dongfeng Zhang
- Qingdao University School of Public Health, Qingdao, China
| | - Weilong Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark; Population Research Unit, Faculty of Social Sciences, University of Helsinki, Finland.
| | - Jesper Lund
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark; Digital Health & Machine Learning Research Group, Hasso Plattner Institute for Digital Engineering, Potsdam, Germany.
| | - Torben Kruse
- Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Jonas Mengel-From
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Kaare Christensen
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark.
| | - Qihua Tan
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, Odense, Denmark; Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
13
|
Lewis CR, Breitenstein RS, Henderson A, Sowards HA, Piras IS, Huentelman MJ, Doane LD, Lemery-Chalfant K. Harsh Parenting Predicts Novel HPA Receptor Gene Methylation and NR3C1 Methylation Predicts Cortisol Daily Slope in Middle Childhood. Cell Mol Neurobiol 2021; 41:783-793. [PMID: 32472381 PMCID: PMC11448560 DOI: 10.1007/s10571-020-00885-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/23/2020] [Indexed: 01/16/2023]
Abstract
Adverse experiences in childhood are associated with altered hypothalamic-pituitary-adrenal (HPA) axis function and negative health outcomes throughout life. It is now commonly accepted that abuse and neglect can alter epigenetic regulation of HPA genes. Accumulated evidence suggests harsh parenting practices such as spanking are also strong predictors of negative health outcomes. We predicted harsh parenting at 2.5 years old would predict HPA gene DNA methylation similarly to abuse and neglect, and cortisol output at 8.5 years old. Saliva samples were collected three times a day across 3 days to estimate cortisol diurnal slopes. Methylation was quantified using the Illumina Infinium MethylationEPIC array BeadChip (850 K) with DNA collected from buccal cells. We used principal components analysis to compute a summary statistic for CpG sites across candidate genes. The first and second components were used as outcome variables in mixed linear regression analyses with harsh parenting as a predictor variable. We found harsh parenting significantly predicted methylation of several HPA axis genes, including novel gene associations with AVPRB1, CRHR1, CRHR2, and MC2R (FDR corrected p < 0.05). Further, we found NR3C1 methylation predicted a steeper diurnal cortisol slope. Our results extend the current literature by demonstrating harsh parenting may influence DNA methylation similarly to more extreme early life experiences such as abuse and neglect. Further, we show NR3C1 methylation is associated with diurnal HPA function. Elucidating the molecular consequences of harsh parenting on health can inform best parenting practices and provide potential treatment targets for common complex disorders.
Collapse
Affiliation(s)
- Candace R Lewis
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA.
- Psychology, Arizona State University, Tempe, AZ, USA.
| | | | - Adrienne Henderson
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | | | - Ignazio S Piras
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Matthew J Huentelman
- Neurogenomics, Translational Genomics Research Institute, 445 N 5th St., Phoenix, AZ, 85004, USA
| | - Leah D Doane
- Psychology, Arizona State University, Tempe, AZ, USA
| | | |
Collapse
|
14
|
Preadult polytoxicomania-strong environmental underpinnings and first genetic hints. Mol Psychiatry 2021; 26:3211-3222. [PMID: 33824432 PMCID: PMC8505259 DOI: 10.1038/s41380-021-01069-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 11/09/2022]
Abstract
Considering the immense societal and personal costs and suffering associated with multiple drug use or "polytoxicomania", better understanding of environmental and genetic causes is crucial. While previous studies focused on single risk factors and selected drugs, effects of early-accumulated environmental risks on polytoxicomania were never addressed. Similarly, evidence of genetic susceptibility to particular drugs is abundant, while genetic predisposition to polytoxicomania is unexplored. We exploited the GRAS data collection, comprising information on N~2000 deep-phenotyped schizophrenia patients, to investigate effects of early-life environmental risk accumulation on polytoxicomania and additionally provide first genetic insight. Preadult accumulation of environmental risks (physical or sexual abuse, urbanicity, migration, cannabis, alcohol) was strongly associated with lifetime polytoxicomania (p = 1.5 × 10-45; OR = 31.4), preadult polytoxicomania with OR = 226.6 (p = 1.0 × 10-33) and adult polytoxicomania with OR = 17.5 (p = 3.4 × 10-24). Parallel accessibility of genetic data from GRAS patients and N~2100 controls for genome-wide association (GWAS) and phenotype-based genetic association studies (PGAS) permitted the creation of a novel multiple GWAS-PGAS approach. This approach yielded 41 intuitively interesting SNPs, potentially conferring liability to preadult polytoxicomania, which await replication upon availability of suitable deep-phenotyped cohorts anywhere world-wide. Concisely, juvenile environmental risk accumulation, including cannabis and alcohol as starter/gateway drugs, strongly predicts polytoxicomania during adolescence and adulthood. This pivotal message should launch more effective sociopolitical measures to prevent this deleterious psychiatric condition.
