1
|
Nguyen CH, Salazar MG, Rincón-Cortés M. Female rats exposed to early life scarcity-adversity are resilient to later life changes in maternal behavior. Horm Behav 2025; 170:105720. [PMID: 40090292 DOI: 10.1016/j.yhbeh.2025.105720] [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: 07/02/2024] [Revised: 01/10/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
In both humans and rodents, maternal care is disturbed by exposure to environmental adversity, including low resource conditions (i.e., poverty, scarcity). Maternal adversity is associated with compromised quality of mother-infant attachment and increased adverse caregiving patterns such as abuse, maltreatment and/or neglect, which disrupt behavioral development in the female offspring. Importantly, maternal behavior is thought to be an intergenerational behavior, meaning that the quality of maternal care a female experiences during early life is thought to influence the quality of care she will display towards her own offspring when she becomes a mother. Here, we tested this idea by employing a rodent model of postpartum environmental adversity based on creating an impoverished nesting environment during postpartum days (PD) 2-9, which disrupts mother-infant interactions and is thought to upregulate hypothalamic-pituitary-adrenal (HPA)-axis function in the pups. We examined the impact of this form of early life adversity on pup stress hormone (i.e., corticosterone- CORT) levels by collecting trunk blood and later life maternal behavior by conducting maternal behavior observations and maternal motivation tests (e.g., T-Maze, pup retrieval, pup-associated conditioned place preference) in the first filial (F1) generation. We report no impact of early life scarcity-adversity/adverse caregiving on pup CORT levels or later life naturalistic or motivated maternal behaviors. In sum, we show that female rat pups who experienced adverse caregiving during early life showed resilience towards developing negative caregiving patterns, as they did not perpetuate the same aberrant maternal behavior that they received from their mothers.
Collapse
Affiliation(s)
- Christine H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Melissa G Salazar
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Millie Rincón-Cortés
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
2
|
Silva-Hurtado TJ, Giua G, Lassalle O, Makrini-Maleville L, Strauss B, Wager-Miller J, Freyermuth JM, Mackie K, Valjent E, Manzoni OJ, Chavis P. Reelin Deficiency and Synaptic Impairment in the Adolescent Prefrontal Cortex Following Initial Synthetic Cannabinoid Exposure. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100426. [PMID: 39926699 PMCID: PMC11804564 DOI: 10.1016/j.bpsgos.2024.100426] [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: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 02/11/2025] Open
Abstract
Background Adolescent cannabinoid exposure can have long-lasting effects on the brain, particularly in the prefrontal cortex, where the reelin protein plays a crucial role in neural organization. Chronic cannabinoid exposure leads to reelin deficiency and behavioral abnormalities, but the underlying mechanisms remain unclear. With the increasing use of synthetic cannabinoids (SCs) among young people, understanding these effects is crucial. Methods We examined the cellular and synaptic consequences of initial SC exposure in adolescent male mice 1 day after a single in vivo exposure to WIN 55,212-2. Our approach combined immunohistochemistry, Western blots, conditional CB1 receptor (CB1R) knockout mouse lines, quantitative polymerase chain reaction, and ex vivo electrophysiology to investigate the effects of SC on reelin expression and synaptic plasticity. Additionally, single-molecule fluorescent in situ hybridization profiling was used to identify cellular coexpression patterns of reelin and CB1Rs. Results Our findings indicate that a single exposure to SC decreased reelin expression in specific prefrontal cortex layers accompanied by disrupted proteolytic fragmentation but not changes in messenger RNA expression. Single-molecule fluorescent in situ hybridization profiling revealed a strong coexpression of CB1R and reelin. Furthermore, our pharmacological and genetic approaches demonstrated that CB1Rs in GABAergic (gamma-aminobutyric acidergic) neurons mediate the SC-induced decrease in reelin. This decrease in reelin results in a reduction in long-term potentiation, phenocopying reelin haploinsufficient mice. Notably, we restored long-term potentiation by infusing reelin bilaterally, establishing a functional link between reelin depletion and synaptic deficits. Conclusions These findings provide new insights into the neural consequences of adolescent cannabinoid consumption and highlight the critical role of reelin in the cellular mechanisms associated with SC initiation during adolescence.
Collapse
Affiliation(s)
- Thenzing J. Silva-Hurtado
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Gabriele Giua
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Olivier Lassalle
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Leila Makrini-Maleville
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Benjamin Strauss
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Jim Wager-Miller
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
- The Gill Institute for Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | | | - Ken Mackie
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
- The Gill Institute for Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Emmanuel Valjent
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Olivier J.J. Manzoni
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Pascale Chavis
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| |
Collapse
|
3
|
Walker SL, Glasper ER. Unraveling sex differences in maternal and paternal care impacts on social behaviors and neurobiological responses to early-life adversity. Front Neuroendocrinol 2025; 76:101162. [PMID: 39561882 PMCID: PMC11811932 DOI: 10.1016/j.yfrne.2024.101162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/21/2024]
Abstract
Early-life stress (ELS) affects the development of prosocial behaviors and social-cognitive function, often leading to structural brain changes and increased psychosocial disorders. Recent studies suggest that mother- and father-child relationships independently influence social development in a sex-specific manner, but the effects of impaired father-child relationships are often overlooked. This review examines preclinical rodent studies to explore how parental neglect impacts neuroplasticity and social behaviors in offspring. We highlight that disruptions in maternal interactions may affect male pups more in uniparental rodents, while impaired paternal interactions in biparental rodents tend to impact female pups more. Due to limited research, the separate effects of maternal and paternal neglect on brain development and social behaviors in biparental species remain unclear. Addressing these gaps could clarify the sex-specific mechanisms underlying social and neurobiological deficits following parental neglect.
Collapse
Affiliation(s)
- Shakeera L Walker
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States
| | - Erica R Glasper
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, United States; Neuroscience Graduate Program, The Ohio State University, Columbus, OH, 43210, United States; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, 43210, United States.
| |
Collapse
|
4
|
Warhaftig G, Almeida D, Turecki G. Early life adversity across different cell- types in the brain. Neurosci Biobehav Rev 2023; 148:105113. [PMID: 36863603 DOI: 10.1016/j.neubiorev.2023.105113] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023]
Abstract
Early life adversity (ELA)- which includes physical, psychological, emotional, and sexual abuse is one of the most common predictors to diverse psychopathologies later in adulthood. As ELA has a lasting impact on the brain at a developmental stage, recent findings from the field highlighted the specific contributions of different cell types to ELA and their association with long lasting consequences. In this review we will gather recent findings describing morphological, transcriptional and epigenetic alterations within neurons, glia and perineuronal nets and their associated cellular subpopulation. The findings reviewed and summarized here highlight important mechanisms underlying ELA and point to therapeutic approaches for ELA and related psychopathologies later in life.
Collapse
Affiliation(s)
- Gal Warhaftig
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Hospital Research Center, Montreal QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal QC H3A 1A1, Canada.
| |
Collapse
|
5
|
The lifetime impact of stress on fear regulation and cortical function. Neuropharmacology 2023; 224:109367. [PMID: 36464208 DOI: 10.1016/j.neuropharm.2022.109367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
A variety of stressful experiences can influence the ability to form and subsequently inhibit fear memory. While nonsocial stress can impact fear learning and memory throughout the lifespan, psychosocial stressors that involve negative social experiences or changes to the social environment have a disproportionately high impact during adolescence. Here, we review converging lines of evidence that suggest that development of prefrontal cortical circuitry necessary for both social experiences and fear learning is altered by stress exposure in a way that impacts both social and fear behaviors throughout the lifespan. Further, we suggest that psychosocial stress, through its impact on the prefrontal cortex, may be especially detrimental during early developmental periods characterized by higher sociability. This article is part of the Special Issue on 'Fear, Anxiety and PTSD'.