Collapse
|
15
|
Uselman TW, Barto DR, Jacobs RE, Bearer EL. Evolution of brain-wide activity in the awake behaving mouse after acute fear by longitudinal manganese-enhanced MRI. Neuroimage 2020; 222:116975. [PMID: 32474079 PMCID: PMC7805483 DOI: 10.1016/j.neuroimage.2020.116975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
Life threatening fear after a single exposure evolves in a subset of vulnerable individuals to anxiety, which may persist for their lifetime. Yet neither the whole brain's response to innate acute fear nor how brain activity evolves over time is known. Sustained neuronal activity may be a factor in the development of a persistent fear response. We couple two experimental protocols to provoke acute fear leading to prolonged fear: Predator stress (PS), a naturalistic approach to induce fear in rodents; and Serotonin transporter knockout mouse (SERT-KO) that responds to PS with sustained defensive behavior. Behavior was monitored before, during and at short and long times after PS in wild type (WT) and SERT-KO mice. Both genotypes responded to PS with defensive behavior. SERT-KO retained defensive behavior for 23 days, while WT mice returned to baseline exploratory behavior by 9 days. Thus, differences in neural activity between WT and SERT-KO 9 days after PS identifies neural correlates of persistent defensive behavior, in mice. We used longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) to identify brain-wide neural activity associated with different behaviors. Mn2+ accumulation in active neurons occurs in awake, behaving mice and is retrospectively imaged. Following the same two cohorts of mice, WT and SERT-KO, longitudinally allowed unbiased quantitative comparisons of brain-wide activity by statistical parametric mapping (SPM). During natural behavior in WT, only low levels of activity-induced Mn2+-accumulation were detected, while much more accumulation appeared immediately after PS in both WT and SERT-KO, and evolved at 9 days to a new activity pattern (p < 0.0001, uncorr., T = 5.4). Patterns of accumulation differed between genotypes, with more regions of the brain and larger volumes within regions involved in SERT-KO than WT. A new computational segmentation analysis, using our InVivo Atlas based on a manganese-enhanced MR image of a living mouse, revealed dynamic changes in the volume of significantly enhanced voxels within each segment that differed between genotypes across 45 of 87 segmented regions. At Day 9 after PS, the striatum and ventral pallidum were active in both genotypes but more so in the SERT-KO. SERT-KO also displayed sustained or increased volume of Mn2+ accumulations between Post-Fear and Day 9 in eight segments where activity was decreased or silenced in WT. C-fos staining, an alternative neural activity marker, of brains from the same mice fixed at conclusion of imaging sessions confirmed that MEMRI detected active neurons. Intensity measurements in 12 regions of interest (ROIs) supported the SPM results. Between group comparisons by SPM and of ROI measurements identified specific regions differing between time points and genotypes. We report brain-wide activity in response to a single exposure of acute fear, and, for the first time, its evolution to new activity patterns over time in individuals vulnerable to persistent fear. Our results show multiple regions with dynamic changes in neural activity and that the balance of activity between segments is disordered in the SERT-KO. Thus, longitudinal MEMRI represents a powerful approach to discover how brain-wide activity evolves from the natural state either after an experience or during a disease process.
Collapse
Affiliation(s)
- Taylor W Uselman
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Daniel R Barto
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Russell E Jacobs
- Zilkha Neurogenetics Institute, University of Southern California, Los Angeles, CA, USA; California Institute of Technology, Pasadena, CA, USA
| | - Elaine L Bearer
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
16
|
Cocaine-induced inheritable epigenetic marks may be altered by changing early postnatal fostering. Neuroreport 2020; 30:1157-1165. [PMID: 31568187 DOI: 10.1097/wnr.0000000000001332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Here, we explored the hypothesis that parental cocaine exposure could alter epigenetic machinery in their drug-naive offspring while early postnatal fostering may further modify the accompanied neurochemical and functional components. Variant drug-naive pups were produced from cocaine-exposed or unexposed C57BL/6 female mice that were matched with their male counterparts for mating. Within 3 days of birth, half of the pups were cross-fostered and nurtured by non-biological lactating dams. The pups were initially examined for locomotor activity and memory performance and subsequently for changes in DNA methylation in promoter regions of cAMP response element modulator (Crem) and Fosb in the prefrontal cortex at 48 days postnatum. The impact of postnatal fostering on these parameters was also investigated. Our results showed that cocaine exposure significantly decreased both Crem and Fosb methylation in the prefrontal cortex of progenitor mice, while similar patterns of methylation were replicated in the brains of drug-naive non-fostered offspring mice but reversed by postnatal fostering. Furthermore, offspring raised by cocaine-exposed dams were impaired in discriminative learning and exhibited memory decline, whereas locomotor activity remains unaltered in all groups of mice. Our data provide some evidence that indirect exposure to cocaine may cause marked epigenetic changes within the cortical networks of drug-naive descendants and that mediation by Crem/Fosb signalling in this brain region may be beneficial, while early postnatal fostering may further engineer molecular switching that may predispose the individual to future risky behaviours as well as accumulative potential to developing cognitive impairment later in life.
Collapse
|
17
|
Colich NL, Rosen ML, Williams ES, McLaughlin KA. Biological aging in childhood and adolescence following experiences of threat and deprivation: A systematic review and meta-analysis. Psychol Bull 2020; 146:721-764. [PMID: 32744840 DOI: 10.1037/bul0000270] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Life history theory argues that exposure to early life adversity (ELA) accelerates development, although existing evidence for this varies. We present a meta-analysis and systematic review testing the hypothesis that ELA involving threat (e.g., violence exposure) will be associated with accelerated biological aging across multiple metrics, whereas exposure to deprivation (e.g., neglect, institutional rearing) and low-socioeconomic status (SES) will not. We meta-analyze 54 studies (n = 116,010) examining associations of ELA with pubertal timing and cellular aging (telomere length and DNA methylation age), systematically review 25 studies (n = 3,253) examining ELA and neural markers of accelerated development (cortical thickness and amygdala-prefrontal cortex functional connectivity) and evaluate whether associations of ELA with biological aging vary according to the nature of adversity experienced. ELA overall was associated with accelerated pubertal timing (d = -0.10) and cellular aging (d = -0.21), but these associations varied by adversity type. Moderator analysis revealed that ELA characterized by threat was associated with accelerated pubertal development (d = -0.26) and accelerated cellular aging (d = -0.43), but deprivation and SES were unrelated to accelerated development. Systematic review revealed associations between ELA and accelerated cortical thinning, with threat-related ELA consistently associated with thinning in ventromedial prefrontal cortex, and deprivation and SES associated with thinning in frontoparietal, default, and visual networks. There was no consistent association of ELA with amygdala-PFC connectivity. These findings suggest specificity in the types of early environmental experiences associated with accelerated biological aging and highlight the importance of evaluating how accelerated aging contributes to health disparities and whether this process can be mitigated through early intervention. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
|
18
|
Ferle V, Repouskou A, Aspiotis G, Raftogianni A, Chrousos G, Stylianopoulou F, Stamatakis A. Synergistic effects of early life mild adversity and chronic social defeat on rat brain microglia and cytokines. Physiol Behav 2019; 215:112791. [PMID: 31870943 DOI: 10.1016/j.physbeh.2019.112791] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023]
Abstract
Exposure to early life stress affects the development and function of the brain and when followed by adversities in adulthood, the negative effects of stress are enhanced. Microglia has been proposed as a potential mediator of this phenomenon. In the present study, we investigated the long-term effects of mild early life stress, the consequences of a stressor in adulthood as well as their interaction on microglial and cytokine (PPARγ, IL-1β and TNFα) levels in the brain of adult male rats. As an early life stress we used a model of maternal neglect, in which the dam is present but non-accessible to the pup for 15 min during postnatal days 10-13; as a stressor in adulthood we exposed animals to chronic social defeat (CSD) for 3 weeks. We determined in the hippocampus, prefrontal cortex and amygdala, the number of Iba-1+ microglial cells, the number of PPARγ+ cells as well as the relative expression of PPARγ, IL-1β and TNFα mRNA by qPCR. Following exposure to CSD, the number of Iba-1+ cells was increased in the hippocampus and the prefrontal cortex of adult animals exposed to mild early life stress, while in the absence of CSD no such difference was observed. Moreover, following CSD PPARγ levels were increased in the hippocampus of adult males exposed as neonates to "maternal neglect". Our findings support the notion that early life stress, even a mild one, primes microglia and enhances its reactivity to a second stressful event, later in life, in accord with the "two-hit" hypothesis.