Collapse
|
6
|
Zhou A, Ancelin ML, Ritchie K, Ryan J. Childhood adverse events and BDNF promoter methylation in later-life. Front Psychiatry 2023; 14:1108485. [PMID: 36911114 PMCID: PMC9998928 DOI: 10.3389/fpsyt.2023.1108485] [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: 11/26/2022] [Accepted: 02/13/2023] [Indexed: 03/14/2023] Open
Abstract
Studies have shown that the effects of early-life stress and trauma can be enduring, with long-term negative effects on health. Epigenetics, including DNA methylation, have been implicated as a potential mechanism for these effects. Brain-derived neurotropic factor (BDNF) is a neurotransmitter involved in learning and memory, and altered BDNF promoter methylation measured in peripheral tissue has been found with early-life stress. However, whether such methylation differences remain stable into later life, is unknown. This study aimed to investigate the association between childhood adversity and BDNF promoter methylation in adults aged 65 years and over. Data came from a large study of older community-dwelling individuals in France (ESPRIT). Information on three major childhood adverse events, namely abuse/maltreatment, war/natural disaster, and financial difficulties/poverty, was obtained by retrospective reporting from participants of ESPRIT study. BDNF promoter I and IV methylation was assessed in blood and buccal tissue. Linear regression analysis was performed, adjusting for age, sex, education, depression, and morbidity. Among 927 participants, there was no strong evidence that childhood abuse/maltreatment or financial difficulties/poverty were associated with BDNF methylation in older individuals. For war/natural disaster, differential methylation at four of twenty-nine CpG sites was observed, however, these would not have remained significant after correction for multiple testing. Together, these findings do not support a long-term association between adverse childhood events and BDNF methylation in older age, but further large prospective studies are needed, which do not target specific genes, but consider DNA methylation across the genome.
Collapse
Affiliation(s)
- Aoshuang Zhou
- Division of Epidemiology, Jockey Club School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Marie-Laure Ancelin
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Karen Ritchie
- Institute for Neurosciences of Montpellier, University of Montpellier, INSERM, Montpellier, France
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Govender P, Ghai M, Okpeku M. Sex-specific DNA methylation: impact on human health and development. Mol Genet Genomics 2022; 297:1451-1466. [PMID: 35969270 DOI: 10.1007/s00438-022-01935-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022]
Abstract
Human evolution has shaped gender differences between males and females. Over the years, scientific studies have proposed that epigenetic modifications significantly influence sex-specific differences. The evolution of sex chromosomes with epigenetics as the driving force may have led to one sex being more adaptable than the other when exposed to various factors over time. Identifying and understanding sex-specific differences, particularly in DNA methylation, will help determine how each gender responds to factors, such as disease susceptibility, environmental exposure, brain development and neurodegeneration. From a medicine and health standpoint, sex-specific methylation studies have shed light on human disease severity, progression, and response to therapeutic intervention. Interesting findings in gender incongruent individuals highlight the role of genetic makeup in influencing DNA methylation differences. Sex-specific DNA methylation studies will empower the biotechnology and pharmaceutical industry with more knowledge to identify biomarkers, design and develop sex bias drugs leading to better treatment in men and women based on their response to different diseases.
Collapse
Affiliation(s)
- Priyanka Govender
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Meenu Ghai
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa.
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| |
Collapse
|
8
|
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
|
9
|
Fitter MH, Stern JA, Straske MD, Allard T, Cassidy J, Riggins T. Mothers’ Attachment Representations and Children’s Brain Structure. Front Hum Neurosci 2022; 16:740195. [PMID: 35370579 PMCID: PMC8967255 DOI: 10.3389/fnhum.2022.740195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 01/24/2022] [Indexed: 12/16/2022] Open
Abstract
Ample research demonstrates that parents’ experience-based mental representations of attachment—cognitive models of close relationships—relate to their children’s social-emotional development. However, no research to date has examined how parents’ attachment representations relate to another crucial domain of children’s development: brain development. The present study is the first to integrate the separate literatures on attachment and developmental social cognitive neuroscience to examine the link between mothers’ attachment representations and 3- to 8-year-old children’s brain structure. We hypothesized that mothers’ attachment representations would relate to individual differences in children’s brain structures involved in stress regulation—specifically, amygdala and hippocampal volumes—in part via mothers’ responses to children’s distress. We assessed 52 mothers’ attachment representations (secure base script knowledge on the Attachment Script Assessment and self-reported attachment avoidance and anxiety on the Experiences in Close Relationships scale) and children’s brain structure. Mothers’ secure base script knowledge was significantly related to children’s smaller left amygdala volume but was unrelated to hippocampal volume; we found no indirect links via maternal responses to children’s distress. Exploratory analyses showed associations between mothers’ attachment representations and white matter and thalamus volumes. Together, these preliminary results suggest that mothers’ attachment representations may be linked to the development of children’s neural circuitry related to stress regulation.
Collapse
Affiliation(s)
- Megan H. Fitter
- Maryland Child and Family Development Lab, University of Maryland, College Park, College Park, MD, United States
- *Correspondence: Megan H. Fitter,
| | - Jessica A. Stern
- BabyLab, University of Virginia, Charlottesville, VA, United States
| | - Martha D. Straske
- Maryland Child and Family Development Lab, University of Maryland, College Park, College Park, MD, United States
| | - Tamara Allard
- Neurocognitive Development Lab, University of Maryland, College Park, College Park, MD, United States
| | - Jude Cassidy
- Maryland Child and Family Development Lab, University of Maryland, College Park, College Park, MD, United States
| | - Tracy Riggins
- Neurocognitive Development Lab, University of Maryland, College Park, College Park, MD, United States
| |
Collapse
|
10
|
Collins NJ, Zimmerman CW, Phillips NLH, Fern S, Doherty TS, Roth TL. Developmental administration of valproic acid alters DNA methylation and maternal behavior. Dev Psychobiol 2022; 64:e22231. [PMID: 35312054 DOI: 10.1002/dev.22231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/01/2021] [Accepted: 11/18/2021] [Indexed: 02/05/2023]
Abstract
Exposure to adversity in early development has powerful and potentially lasting consequences on behavior. Previous work in our laboratory using female Long-Evans rats has demonstrated that exposure to early-life maltreatment manifests into alterations in dam behavior, including a perpetuation of the maltreatment phenotype. These observed behavioral changes coincide with changes in epigenetic activity in the prefrontal cortex (PFC). Further, treating dams with a chromatin modifying agent (Zebularine) normalizes methylation and maltreatment phenotypes, suggesting a link between epigenetic programming and phenotypic outcomes. Here, we sought to investigate if administration of a chromatin modifying agent concurrent with the experience of maltreatment normalizes epigenetic activity associated with maltreatment and alters behavioral trajectories. Administration of valproic acid (VPA) transiently lowered levels of global DNA methylation in the PFC, regardless of exposure to nurturing care or maltreatment. When VPA-exposed animals reached adulthood, they engaged in more adverse behaviors toward their offspring. These data provide further evidence linking epigenetic changes in the developing brain with effects on behavior.
Collapse
Affiliation(s)
- Nicholas J Collins
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Catherine W Zimmerman
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Natalia L H Phillips
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Samantha Fern
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
11
|
Provenzi L, Villa M, Mambretti F, Citterio A, Grumi S, Bertazzoli E, Biasucci G, Decembrino L, Gardella B, Giacchero R, Magnani ML, Nacinovich R, Pisoni C, Prefumo F, Orcesi S, Scelsa B, Giorda R, Borgatti R. Is Brain-Derived Neurotropic Factor Methylation Involved in the Association Between Prenatal Stress and Maternal Postnatal Anxiety During the COVID-19 Pandemic? Front Psychiatry 2022; 13:950455. [PMID: 35911240 PMCID: PMC9329563 DOI: 10.3389/fpsyt.2022.950455] [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: 05/22/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The COVID-19 pandemic is a collective trauma that may expose susceptible individuals to high levels of stress. Pregnant women represent a high-risk population, considering that pregnancy is a period of heightened neuroplasticity and susceptibility to stress through epigenetic mechanisms. Previous studies showed that the methylation status of the BDNF gene is linked with prenatal stress exposure. The goals of this study were (a) to assess the association between pandemic-related stress and postnatal anxiety and (b) to investigate the potential role of maternal BDNF methylation as a significant mediator of this association. METHODS In the present study, we report data on the association among pandemic-related stress during pregnancy, maternal BDNF methylation, and postnatal anxiety symptoms. Pandemic-related stress and postnatal anxiety were assessed through self-report instruments. BDNF methylation was estimated in 11 CpG sites in DNA from mothers' buccal cells. Complete data were available from 108 mothers. RESULTS Results showed that pandemic-related stress was associated with an increased risk of postnatal anxiety, r = 0.20, p < 0.05. CpG-specific BDNF methylation was significantly associated with both prenatal pandemic-related stress, r = 0.21, p < 0.05, and postnatal maternal anxious symptoms, r = 0.25, p = 0.01. Moreover, a complete mediation by the BDNF CpG6 methylation emerged between pandemic-related stress during pregnancy and postnatal maternal anxiety, ACME = 0.66, p < 0.05. CONCLUSION These findings suggest that BDNF epigenetic regulation by pandemic-related stress might contribute to increase the risk of anxiety in mothers. Policymakers should prioritize the promotion of health and wellbeing in pregnant women and mothers during the present healthcare emergency.