Collapse
Affiliation(s)
- Vasiliki Ferle
- Department of Basic Sciences, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Greece
| | - Anastasia Repouskou
- Faculty of Dentistry, School of Health Sciences, National and Kapodistrian University of Athens, Greece.
| | - George Aspiotis
- Department of Basic Sciences, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Greece
| | - Androniki Raftogianni
- Department of Basic Sciences, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Greece
| | - George Chrousos
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, Aghia Sofia Children's Hospital, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Fotini Stylianopoulou
- Department of Basic Sciences, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Greece.
| | - Antonios Stamatakis
- Department of Basic Sciences, Faculty of Nursing, School of Health Sciences, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
19
|
Newhouse DJ, Barcelo-Serra M, Tuttle EM, Gonser RA, Balakrishnan CN. Parent and offspring genotypes influence gene expression in early life. Mol Ecol 2019; 28:4166-4180. [PMID: 31421010 DOI: 10.1111/mec.15205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Parents can have profound effects on offspring fitness. Little, however, is known about the mechanisms through which parental genetic variation influences offspring physiology in natural systems. White-throated sparrows (Zonotrichia albicollis, WTSP) exist in two genetic morphs, tan and white, controlled by a large polymorphic supergene. Morphs mate disassortatively, resulting in two pair types: tan male × white female (T × W) pairs, which provide biparental care and white male × tan female (W × T) pairs, which provide female-biased care. To investigate how parental composition impacts offspring, we performed RNA-seq on whole blood of WTSP nestlings sampled from nests of both pair types. Parental pair type had a large effect on nestling gene expression, with 881 genes differentially expressed (DE) and seven correlated gene coexpression modules. The DE genes and modules expressed at higher levels in W × T nests with female-biased parental care function in metabolism and stress-related pathways resulting from the overrepresentation of proteolysis and stress-response genes (e.g., SOD2, NR3C1). These results show that parental genotypes and/or associated behaviours influence nestling physiology, and highlight avenues of further research investigating the ultimate implications for the maintenance of this polymorphism. Nestlings also exhibited morph-specific gene expression, with 92 differentially expressed genes, comprising immunity genes and genes encompassed by the supergene. Remarkably, we identified the same regulatory hub genes in these blood-derived expression networks as were previously identified in adult WTSP brains (EPM2A, BPNT1, TAF5L). These hub genes were located within the supergene, highlighting the importance of this gene complex in structuring regulatory networks across diverse tissues.
Collapse
Affiliation(s)
- Daniel J Newhouse
- Department of Biology, East Carolina University, Greenville, NC, USA
| | | | | | | | | |
Collapse
|
20
|
Arndt DL, Wukitsch TJ, Garcia EJ, Cain M. Histone deacetylase inhibition differentially attenuates cue-induced reinstatement: An interaction of environment and acH3K9 expression in the dorsal striatum. Behav Neurosci 2019; 133:478-488. [PMID: 31343201 DOI: 10.1037/bne0000333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Substance use disorder is driven by complex gene-environment interactions. Epigenetic histone regulation is a significant contributor to several behavioral phenotypes of drug abuse. The primary epigenetic mechanisms that drive drug taking and drug seeking are still being investigated, and it is unclear how environmental conditions alter epigenetic histone acetylation to change behaviors geared toward drug reward. This study examined the effects of environmental condition on amphetamine self-administration, and whether drug-taking and drug-seeking behaviors could be influenced through inhibition of an epigenetic regulator, histone deacetylase (HDAC). Male rats reared for 30 days in enriched (EC), isolated (IC), or standard conditions (SC) prior to amphetamine (0.03, 0.05, 0.1 mg/kg/infusion, IV) self-administration, extinction, and reinstatement sessions. The HDAC inhibitor, Trichostatin A (TsA; 0.3 mg/kg, IV), was injected 30 min prior to operant sessions. After amphetamine-induced reinstatement (0.25 mg/kg, subcutaneous [s.c.]), tissue was extracted for Western blot analyses of acetylated histone H3 lysine 9 (acH3K9) in the nucleus accumbens (NAc) and dorsal striatum (DSt). While TsA did not significantly affect amphetamine self-administration or extinction, TsA decreased cue-, but not drug-induced reinstatement in IC rats only. In the DSt, but not in the NAc, IC rats exhibited significantly less acH3K9 expression than EC and SC rats, irrespective of TsA treatment. HDAC inhibition decreases cue-induced reinstatement of amphetamine seeking in IC rats. While IC rats exhibit less acH3K9 expression in the DSt, future studies are needed to elucidate the critical epigenetic factors that drive substance abuse, particularly in vulnerable populations. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
| | | | | | - Mary Cain
- Department of Psychological Sciences
| |
Collapse
|
21
|
Saboory E, Mohammadi S, Dindarian S, Mohammadi H. Prenatal stress and elevated seizure susceptibility: Molecular inheritable changes. Epilepsy Behav 2019; 96:122-131. [PMID: 31132613 DOI: 10.1016/j.yebeh.2019.04.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/17/2019] [Accepted: 04/24/2019] [Indexed: 01/08/2023]
Abstract
Stressful episodes are common during early-life and may have a wide range of negative effects on both physical and mental status of the offspring. In addition to various neurobehavioral complications induced by prenatal stress (PS), seizure is a common complication with no fully explained cause. In this study, the association between PS and seizure susceptibility was reviewed focusing on sex differences and various underlying mechanisms. The role of drugs in the initiation of seizure and the effects of PS on the nervous system that prone the brain for seizure, especially the hypothalamic-pituitary-adrenal (HPA) axis, are also discussed in detail by reviewing the papers studying the effect of PS on glutamatergic, gamma-aminobutyric acid (GABA)ergic, and adrenergic systems in the context of seizure and epilepsy. Finally, epigenetic changes in epilepsy are described, and the underlying mechanisms of this change are expanded. As the effects of PS may be life-lasting, it is possible to prevent future psychiatric and behavioral disorders including epilepsy by preventing avoidable PS risk factors.