Collapse
Affiliation(s)
- Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Marco Villa
- Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | | | | | - Serena Grumi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | | | | | | | - Barbara Gardella
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | | | - Renata Nacinovich
- ASST Monza, Monza, Italy.,Department of Medicine and Surgery, Università Bicocca, Milan, Italy
| | | | - Federico Prefumo
- ASST Spedali Civili, Brescia, Italy.,Division of Obstetrics and Gynecology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | | | - Roberto Giorda
- Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
12
|
Timmerman BM, Mooney-Leber SM, Brummelte S. The effects of neonatal procedural pain and maternal isolation on hippocampal cell proliferation and reelin concentration in neonatal and adult male and female rats. Dev Psychobiol 2021; 63:e22212. [PMID: 34813104 DOI: 10.1002/dev.22212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Preterm births accounted for over 10% of all U.S. live births in 2019 and the rate is rising. Neonatal stressors, especially procedural pain, experienced by preterm infants in the neonatal intensive care unit (NICU) have been associated with neurodevelopmental impairments. Parental care can alleviate stress during stressful or painful procedures; however, infants in the NICU often receive reduced parental care compared with their peers. Animal studies suggest that decreased maternal care similarly impairs neurodevelopment but also influences the effects of neonatal pain. It is important to mimic both stressors in animal models of neonatal stress exposure. In this study, researchers investigated the individual and combined impact of neonatal pain and maternal isolation on reelin protein levels and cellular proliferation in the hippocampal dentate gyrus of 8 days old and adult rats. Exposure to either stressor individually, but not both, increased reelin levels in the dentate gyrus of adult females without significantly altering reelin levels in adult males. However, cell proliferation levels at either age were unaffected by the early-life stressors. These results suggest that each early-life stressor has a unique effect on markers of brain development and more research is needed to further investigate their distinct influences.
Collapse
Affiliation(s)
- Brian M Timmerman
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Sean M Mooney-Leber
- Department of Psychology, University of Wisconsin-Stevens Points, Stevens Point, Wisconsin, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA.,Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
13
|
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
|
14
|
The Impact of Stress Within and Across Generations: Neuroscientific and Epigenetic Considerations. Harv Rev Psychiatry 2021; 29:303-317. [PMID: 34049337 DOI: 10.1097/hrp.0000000000000300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The impact of stress and trauma on biological systems in humans can be substantial. They can result in epigenetic changes, accelerated brain development and sexual maturation, and predisposition to psychopathology. Such modifications may be accompanied by behavioral, emotional, and cognitive overtones during one's lifetime. Exposure during sensitive periods of neural development may lead to long-lasting effects that may not be affected by subsequent environmental interventions. The cumulative effects of life stressors in an individual may affect offspring's methylome makeup and epigenetic clocks, neurohormonal modulation and stress reactivity, and physiological and reproductive development. While offspring may suffer deleterious effects from parental stress and their own early-life adversity, these factors may also confer traits that prove beneficial and enhance fitness to their own environment. This article synthesizes the data on how stress shapes biological and behavioral dimensions, drawing from preclinical and human models. Advances in this field of knowledge should potentially allow for an improved understanding of how interventions may be increasingly tailored according to individual biomarkers and developmental history.
Collapse
|
15
|
Shupe EA, Clinton SM. Neonatal resource scarcity alters maternal care and impacts offspring core temperature and growth in rats. Dev Psychobiol 2021; 63:e22144. [PMID: 34053070 DOI: 10.1002/dev.22144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Stressful experiences during childhood, including poverty and inconsistent parental care, can enhance vulnerability for worsened physical and mental health outcomes in adulthood. Using Sprague Dawley rats, the present study explored the impact of limited resource availability on maternal behavior and physiological and emotional behavior outcomes in the offspring. Early life adversity was induced by incorporating aspects of the limited bedding and nesting and scarcity models, wherein limited resource availability has previously been shown to provoke unpredictable or adverse maternal care respectively. In our hands, neonatal limited bedding (NLB) stress during postnatal days (P)2-9 altered maternal care, augmenting pup-directed behaviors and reducing self-directed behaviors, and modestly increased the frequency of transitions between discrete behaviors across consecutive timed observations. NLB-exposed pups had lower core body temperatures immediately following the stressful manipulation and exhibited decreased body weight gain across development. However, NLB exposure did not impact adult offspring's social or emotional behavior outcomes in the three-chamber social interaction, novelty-suppressed feeding, splash, or forced swim tests. These findings add to the literature demonstrating that early life adversity impacts maternal care in rodents and can disrupt certain metabolic and thermoregulatory outcomes in the offspring.
Collapse
Affiliation(s)
- Elizabeth A Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, Virginia, USA
| | - Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University (Virginia Tech), Blacksburg, Virginia, USA
| |
Collapse
|
16
|
Brudzynski SM. Biological Functions of Rat Ultrasonic Vocalizations, Arousal Mechanisms, and Call Initiation. Brain Sci 2021; 11:brainsci11050605. [PMID: 34065107 PMCID: PMC8150717 DOI: 10.3390/brainsci11050605] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 01/21/2023] Open
Abstract
This review summarizes all reported and suspected functions of ultrasonic vocalizations in infant and adult rats. The review leads to the conclusion that all types of ultrasonic vocalizations subserving all functions are vocal expressions of emotional arousal initiated by the activity of the reticular core of the brainstem. The emotional arousal is dichotomic in nature and is initiated by two opposite-in-function ascending reticular systems that are separate from the cognitive reticular activating system. The mesolimbic cholinergic system initiates the aversive state of anxiety with concomitant emission of 22 kHz calls, while the mesolimbic dopaminergic system initiates the appetitive state of hedonia with concomitant emission of 50 kHz vocalizations. These two mutually exclusive arousal systems prepare the animal for two different behavioral outcomes. The transition from broadband infant isolation calls to the well-structured adult types of vocalizations is explained, and the social importance of adult rat vocal communication is emphasized. The association of 22 kHz and 50 kHz vocalizations with aversive and appetitive states, respectively, was utilized in numerous quantitatively measured preclinical models of physiological, psychological, neurological, neuropsychiatric, and neurodevelopmental investigations. The present review should help in understanding and the interpretation of these models in biomedical research.
Collapse
Affiliation(s)
- Stefan M Brudzynski
- Department of Psychology, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
17
|
Ahmed OG, Shehata GA, Ali RM, Makboul R, Abd Allah ESH, Abd El-Rady NM. Folic acid ameliorates neonatal isolation-induced autistic like behaviors in rats: epigenetic modifications of BDNF and GFAP promotors. Appl Physiol Nutr Metab 2021; 46:964-975. [PMID: 33635721 DOI: 10.1139/apnm-2020-0923] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The current study investigated the role of epigenetic dysregulation of brain derived neurotrophic factor (BDNF) and glial fibrillary acidic protein (GFAP) genes and oxidative stress as possible mechanisms of autistic-like behaviors in neonatal isolation model in rats and the impact of folic acid administration on these parameters. Forty Wistar albino pups were used as follows: control, folic acid administered, isolated, and isolated folic acid treated groups. Isolated pups were separated from their mothers for 90 min daily from postnatal day (PND) 1 to 11. Pups (isolated or control) received either the vehicle or folic acid (4 mg/kg/day) orally from PND 1 to 29. Behavioral tests were done from PND 30 to 35. Oxidative stress markers and antioxidant defense in the frontal cortex homogenate were determined. DNA methylation of BDNF and GFAP genes was determined by qPCR. Histopathological examination was carried out. Neonatal isolation produced autistic-like behaviors that were associated with BDNF and GFAP hypomethylation, increased oxidative stress, increased inflammatory cell infiltration, and structural changes in the frontal cortex. Folic acid administration concurrently with isolation reduced neonatal isolation-induced autistic-like behaviors, decreased oxidative stress, regained BDNF and GFAP gene methylation, and ameliorated structural changes in the frontal cortices of isolated folic acid treated rats. Novelty: Neonatal isolation induces "autistic-like" behavior and these behaviors are reversed by folic acid supplementation. Neonatal isolation induces DNA hypomethylation of BDNF and GFAP, increased oxidative stress markers, and neuroinflammation. All of these changes were reversed by daily folic acid supplementation.