Collapse
Affiliation(s)
- Ehsan Saboory
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Sedra Mohammadi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
| | - Sina Dindarian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Hozan Mohammadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Chronic clozapine treatment improves the alterations of prepulse inhibition and BDNF mRNA expression in the medial prefrontal cortex that are induced by adolescent social isolation. Behav Pharmacol 2019; 30:311-319. [DOI: 10.1097/fbp.0000000000000419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
23
|
Dunn EC, Soare TW, Zhu Y, Simpkin AJ, Suderman MJ, Klengel T, Smith ADAC, Ressler KJ, Relton CL. Sensitive Periods for the Effect of Childhood Adversity on DNA Methylation: Results From a Prospective, Longitudinal Study. Biol Psychiatry 2019; 85:838-849. [PMID: 30905381 PMCID: PMC6552666 DOI: 10.1016/j.biopsych.2018.12.023] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 12/04/2018] [Accepted: 12/16/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Exposure to early-life adversity is known to predict DNA methylation (DNAm) patterns that may be related to psychiatric risk. However, few studies have investigated whether adversity has time-dependent effects based on the age at exposure. METHODS Using a two-stage structured life course modeling approach, we tested the hypothesis that there are sensitive periods when adversity induces greater DNAm changes. We tested this hypothesis in relation to two alternatives: an accumulation hypothesis, in which the effect of adversity increases with the number of occasions exposed, regardless of timing; and a recency model, in which the effect of adversity is stronger for more proximal events. Data came from the Accessible Resource for Integrated Epigenomic Studies, a subsample of mother-child pairs from the Avon Longitudinal Study of Parents and Children (n = 691-774). RESULTS After covariate adjustment and multiple testing correction, we identified 38 CpG sites that were differentially methylated at 7 years of age following exposure to adversity. Most loci (n = 35) were predicted by the timing of adversity, namely exposures before 3 years of age. Neither the accumulation nor recency of the adversity explained considerable variability in DNAm. A standard epigenome-wide association study of lifetime exposure (vs. no exposure) failed to detect these associations. CONCLUSIONS The developmental timing of adversity explains more variability in DNAm than the accumulation or recency of exposure. Very early childhood appears to be a sensitive period when exposure to adversity predicts differential DNAm patterns. Classification of individuals as exposed versus unexposed to early-life adversity may dilute observed effects.
Collapse
Affiliation(s)
- Erin C Dunn
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts.
| | - Thomas W Soare
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Stanley Center for Psychiatric Research, The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Yiwen Zhu
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew J Simpkin
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Matthew J Suderman
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts; Department of Psychiatry and Psychotherapy, University Medical Center Gottingen, Germany
| | - Andrew D A C Smith
- Applied Statistics Group, University of the West of England, Bristol, United Kingdom
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom; Institute of Genetic Medicine, University of Newcastle, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
24
|
The interaction between oxytocin receptor gene methylation and maternal behavior on children's early theory of mind abilities. Dev Psychopathol 2019; 32:511-519. [PMID: 31030686 DOI: 10.1017/s0954579419000257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Theory of mind, the ability to represent the mental states of others, is an important social cognitive process, which contributes to the development of social competence. Recent research suggests that interactions between gene and environmental factors, such as oxytocin receptor gene (OXTR) polymorphisms and maternal parenting behavior, may underlie individual differences in children's theory of mind. However, the potential influence of DNA methylation of OXTR remains unclear. The current study investigated the roles of OXTR methylation, maternal behavior, and their statistical interaction on toddlers' early emerging theory of mind abilities. Participants included a community sample of 189 dyads of mothers and their 2- to 3-year-old children, whose salivary DNA was analyzed. Results indicated that more maternal structuring behavior was associated with better performance, on a battery of three theory of mind tasks, while higher OXTR methylation within exon 3 was associated with poorer performance. A significant interaction also emerged, such that OXTR methylation was related to theory of mind among children whose mothers displayed less structuring, when controlling for children's age, sex, ethnicity, number of child-aged siblings, verbal ability, and maternal education. Maternal structuring behavior may buffer the potential negative impact of hypermethylation on OXTR gene expression and function.