Collapse
Affiliation(s)
- Omyma G Ahmed
- Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ghaydaa A Shehata
- Neurology and Psychiatry Department, Faculty of Medicine, Assiut University, Egypt
| | - Rasha M Ali
- Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Rania Makboul
- Pathology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Eman S H Abd Allah
- Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Nessren M Abd El-Rady
- Medical Physiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
18
|
Duffy HBD, Roth TL. Increases in Bdnf DNA Methylation in the Prefrontal Cortex Following Aversive Caregiving Are Reflected in Blood Tissue. Front Hum Neurosci 2020; 14:594244. [PMID: 33324186 PMCID: PMC7721665 DOI: 10.3389/fnhum.2020.594244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022] Open
Abstract
Child maltreatment not only leads to epigenetic changes, but also increases the risk of related behavioral deficits and mental disorders. These issues presumably are most closely associated with epigenetic changes in the brain, but epigenetic changes in peripheral tissues like blood are often examined instead, due to their accessibility. As such, the reliability of using the peripheral epigenome as a proxy for that of the brain is imperative. Previously, our lab has found aberrant methylation at the Brain-derived neurotrophic factor (Bdnf) gene in the prefrontal cortex of rats following aversive caregiving. The current study examined whether aversive caregiving alters Bdnf DNA methylation in the blood compared to the prefrontal cortex. It was revealed that DNA methylation associated with adversity increased in both tissues, but this methylation was not correlated between tissues. These findings indicate that group trends in Bdnf methylation between blood and the brain are comparable, but variation exists among individual subjects.
Collapse
Affiliation(s)
- Hannah B D Duffy
- 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
|
19
|
Prenatal maternal stress is associated with increased sensitivity to neuropathic pain and sex-specific changes in supraspinal mRNA expression of epigenetic- and stress-related genes in adulthood. Behav Brain Res 2020; 380:112396. [DOI: 10.1016/j.bbr.2019.112396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
|
20
|
Total DNA methylation in the brain in response to decitabine treatment in female rats. EUROPEAN PHARMACEUTICAL JOURNAL 2019. [DOI: 10.2478/afpuc-2019-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Hypomethylating agent decitabine is being used in the treatment of certain types of leukaemia in combination with other anticancer drugs. Aberrant DNA methylation has been suggested to occur in pathological states including depression. Scarce data in male rats suggest antidepressant effects of decitabine. The main aim of our studies is to test the hypothesis that the inhibition of DNA methylation results in antidepressant effects in female rats. Before doing so, we decided to verify the effects of decitabine on DNA methylation in females. The findings demonstrate that the treatment with decitabine at the dose shown previously to inhibit DNA methylation in males, had no effect on total DNA methylation in two brain regions, namely the hippocampus and frontal cortex of female rats. In conclusion, the present study allows us to suggest that the effect of decitabine on DNA methylation in the brain is sex dependent.
Collapse
|
21
|
Torres-Berrío A, Issler O, Parise EM, Nestler EJ. Unraveling the epigenetic landscape of depression: focus on early life stress
. DIALOGUES IN CLINICAL NEUROSCIENCE 2019; 21:341-357. [PMID: 31949402 PMCID: PMC6952747 DOI: 10.31887/dcns.2019.21.4/enestler] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Depression is a devastating psychiatric disorder caused by a combination of genetic predisposition and life events, mainly exposure to stress. Early life stress (ELS) in particular is known to "scar" the brain, leading to an increased susceptibility to developing depression later in life via epigenetic mechanisms. Epigenetic processes lead to changes in gene expression that are not due to changes in DNA sequence, but achieved via modulation of chromatin modifications, DNA methylation, and noncoding RNAs. Here we review common epigenetic mechanisms including the enzymes that take part in reading, writing, and erasing specific epigenetic marks. We then describe recent developments in understanding how ELS leads to changes in the epigenome that are manifested in increased susceptibility to depression-like abnormalities in animal models. We conclude with highlighting the need for future studies that will potentially enable the utilisation of the understanding of epigenetic changes linked to ELS for the development of much-needed novel therapeutic strategies and biomarker discovery.
.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Orna Issler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric M Parise
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Eric J Nestler
- Author affiliations: Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, US
| |
Collapse
|
22
|
Modelling resilience in adolescence and adversity: a novel framework to inform research and practice. Transl Psychiatry 2019; 9:316. [PMID: 31772187 PMCID: PMC6879584 DOI: 10.1038/s41398-019-0651-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 10/24/2019] [Accepted: 11/06/2019] [Indexed: 02/07/2023] Open
Abstract
Recent conceptualisations of resilience have advanced the notion that it is a dynamic and multifaceted construct. However, its adaptive components, especially those forged by adversity, have not been fully realised, and its neurobiological and psychosocial underpinnings are yet to be meaningfully integrated. In part, this is because a developmental perspective is often neglected in the formulation of resilience. In this review, we consider the findings of resilience research, with a specific emphasis on the developmental period of adolescence. To bridge the gaps in our current understanding, we propose a model of resilience that is predicated on experiencing adversity. Specifically, our model provides a sophisticated insight into the components of resilience, which, together with intrinsic features, involves facilitation of, and skill acquisition via strengthening processes we term tempering and fortification. The model also points to the potential trajectories of adversity-driven resilience and forms the basis of a framework that allows for individual variance in resilience, and the identification of both neurobiological and psychosocial targets for prevention and therapeutic interventions.
Collapse
|
23
|
Holmes L, Shutman E, Chinaka C, Deepika K, Pelaez L, Dabney KW. Aberrant Epigenomic Modulation of Glucocorticoid Receptor Gene (NR3C1) in Early Life Stress and Major Depressive Disorder Correlation: Systematic Review and Quantitative Evidence Synthesis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16214280. [PMID: 31689998 PMCID: PMC6861987 DOI: 10.3390/ijerph16214280] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022]
Abstract
Early life stress (ELS) induced by psychological trauma, child maltreatment, maternal separation, and domestic violence predisposes to psycho-behavioral pathologies during adulthood, namely major depressive disorder (MDD), anxiety, and bipolar affective disorder. While environmental data are available in illustrating this association, data remain to be established on the epigenomic underpinning of the nexus between ELS and MDD predisposition. Specifically, despite the observed aberrant epigenomic modulation of the NR3C1, a glucocorticoid receptor gene, in early social adversity and social threats in animal and human models, reliable scientific data for intervention mapping in reducing social adversity and improving human health is required. We sought to synthesize the findings of studies evaluating (a) epigenomic modulations, mainly DNA methylation resulting in MDD following ELS, (b) epigenomic modifications associated with ELS, and (c) epigenomic alterations associated with MDD. A systematic review and quantitative evidence synthesis (QES) were utilized with the random effect meta-analytic procedure. The search strategy involved both the PubMed and hand search of relevant references. Of the 1534 studies identified through electronic search, 592 studies were screened, 11 met the eligibility criteria for inclusion in the QES, and 5 examined ELS and MDD; 4 studies assessed epigenomic modulation and ELS, while 2 studies examined epigenomic modulations and MDD. The dense DNA methylation of the 1F exon of the NR3C1, implying the hypermethylated region of the glucocorticoid receptor gene, was observed in the nexus between ELS and MDD, common effect size (CES) = 14.96, 95%CI, 10.06-19.85. With respect to epigenomic modulation associated with child ELS, hypermethylation was observed, CES = 23.2%, 95%CI, 8.00-38.48. In addition, marginal epigenomic alteration was indicated in MDD, where hypermethylation was associated with increased risk of MDD, CES = 2.12%, 95%CI, -0.63-4.86. Substantial evidence supports the implication of NR3C1 and environmental interaction, mainly DNA methylation, in the predisposition to MDD following ELS. This QES further supports aberrant epigenomic modulation identified in ELS as well as major depressive episodes involving dysfunctional glucocorticoid-mediated negative feedback as a result of allostatic overload. These findings recommend prospective investigation of social adversity and its predisposition to the MDD epidemic via aberrant epigenomic modulation. Such data will facilitate early intervention mapping in reducing MDD in the United States population.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA.