Collapse
|
25
|
Keller SM, Doherty TS, Roth TL. Pharmacological Manipulation of DNA Methylation in Adult Female Rats Normalizes Behavioral Consequences of Early-Life Maltreatment. Front Behav Neurosci 2018; 12:126. [PMID: 30008666 PMCID: PMC6034089 DOI: 10.3389/fnbeh.2018.00126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/06/2018] [Indexed: 01/03/2023] Open
Abstract
Exposure to adversity early in development alters brain and behavioral trajectories. Data continue to accumulate that epigenetic mechanisms are a mediating factor between early-life adversity and adult behavioral phenotypes. Previous work from our laboratory has shown that female Long-Evans rats exposed to maltreatment during infancy display both aberrant forced swim behavior and patterns of brain DNA methylation in adulthood. Therefore, we examined the possibility of rescuing the aberrant forced swim behavior in maltreated-adult females by administering an epigenome-modifying drug (zebularine) at a dose previously shown to normalize DNA methylation. We found that zebularine normalized behavior in the forced swim test in maltreated females such that they performed at the levels of controls (females that had been exposed to only nurturing care during infancy). These data help link DNA methylation to an adult phenotype in our maltreatment model, and more broadly provide additional evidence that non-targeted epigenetic manipulations can change behavior associated with early-life adversity.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
26
|
Cecil CA, Walton E, Jaffee SR, O’Connor T, Maughan B, Relton CL, Smith RG, McArdle W, Gaunt TR, Ouellet-Morin I, Barker ED. Neonatal DNA methylation and early-onset conduct problems: A genome-wide, prospective study. Dev Psychopathol 2018; 30:383-397. [PMID: 28595673 PMCID: PMC7612607 DOI: 10.1017/s095457941700092x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Early-onset conduct problems (CP) are a key predictor of adult criminality and poor mental health. While previous studies suggest that both genetic and environmental risks play an important role in the development of early-onset CP, little is known about potential biological processes underlying these associations. In this study, we examined prospective associations between DNA methylation (cord blood at birth) and trajectories of CP (4-13 years), using data drawn from the Avon Longitudinal Study of Parents and Children. Methylomic variation at seven loci across the genome (false discovery rate < 0.05) differentiated children who go on to develop early-onset (n = 174) versus low (n = 86) CP, including sites in the vicinity of the monoglyceride lipase (MGLL) gene (involved in endocannabinoid signaling and pain perception). Subthreshold associations in the vicinity of three candidate genes for CP (monoamine oxidase A [MAOA], brain-derived neurotrophic factor [BDNF], and FK506 binding protein 5 [FKBP5]) were also identified. Within the early-onset CP group, methylation levels of the identified sites did not distinguish children who will go on to persist versus desist in CP behavior over time. Overall, we found that several of the identified sites correlated with prenatal exposures, and none were linked to known genetic methylation quantitative trait loci. Findings contribute to a better understanding of epigenetic patterns associated with early-onset CP.
Collapse
Affiliation(s)
| | - Esther Walton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, UK
| | - Sara R. Jaffee
- Department of Psychology, University of Pennsylvania, USA
| | | | - Barbara Maughan
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Caroline L. Relton
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, UK
| | | | - Wendy McArdle
- School of Social and Community Medicine, University of Bristol, UK
| | - Tom R. Gaunt
- School of Social and Community Medicine, University of Bristol, UK
| | | | - Edward D. Barker
- Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
27
|
Bendre M, Comasco E, Checknita D, Tiihonen J, Hodgins S, Nilsson KW. Associations Between MAOA-uVNTR Genotype, Maltreatment, MAOA Methylation, and Alcohol Consumption in Young Adult Males. Alcohol Clin Exp Res 2018; 42:508-519. [PMID: 29222910 DOI: 10.1111/acer.13578] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 12/04/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epigenetic mechanisms are candidate moderators of the effect of maltreatment on brain and behavior. Interactions between maltreatment and the monoamine oxidase A upstream variable number tandem repeat genotype (MAOA-uVNTR) are associated with alcohol-related problems. However, presently it is not known whether DNA methylation moderates this association. The study focused on 53 young adult males and aimed to determine whether MAOA methylation moderated the association of alcohol-related problems with the interaction of MAOA-uVNTR and maltreatment, and whether alcohol consumption moderated the association of MAOA methylation with the interaction of MAOA-uVNTR and maltreatment. METHODS MAOA-uVNTR genotypes with ≤ 3 and > 3 repeats were categorized as short (S) and long (L), respectively. Data on maltreatment were obtained retrospectively, using self-reported questionnaires. DNA methylation of 16 candidate CpGs within part of the MAOA first exon and intron was assessed and grouped based on principal component analyses. Alcohol-related problems were assessed using the Alcohol Use Disorders Identification Test (AUDIT). Alcohol consumption was measured using AUDIT-C. Moderation effects were assessed and probed using the moderated moderation model and Johnson-Neyman's method, respectively. RESULTS Carriers of the S allele, who experienced maltreatment and displayed lower Component 1 (mean of CpGs 13-16 in the first intron) MAOA methylation levels, reported higher AUDIT score in contrast to L-allele carriers. Carriers of the S allele, who reported higher AUDIT-C score and experienced maltreatment, displayed lower Component 3 (mean of CpGs 2-6 in the first exon) MAOA methylation levels than L-allele carriers. CONCLUSIONS Intronic methylation moderated the association of alcohol-related problems with the interaction of MAOA-uVNTR and maltreatment. Alcohol consumption moderated the association of exonic methylation with the interaction of MAOA-uVNTR and maltreatment. These results suggest that epigenetic factors as well as genotype and maltreatment play a role in the development of alcohol misuse among young adult males.
Collapse
Affiliation(s)
- Megha Bendre
- Department of Neuroscienc, Uppsala University, Uppsala, Sweden.,Centre for Clinical Research, Uppsala University, County Hospital, Västerås, Sweden
| | - Erika Comasco
- Department of Neuroscienc, Uppsala University, Uppsala, Sweden
| | - Dave Checknita
- Department of Neuroscienc, Uppsala University, Uppsala, Sweden.,Centre for Clinical Research, Uppsala University, County Hospital, Västerås, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jari Tiihonen
- Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Sheilagh Hodgins
- Institut Universitaire en Santé Mentale de Montréal, Université de Montréal, Montréal, Canada
| | - Kent W Nilsson
- Centre for Clinical Research, Uppsala University, County Hospital, Västerås, Sweden
| |
Collapse
|
28
|
Boschen KE, Keller SM, Roth TL, Klintsova AY. Epigenetic mechanisms in alcohol- and adversity-induced developmental origins of neurobehavioral functioning. Neurotoxicol Teratol 2018; 66:63-79. [PMID: 29305195 DOI: 10.1016/j.ntt.2017.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
The long-term effects of developmental alcohol and stress exposure are well documented in both humans and non-human animal models. Damage to the brain and attendant life-long impairments in cognition and increased risk for psychiatric disorders are debilitating consequences of developmental exposure to alcohol and/or psychological stress. Here we discuss evidence for a role of epigenetic mechanisms in mediating these consequences. While we highlight some of the common ways in which stress or alcohol impact the epigenome, we point out that little is understood of the epigenome's response to experiencing both stress and alcohol exposure, though stress is a contributing factor as to why women drink during pregnancy. Advancing our understanding of this relationship is of critical concern not just for the health and well-being of individuals directly exposed to these teratogens, but for generations to come.
Collapse
Affiliation(s)
- K E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, United States
| | - S M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - T L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - A Y Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
29
|
Ajonijebu DC, Abboussi O, Russell VA, Mabandla MV, Daniels WMU. Epigenetics: a link between addiction and social environment. Cell Mol Life Sci 2017; 74:2735-2747. [PMID: 28255755 PMCID: PMC11107568 DOI: 10.1007/s00018-017-2493-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 01/14/2023]
Abstract
The detrimental effects of drug abuse are apparently not limited to individuals but may also impact the vulnerability of their progenies to develop addictive behaviours. Epigenetic signatures, early life experience and environmental factors, converge to influence gene expression patterns in addiction phenotypes and consequently may serve as mediators of behavioural trait transmission between generations. The majority of studies investigating the role of epigenetics in addiction do not consider the influence of social interactions. This shortcoming in current experimental approaches necessitates developing social models that reflect the addictive behaviour in a free-living social environment. Furthermore, this review also reports on the advancement of interventions for drug addiction and takes into account the emerging roles of histone deacetylase (HDAC) inhibitors in the etiology of drug addiction and that HDAC may be a potential therapeutic target at nucleosomal level to improve treatment outcomes.