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Emily Shutman
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, Haverford College, Haverford, PA 19041, USA.
| | - Chinacherem Chinaka
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Department of public health, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
- Community Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA.
| | - Kerti Deepika
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Lavisha Pelaez
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Kirk W Dabney
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
24
|
Doherty TS, Bozeman AL, Roth TL, Brumley MR. DNA methylation and behavioral changes induced by neonatal spinal transection. Infant Behav Dev 2019; 57:101381. [PMID: 31557646 PMCID: PMC6878986 DOI: 10.1016/j.infbeh.2019.101381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/15/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023]
Abstract
Although the importance of epigenetic mechanisms in behavioral development has been gaining attention in recent years, research has largely focused on the brain. To our knowledge, no studies to date have investigated epigenetic changes in the developing spinal cord to determine the dynamic manner in which the spinal epigenome may respond to environmental input during behavioral development. Animal studies demonstrate that spinal cord plasticity is heightened during early development, is somewhat preserved following neonatal transection, and that spinal injured animals are responsive to sensory feedback. Because epigenetic alterations have been implicated in brain plasticity and are highly responsive to experience, these alterations are promising candidates for molecular substrates of spinal plasticity as well. Thus, the current study investigated behavioral changes in the development of weight-bearing locomotion and epigenetic modifications in the spinal cord of infant rats following a neonatal low-thoracic spinal transection or sham surgery on postnatal day (P)1. Specifically, global levels of methylation and methylation status of the brain-derived neurotrophic factor (Bdnf) gene, a neurotrophin heavily involved in both CNS and behavioral plasticity, particularly in development, were examined in lumbar tissue harvested on P10 from sham and spinal-transected subjects. Behavioral results demonstrate that compared to shams, spinal-transected subjects exhibit significantly reduced partial-weight bearing hindlimb activity. Molecular data demonstrate group differences in global lumbar methylation levels as well as exon-specific group differences in Bdnf methylation. This study represents an initial step toward understanding the relationship between epigenetic mechanisms and plasticity associated with spinal cord and locomotor development.
Collapse
Affiliation(s)
- Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, United States
| | - Aimee L Bozeman
- Department of Psychology, Idaho State University, Pocatello, ID, 83209, United States
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, United States
| | - Michele R Brumley
- Department of Psychology, Idaho State University, Pocatello, ID, 83209, United States.
| |
Collapse
|
25
|
Holmes L, Chinaka C, Elmi H, Deepika K, Pelaez L, Enwere M, Akinola OT, Dabney KW. Implication of Spiritual Network Support System in Epigenomic Modulation and Health Trajectory. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E4123. [PMID: 31717711 PMCID: PMC6862316 DOI: 10.3390/ijerph16214123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022]
Abstract
With challenges in understanding the multifactorial etiologies of disease and individual treatment effect heterogeneities over the past four decades, much has been acquired on how physical, chemical and social environments affect human health, predisposing certain subpopulations to adverse health outcomes, especially the socio-environmentally disadvantaged (SED). Current translational data on gene and adverse environment interaction have revealed how adverse gene-environment interaction, termed aberrant epigenomic modulation, translates into impaired gene expression via messenger ribonucleic acid (mRNA) dysregulation, reflecting abnormal protein synthesis and hence dysfunctional cellular differentiation and maturation. The environmental influence on gene expression observed in most literature includes physical, chemical, physicochemical and recently social environment. However, data are limited on spiritual or religious environment network support systems, which reflect human psychosocial conditions and gene interaction. With this limited information, we aimed to examine the available data on spiritual activities characterized by prayers and meditation for a possible explanation of the nexus between the spiritual network support system (SNSS) as a component of psychosocial conditions, implicated in social signal transduction, and the gene expression correlate. With the intent to incorporate SNSS in human psychosocial conditions, we assessed the available data on bereavement, loss of spouse, loneliness, social isolation, low socio-economic status (SES), chronic stress, low social status, social adversity (SA) and early life stress (ELS), as surrogates for spiritual support network connectome. Adverse human psychosocial conditions have the tendency for impaired gene expression through an up-regulated conserved transcriptional response to adversity (CTRA) gene expression via social signal transduction, involving the sympathetic nervous system (SNS), beta-adrenergic receptors, the hypothalamus-pituitary-adrenal (HPA) axis and the glucocorticoid response. This review specifically explored CTRA gene expression and the nuclear receptor subfamily 3 group C member 1 (NR3C1) gene, a glucocorticoid receptor gene, in response to stress and the impaired negative feedback, given allostatic overload as a result of prolonged and sustained stress and social isolation as well as the implied social interaction associated with religiosity. While more remains to be investigated on psychosocial and immune cell response and gene expression, current data on human models do implicate appropriate gene expression via the CTRA and NR3C1 gene in the SNSS as observed in meditation, yoga and thai-chi, implicated in malignant neoplasm remission. However, prospective epigenomic studies in this context are required in the disease causal pathway, prognosis and survival, as well as cautious optimism in the application of these findings in clinical and public health settings, due to unmeasured and potential confoundings implicated in these correlations.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Chinacherem Chinaka
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Hikma Elmi
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Kerti Deepika
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Lavisha Pelaez
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Michael Enwere
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Walden University, Minneapolis, MN 55401, USA
| | - Olumuyiwa T. Akinola
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Kirk W. Dabney
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
26
|
BDNF genetic variants and methylation: effects on cognition in major depressive disorder. Transl Psychiatry 2019; 9:265. [PMID: 31636250 PMCID: PMC6803763 DOI: 10.1038/s41398-019-0601-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/29/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) gene regulation has been linked to the pathophysiology of major depressive disorder (MDD). MDD patients show cognitive deficits, and altered BDNF regulation has a relevant role in neurocognitive functions. Our goal was to explore the association between BDNF genetic and epigenetic variations with neurocognitive performance in a group of MDD patients and healthy controls considering possible modulating factors. The sample included 134 subjects, 64 MDD patients, and 70 healthy controls. Clinical data, childhood maltreatment, and neurocognitive performance were assessed in all participants. Eleven single nucleotide polymorphisms (SNPs) and two promoter regions in the BDNF gene were selected for genotype and methylation analysis. The role of interactions between BDNF genetic and epigenetic variations with MDD diagnosis, sex, and Childhood Trauma Questionnaire (CTQ) scores was also explored. We observed significant associations between neurocognitive performance and two BDNF SNPs (rs908867 and rs925946), an effect that was significantly mediated by methylation values at specific promoter I sites. We identified significant associations between neurocognitive results and methylation status as well as its interactions with MDD diagnosis, sex, and CTQ scores. Our results support the hypothesis that BDNF gene SNPs and methylation status, as well as their interactions with modulating factors, can influence cognition. Further studies are required to confirm the effect of BDNF variations and cognitive function in larger samples.
Collapse
|
27
|
McKee SE, Reyes TM. Effect of supplementation with methyl-donor nutrients on neurodevelopment and cognition: considerations for future research. Nutr Rev 2019; 76:497-511. [PMID: 29701796 DOI: 10.1093/nutrit/nuy007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pregnancy represents a critical period in fetal development, such that the prenatal environment can, in part, establish a lifelong trajectory of health or disease for the offspring. Poor nutrition (macro- or micronutrient deficiencies) can adversely affect brain development and significantly increase offspring risk for metabolic and neurological disease development. The concentration of dietary methyl-donor nutrients is known to alter DNA methylation in the brain, and alterations in DNA methylation can have long-lasting effects on gene expression and neuronal function. The decreased availability of methyl-donor nutrients to the developing fetus in models of poor maternal nutrition is one mechanism hypothesized to link maternal malnutrition and disease risk in offspring. Animal studies indicate that supplementation of both maternal and postnatal (early- and later-life) diets with methyl-donor nutrients can attenuate disease risk in offspring; however, clinical research is more equivocal. The objective of this review is to summarize how specific methyl-donor nutrient deficiencies and excesses during pre- and postnatal life alter neurodevelopment and cognition. Emphasis is placed on reviewing the current literature, highlighting challenges within nutrient supplementation research, and considering potential strategies to ensure robust findings in future studies.
Collapse
Affiliation(s)
- Sarah E McKee
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa M Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Keller SM, Nowak A, Roth TL. Female pups receive more maltreatment from stressed dams. Dev Psychobiol 2019; 61:824-831. [PMID: 30810229 PMCID: PMC6711830 DOI: 10.1002/dev.21834] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 01/10/2023]
Abstract
The effects of exposure to developmental stress often diverge for males and females. Using the scarcity-adversity model of low nesting resources outside the home cage, our lab has discovered sex differences in both behavioral and epigenetic consequences of repeated exposure to caregiver maltreatment. For the measures we have performed to date, we have found more consequences for females. The reasons underlying this sex disparity are unknown. In the current experiment, we aimed to discern the quality of maternal care received by male and female pups in our model. As we have previously found more behavioral and epigenetic consequences in females, we hypothesized that females receive more adverse care compared to their male littermates. Our hypothesis was supported; in our maltreatment condition, we found that female pups received more adverse care than males. This sex difference in adverse care was not present in our two control conditions (cross-foster and normal maternal care). These data lend support to the notion that one reason females in our model incur more behavioral and epigenetic consequences is a result of greater mistreatment by the dam.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Anna Nowak
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
29
|
Orso R, Creutzberg KC, Wearick-Silva LE, Wendt Viola T, Tractenberg SG, Benetti F, Grassi-Oliveira R. How Early Life Stress Impact Maternal Care: A Systematic Review of Rodent Studies. Front Behav Neurosci 2019; 13:197. [PMID: 31555106 PMCID: PMC6724664 DOI: 10.3389/fnbeh.2019.00197] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Maternal care refers to the behavior performed by the dam to nourish and protect her litter during its early development. Frequent and high-quality performance of such maternal behaviors is critical for the neurodevelopment of the pups. Maternal exposure to stress during early development can impair maternal care and amplify the deleterious effects of poor maternal caregiving and neglect. As such, a thorough understanding of the effects caused by several models of early life stress on maternal care may yield more insights into the relationship between stress and maternal behavior. Methods: A systematic review was performed to identify and address the effects of early life stress on maternal behavior. The search was conducted using three online databases: PUBMED, Embase, and Web of Science. To provide clear evidence of the impact of stress on maternal care, in every study, the stress group was always compared to a control group. Outcomes were categorized into eight different behaviors: (1) licking/grooming; (2) arched-back nursing; (3) blanket-nursing/passive nursing; (4) nest building; (5) contact with pups; (6) harmful/adverse caregiving; (7) no contact; (8) nest exits. Additionally, the methodological quality of the studies was evaluated. Results: A total of 12 different early life stress protocols were identified from the 56 studies included in this systematic review. Our data demonstrate that different stress models can promote specific maternal patterns of behavior. Regarding the maternal separation protocol, we observed an overall increase in nursing and licking/grooming behaviors, which are essential for pup development. An increase in the number of nest exits, which represents a fragmentation of maternal care, was observed in the limited bedding protocol, but the total amount of maternal care appears to remain similar between groups. Conclusions: Each stress protocol has unique characteristics that increase the difficulty of rendering comparisons of maternal behavior. The increase in maternal care observed in the maternal separation protocol may be an attempt to overcompensate for the time off-nest. Fragmented maternal care is a key component of the limited bedding protocol. Moreover, the methodological approaches to evaluate maternal behavior, such as time, duration, and behavior type should be more homogeneous across studies.
Collapse
Affiliation(s)
- Rodrigo Orso
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Kerstin Camile Creutzberg
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Luis Eduardo Wearick-Silva
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Thiago Wendt Viola
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Saulo Gantes Tractenberg
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Fernando Benetti
- Laboratório de Neurofisiologia Cognitiva e do Desenvolvimento, Department of Physiology, Instituto de Ciências Básicas da Saúde, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab (DCNL), School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Brain Institute (Instituto do Cérebro), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
30
|
Keller SM, Doherty TS, Roth TL. Pharmacological manipulation of DNA methylation normalizes maternal behavior, DNA methylation, and gene expression in dams with a history of maltreatment. Sci Rep 2019; 9:10253. [PMID: 31311968 PMCID: PMC6635500 DOI: 10.1038/s41598-019-46539-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/25/2019] [Indexed: 01/02/2023] Open
Abstract
The quality of parental care received during development profoundly influences an individual's phenotype, including that of maternal behavior. We previously found that female rats with a history of maltreatment during infancy mistreat their own offspring. One proposed mechanism through which early-life experiences influence behavior is via epigenetic modifications. Indeed, our lab has identified a number of brain epigenetic alterations in female rats with a history of maltreatment. Here we sought to investigate the role of DNA methylation in aberrant maternal behavior. We administered zebularine, a drug known to alter DNA methylation, to dams exposed during infancy to the scarcity-adversity model of low nesting resources, and then characterized the quality of their care towards their offspring. First, we replicate that dams with a history of maltreatment mistreat their own offspring. Second, we show that maltreated-dams treated with zebularine exhibit lower levels of adverse care toward their offspring. Third, we show that administration of zebularine in control dams (history of nurturing care) enhances levels of adverse care. Lastly, we show altered methylation and gene expression in maltreated dams normalized by zebularine. These findings lend support to the hypothesis that epigenetic alterations resulting from maltreatment causally relate to behavioral outcomes.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
31
|
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]
|
32
|
Preventing epigenetic traces of caregiver maltreatment: A role for HDAC inhibition. Int J Dev Neurosci 2019; 78:178-184. [PMID: 31075305 DOI: 10.1016/j.ijdevneu.2019.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/24/2019] [Accepted: 05/06/2019] [Indexed: 01/07/2023] Open
Abstract
Reorganization of the brain's epigenetic landscape occurs alongside early adversity in both human and non-human animals. Whether this reorganization is simply incidental to or is a causal mechanism of the behavioral abnormalities that result from early adversity is important to understand. Using the scarcity-adversity model of low nesting resources in Long Evans rats, our lab has previously reported specific epigenetic and behavioral trajectories occurring in response to early disruption of the caregiving environment. To further probe that relationship, the current work investigates the ability of the epigenome-modifying drug sodium butyrate to prevent maltreatment-induced methylation changes when administered alongside maltreatment. Following exposure to the scarcity-adversity model, during which drug was administered prior to each caregiving session, methylation of Brain-derived Neurotrophic Factor (Bdnf) IX DNA was examined in the Prefrontal Cortex (PFC) of male and female pups at postnatal day (PN) 8. As our previous work reports, increased methylation at this exon of Bdnf in the PFC is a stable epigenetic change across the lifespan that occurs in response to early maltreatment, thus giving us a suitable starting point to investigate pharmacological prevention of maltreatment-induced epigenetic marks. Here we also examined off-target effects of sodium butyrate by assessing methylation in another region of Bdnf (exon IV) not affected in the infant brain as well as global levels of methylation in the brain region of interest. Results indicate that a 400 mg/kg (but not 300 mg/kg) dose of sodium butyrate is effective in preventing the maltreatment-induced rise in methylation at Bdnf exon IX in the PFC of male (but not female) infant pups. Administration of sodium butyrate did not affect the methylation status of Bdnf IV or overall levels of global methylation in the PFC, suggesting potential specificity of this drug. These data provide us an avenue forward for investigating whether the relationship between adversity-induced epigenetic outcomes in our model can be manipulated to improve behavioral outcomes.