Collapse
Affiliation(s)
- Duyilemi C Ajonijebu
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Oualid Abboussi
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa.
| | - Vivienne A Russell
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Musa V Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - William M U Daniels
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
- School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
30
|
Chen Y, Baram TZ. Toward Understanding How Early-Life Stress Reprograms Cognitive and Emotional Brain Networks. Neuropsychopharmacology 2016; 41:197-206. [PMID: 26105143 PMCID: PMC4677123 DOI: 10.1038/npp.2015.181] [Citation(s) in RCA: 322] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/26/2015] [Accepted: 06/19/2015] [Indexed: 01/19/2023]
Abstract
Vulnerability to emotional disorders including depression derives from interactions between genes and environment, especially during sensitive developmental periods. Adverse early-life experiences provoke the release and modify the expression of several stress mediators and neurotransmitters within specific brain regions. The interaction of these mediators with developing neurons and neuronal networks may lead to long-lasting structural and functional alterations associated with cognitive and emotional consequences. Although a vast body of work has linked quantitative and qualitative aspects of stress to adolescent and adult outcomes, a number of questions are unclear. What distinguishes 'normal' from pathologic or toxic stress? How are the effects of stress transformed into structural and functional changes in individual neurons and neuronal networks? Which ones are affected? We review these questions in the context of established and emerging studies. We introduce a novel concept regarding the origin of toxic early-life stress, stating that it may derive from specific patterns of environmental signals, especially those derived from the mother or caretaker. Fragmented and unpredictable patterns of maternal care behaviors induce a profound chronic stress. The aberrant patterns and rhythms of early-life sensory input might also directly and adversely influence the maturation of cognitive and emotional brain circuits, in analogy to visual and auditory brain systems. Thus, unpredictable, stress-provoking early-life experiences may influence adolescent cognitive and emotional outcomes by disrupting the maturation of the underlying brain networks. Comprehensive approaches and multiple levels of analysis are required to probe the protean consequences of early-life adversity on the developing brain. These involve integrated human and animal-model studies, and approaches ranging from in vivo imaging to novel neuroanatomical, molecular, epigenomic, and computational methodologies. Because early-life adversity is a powerful determinant of subsequent vulnerabilities to emotional and cognitive pathologies, understanding the underlying processes will have profound implications for the world's current and future children.
Collapse
Affiliation(s)
- Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
31
|
Mitchell C, Schneper LM, Notterman DA. DNA methylation, early life environment, and health outcomes. Pediatr Res 2016; 79:212-9. [PMID: 26466079 PMCID: PMC4798238 DOI: 10.1038/pr.2015.193] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 07/27/2015] [Indexed: 11/09/2022]
Abstract
Epigenetics, and especially DNA methylation, have recently become provocative biological explanations for early-life environmental effects on later health. Despite the large increase in papers on the topic over the last few years, many questions remain with regards to the biological feasibility of this mechanism and the strength of the evidence to date. In this review, we examine the literature on early-life effects on epigenetic patterns, with special emphasis on social environmental influences. First, we review the basic biology of epigenetic modification of DNA and debate the role of early-life stressful, protective, and positive environments on gene-specific, system-specific, and whole-genome epigenetic patterns later in life. Second, we compare the epigenetic literatures of both humans and other animals and review the research linking epigenetic patterns to health in order to complete the mechanistic pathway. Third, we discuss physical environmental and social environmental effects, which have to date, generally not been jointly considered. Finally, we close with a discussion of the current state of the area's research, its future direction, and its potential use in pediatric health.
Collapse
Affiliation(s)
- Colter Mitchell
- Survey Research Center and Population Studies Center, University of Michigan, Ann Arbor, Michigan
| | - Lisa M. Schneper
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| |
Collapse
|
32
|
Lemieux A, al'Absi M. Stress psychobiology in the context of addiction medicine: from drugs of abuse to behavioral addictions. PROGRESS IN BRAIN RESEARCH 2015; 223:43-62. [PMID: 26806770 DOI: 10.1016/bs.pbr.2015.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In this chapter, we briefly review the basic biology of psychological stress and the stress response. We propose that psychological stress and the neurobiology of the stress response play in substance use initiation, maintenance, and relapse. The proposed mechanisms for this include, on the one hand, the complex interactions between biological mediators of the stress response and the dopaminergic reward system and, on the other hand, mediators of the stress response and other systems crucial in moderating key addiction-related behaviors such as endogenous opioids, the sympathetic-adrenal-medullary system, and endocannabinoids. Exciting new avenues of study including genomics, sex as a moderator of the stress response, and behavioral addictions (gambling, hypersexuality, dysfunctional internet use, and food as an addictive substance) are also briefly presented within the context of stress as a moderator of the addictive process.
Collapse
Affiliation(s)
| | - Mustafa al'Absi
- University of Minnesota School of Medicine, Duluth, MN, USA.
| |
Collapse
|