Collapse
|
33
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
34
|
Chistiakov DA, Chekhonin VP. Early-life adversity-induced long-term epigenetic programming associated with early onset of chronic physical aggression: Studies in humans and animals. World J Biol Psychiatry 2019; 20:258-277. [PMID: 28441915 DOI: 10.1080/15622975.2017.1322714] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objectives: To examine whether chronic physical aggression (CPA) in adulthood can be epigenetically programmed early in life due to exposure to early-life adversity. Methods: Literature search of public databases such as PubMed/MEDLINE and Scopus. Results: Children/adolescents susceptible for CPA and exposed to early-life abuse fail to efficiently cope with stress that in turn results in the development of CPA later in life. This phenomenon was observed in humans and animal models of aggression. The susceptibility to aggression is a complex trait that is regulated by the interaction between environmental and genetic factors. Epigenetic mechanisms mediate this interaction. Subjects exposed to stress early in life exhibited long-term epigenetic programming that can influence their behaviour in adulthood. This programming affects expression of many genes not only in the brain but also in other systems such as neuroendocrine and immune. Conclusions: The propensity to adult CPA behaviour in subjects experienced to early-life adversity is mediated by epigenetic programming that involves long-term systemic epigenetic alterations in a whole genome.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Vladimir P Chekhonin
- a Department of Fundamental and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia.,b Department of Medical Nanobiotechnology , Pirogov Russian State Medical University (RSMU) , Moscow , Russia
| |
Collapse
|
35
|
Tiwari A, Gonzalez A. Biological alterations affecting risk of adult psychopathology following childhood trauma: A review of sex differences. Clin Psychol Rev 2018; 66:69-79. [DOI: 10.1016/j.cpr.2018.01.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 01/10/2023]
|
36
|
Watamura SE, Roth TL. Looking back and moving forward: Evaluating and advancing translation from animal models to human studies of early life stress and DNA methylation. Dev Psychobiol 2018; 61:323-340. [PMID: 30426484 DOI: 10.1002/dev.21796] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/06/2018] [Accepted: 09/25/2018] [Indexed: 01/08/2023]
Abstract
Advances in epigenetic methodologies have deepened theoretical explanations of mechanisms linking early life stress (ELS) and disease outcomes and suggest promising targets for intervention. To date, however, human studies have not capitalized on the richness of diverse animal models to derive and systematically evaluate specific and testable hypotheses. To promote cross-species dialog and scientific advance, here we provide a classification scheme to systematically evaluate the match between characteristics of human and animal studies of ELS and DNA methylation. Three preclinical models were selected that are highly cited, and that differ in the nature and severity of the ELS manipulation as well as in the affected epigenetic loci (the licking and grooming, maternal separation, and caregiver maltreatment models). We evaluated the degree to which human studies matched these preclinical models with respect to the timing of ELS and of DNA methylation assessment, as well as the type of ELS, whether sex differences were explicitly examined, the tissue sampled, and the targeted loci. Results revealed <50% match (range of 8-83%) between preclinical models and human work on these variables. Immediate and longer-term suggestions to improve translational specificity are offered, with the goal of accelerating scientific advance.
Collapse
Affiliation(s)
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
37
|
Neurobiology of Infant Sensitive Period for Attachment and Its Reinstatement Through Maternal Social Buffering. MINNESOTA SYMPOSIA ON CHILD PSYCHOLOGY 2018. [DOI: 10.1002/9781119461746.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
38
|
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
|
39
|
Burns SB, Szyszkowicz JK, Luheshi GN, Lutz PE, Turecki G. Plasticity of the epigenome during early-life stress. Semin Cell Dev Biol 2018; 77:115-132. [DOI: 10.1016/j.semcdb.2017.09.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/08/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022]
|
40
|
Doherty TS, Roth TL. Epigenetic Landscapes of the Adversity-Exposed Brain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:1-19. [PMID: 29933946 DOI: 10.1016/bs.pmbts.2017.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
It is understood that adversity during development has the power to alter behavioral trajectories, and the role of the epigenome in that relationship is currently under intense investigation. Several studies in both nonhuman animals and humans have established a link between early adversity and epigenetic regulation of genes heavily implicated in the stress response, plasticity and cognition, and psychiatric disorders such as depression and anxiety. Thus the relatively recent surge of studies centering on the epigenetic outcomes of stress has great potential to inform treatments and interventions for psychiatric disorder precipitated by early adversity. Here we review what we know and what we do not know, and suggest approaches to help further elucidate the relationship between early adversity, epigenetics, and behavior.
Collapse
Affiliation(s)
| | - Tania L Roth
- University of Delaware, Newark, DE, United States.
| |
Collapse
|
41
|
Kundakovic M. Sex-Specific Epigenetics: Implications for Environmental Studies of Brain and Behavior. Curr Environ Health Rep 2018; 4:385-391. [PMID: 28986864 DOI: 10.1007/s40572-017-0172-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW This review discusses the current state of knowledge on sex differences in the epigenetic regulation in the brain and highlights its relevance for the environmental studies of brain and behavior. RECENT FINDINGS Recent evidence shows that epigenetic mechanisms are involved in the control of brain sexual differentiation and in memory-enhancing effects of estradiol in females. In addition, several studies have implicated epigenetic dysregulation as an underlying mechanism for sex-specific neurobehavioral effects of environmental exposures. The area of sex-specific neurepigenetics has a great potential to improve our understanding of brain function in health and disease. Future neuropigenetic studies will require the inclusion of males and females and would ideally account for the fluctuating hormonal status in females which is likely to affect the epigenome. The implementation of cutting-edge methods that include epigenomic characterization of specific cell types using latest next-generation sequencing approaches will further advance the area.
Collapse
Affiliation(s)
- Marija Kundakovic
- Department of Biological Sciences, Fordham University, 441 E. Fordham Road, Larkin Hall, Room 160, Bronx, NY, 10458, USA.
| |
Collapse
|
42
|
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
|
43
|
Barnett Burns S, Almeida D, Turecki G. The Epigenetics of Early Life Adversity: Current Limitations and Possible Solutions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:343-425. [DOI: 10.1016/bs.pmbts.2018.01.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Blaze J, Asok A, Borrelli K, Tulbert C, Bollinger J, Ronca AE, Roth TL. Intrauterine exposure to maternal stress alters Bdnf IV DNA methylation and telomere length in the brain of adult rat offspring. Int J Dev Neurosci 2017; 62:56-62. [PMID: 28330827 PMCID: PMC5600826 DOI: 10.1016/j.ijdevneu.2017.03.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 12/16/2022] Open
Abstract
DNA methylation (addition of methyl groups to cytosines) and changes in telomere length (TTAGGG repeats on the ends of chromosomes) are two molecular modifications that result from stress and could contribute to the long-term effects of intrauterine exposure to maternal stress on offspring behavior. Here, we measured methylation of DNA associated with the Brain-derived neurotrophic factor (Bdnf) gene, a gene important in development and plasticity, and telomere length in the brains of adult rat male and female offspring whose mothers were exposed to unpredictable and variable stressors throughout gestation. Males exposed to prenatal stress had greater methylation (Bdnf IV) in the medial prefrontal cortex (mPFC) compared to non-stressed male controls and stressed females. Further, prenatally-stressed animals had shorter telomeres than controls in the mPFC. Together findings indicate a long-term impact of prenatal stress on brain DNA methylation and telomere biology with relevance for behavioral and health outcomes, and contribute to a growing literature linking stress to intergenerational molecular changes.
Collapse
Affiliation(s)
- Jennifer Blaze
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE,United States
| | - Arun Asok
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE,United States
| | - Kristyn Borrelli
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE,United States
| | - Christina Tulbert
- Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Justin Bollinger
- Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - April E Ronca
- Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, United States; Program in Neuroscience, Wake Forest School of Medicine, Winston-Salem, NC, United States; Space Biosciences Research Division, NASA Ames Research Center, Moffett Field, CA
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE,United States.
| |
Collapse
|
45
|
|
46
|
Raineki C, Bodnar TS, Holman PJ, Baglot SL, Lan N, Weinberg J. Effects of early-life adversity on immune function are mediated by prenatal environment: Role of prenatal alcohol exposure. Brain Behav Immun 2017; 66:210-220. [PMID: 28698116 PMCID: PMC5650917 DOI: 10.1016/j.bbi.2017.07.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/06/2017] [Accepted: 07/03/2017] [Indexed: 01/07/2023] Open
Abstract
The contribution of the early postnatal environment to the pervasive effects of prenatal alcohol exposure (PAE) is poorly understood. Moreover, PAE often carries increased risk of exposure to adversity/stress during early life. Dysregulation of immune function may play a role in how pre- and/or postnatal adversity/stress alters brain development. Here, we combine two animal models to examine whether PAE differentially increases vulnerability to immune dysregulation in response to early-life adversity. PAE and control litters were exposed to either limited bedding (postnatal day [PN] 8-12) to model early-life adversity or normal bedding, and maternal behavior and pup vocalizations were recorded. Peripheral (serum) and central (amygdala) immune (cytokines and C-reactive protein - CRP) responses of PAE animals to early-life adversity were evaluated at PN12. Insufficient bedding increased negative maternal behavior in both groups. Early-life adversity increased vocalization in all animals; however, PAE pups vocalized less than controls. Early-life adversity reduced serum TNF-α, KC/GRO, and IL-10 levels in control but not PAE animals. PAE increased serum CRP, and levels were even higher in pups exposed to adversity. Finally, PAE reduced KC/GRO and increased IL-10 levels in the amygdala. Our results indicate that PAE alters immune system development and both behavioral and immune responses to early-life adversity, which could have subsequent consequences for brain development and later life health.
Collapse
Affiliation(s)
- Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada.
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Parker J Holman
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Samantha L Baglot
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Ni Lan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
47
|
Doherty TS, Blaze J, Keller SM, Roth TL. Phenotypic outcomes in adolescence and adulthood in the scarcity-adversity model of low nesting resources outside the home cage. Dev Psychobiol 2017; 59:703-714. [PMID: 28767135 PMCID: PMC5569321 DOI: 10.1002/dev.21547] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 06/27/2017] [Indexed: 01/10/2023]
Abstract
Early life adversity is known to disrupt behavioral trajectories and many rodent models have been developed to characterize these stress-induced outcomes. One example is the scarcity-adversity model of low nesting resources. This model employs resource scarcity (i.e., low nesting materials) to elicit adverse caregiving conditions (including maltreatment) toward rodent neonates. Our lab utilizes a version of this model wherein caregiving exposures occur outside the home cage during the first postnatal week. The aim of this study was to determine adolescent and adult phenotypic outcomes associated with this model, including assessment of depressive- and anxiety-like behaviors and performance in different cognitive domains. Exposure to adverse caregiving had no effect on adolescent behavioral performance whereas exposure significantly impaired adult behavioral performance. Further, adult behavioral assays revealed substantial differences between sexes. Overall, data demonstrate the ability of repeated exposure to brief bouts of maltreatment outside the home cage in infancy to impact the development of several behavioral domains later in life.
Collapse
Affiliation(s)
- Tiffany S. Doherty
- Department of Psychological and Brain Sciences, University of Delaware,
Newark DE 19716
| | | | | | - Tania L. Roth
- Department of Psychological and Brain Sciences, University of Delaware,
Newark DE 19716
| |
Collapse
|
48
|
Walker CD, Bath KG, Joels M, Korosi A, Larauche M, Lucassen PJ, Morris MJ, Raineki C, Roth TL, Sullivan RM, Taché Y, Baram TZ. Chronic early life stress induced by limited bedding and nesting (LBN) material in rodents: critical considerations of methodology, outcomes and translational potential. Stress 2017; 20:421-448. [PMID: 28617197 PMCID: PMC5705407 DOI: 10.1080/10253890.2017.1343296] [Citation(s) in RCA: 263] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 02/07/2023] Open
Abstract
The immediate and long-term effects of exposure to early life stress (ELS) have been documented in humans and animal models. Even relatively brief periods of stress during the first 10 days of life in rodents can impact later behavioral regulation and the vulnerability to develop adult pathologies, in particular an impairment of cognitive functions and neurogenesis, but also modified social, emotional, and conditioned fear responses. The development of preclinical models of ELS exposure allows the examination of mechanisms and testing of therapeutic approaches that are not possible in humans. Here, we describe limited bedding and nesting (LBN) procedures, with models that produce altered maternal behavior ranging from fragmentation of care to maltreatment of infants. The purpose of this paper is to discuss important issues related to the implementation of this chronic ELS procedure and to describe some of the most prominent endpoints and consequences, focusing on areas of convergence between laboratories. Effects on the hypothalamic-pituitary adrenal (HPA) axis, gut axis and metabolism are presented in addition to changes in cognitive and emotional functions. Interestingly, recent data have suggested a strong sex difference in some of the reported consequences of the LBN paradigm, with females being more resilient in general than males. As both the chronic and intermittent variants of the LBN procedure have profound consequences on the offspring with minimal external intervention from the investigator, this model is advantageous ecologically and has a large translational potential. In addition to the direct effect of ELS on neurodevelopmental outcomes, exposure to adverse early environments can also have intergenerational impacts on mental health and function in subsequent generation offspring. Thus, advancing our understanding of the effect of ELS on brain and behavioral development is of critical concern for the health and wellbeing of both the current population, and for generations to come.
Collapse
Affiliation(s)
- Claire-Dominique Walker
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 Lasalle Blvd, Montreal, QC H4H 1R3, Canada
| | - Kevin G. Bath
- Department of Neuroscience, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Marian Joels
- Department Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Aniko Korosi
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Paul J. Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, NSW, Australia
| | - Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, Department of Child and Adolescent Psychiatry, New York University Langone Medical School, New York, NY 10016, USA
| | - Tania L. Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, Department of Child and Adolescent Psychiatry, New York University Langone Medical School, New York, NY 10016, USA
| | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Tallie Z. Baram
- Department of Pediatrics, of Anatomy & Neurobiology and of Neurology, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
49
|
BECon: a tool for interpreting DNA methylation findings from blood in the context of brain. Transl Psychiatry 2017; 7:e1187. [PMID: 28763057 PMCID: PMC5611738 DOI: 10.1038/tp.2017.171] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/14/2017] [Accepted: 06/17/2017] [Indexed: 12/11/2022] Open
Abstract
Tissue differences are one of the largest contributors to variability in the human DNA methylome. Despite the tissue-specific nature of DNA methylation, the inaccessibility of human brain samples necessitates the frequent use of surrogate tissues such as blood, in studies of associations between DNA methylation and brain function and health. Results from studies of surrogate tissues in humans are difficult to interpret in this context, as the connection between blood-brain DNA methylation is tenuous and not well-documented. Here, we aimed to provide a resource to the community to aid interpretation of blood-based DNA methylation results in the context of brain tissue. We used paired samples from 16 individuals from three brain regions and whole blood, run on the Illumina 450 K Human Methylation Array to quantify the concordance of DNA methylation between tissues. From these data, we have made available metrics on: the variability of cytosine-phosphate-guanine dinucleotides (CpGs) in our blood and brain samples, the concordance of CpGs between blood and brain, and estimations of how strongly a CpG is affected by cell composition in both blood and brain through the web application BECon (Blood-Brain Epigenetic Concordance; https://redgar598.shinyapps.io/BECon/). We anticipate that BECon will enable biological interpretation of blood-based human DNA methylation results, in the context of brain.
Collapse
|
50
|
Caregiver maltreatment causes altered neuronal DNA methylation in female rodents. Dev Psychopathol 2017; 29:477-489. [PMID: 28401839 DOI: 10.1017/s0954579417000128] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Negative experiences with a caregiver during infancy can result in long-term changes in brain function and behavior, but underlying mechanisms are not well understood. It is our central hypothesis that brain and behavior changes are conferred by early childhood adversity through epigenetic changes involving DNA methylation. Using a rodent model of early-life caregiver maltreatment (involving exposure to an adverse caregiving environment for postnatal days 1-7), we have previously demonstrated abnormal methylation of DNA associated with the brain-derived neurotrophic factor (Bdnf) gene in the medial prefrontal cortex (mPFC) of adult rats. The aim of the current study was to characterize Bdnf DNA methylation in specific cell populations within the mPFC. In the prefrontal cortex, there is approximately twice as many neurons as glia, and studies have recently shown differential and distinctive DNA methylation patterns in neurons versus nonneurons. Here, we extracted nuclei from the mPFC of adult animals that had experienced maltreatment and used fluorescence-activated cell sorting to isolate cell types before performing bisulfite sequencing to estimate methylation of cytosine-guanine sites. Our data indicate that early-life stress induced methylation of DNA associated with Bdnf IV in a cell-type and sex-specific manner. Specifically, females that experienced early-life maltreatment exhibited greater neuronal cytosine-guanine methylation compared to controls, while no changes were detected in Bdnf methylation in males regardless of cell type. These changes localize the specificity of our previous findings to mPFC neurons and highlight the capacity of maltreatment to cause methylation changes that are likely to have functional consequences for neuronal function.
Collapse
|