201
|
Flaherty E, Legano L, Idzerda S, Sirotnak AP, Budzak AE, Gavril AR, Haney SB, Laskey A“T, Messner SA, Moles RL, Palsuci VJ. Ongoing Pediatric Health Care for the Child Who Has Been Maltreated. Pediatrics 2019; 143:peds.2019-0284. [PMID: 30886109 DOI: 10.1542/peds.2019-0284] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pediatricians provide continuous medical care and anticipatory guidance for children who have been reported to state child protection agencies, including tribal child protection agencies, because of suspected child maltreatment. Because families may continue their relationships with their pediatricians after these reports, these primary care providers are in a unique position to recognize and manage the physical, developmental, academic, and emotional consequences of maltreatment and exposure to childhood adversity. Substantial information is available to optimize follow-up medical care of maltreated children. This new clinical report will provide guidance to pediatricians about how they can best oversee and foster the optimal physical health, growth, and development of children who have been maltreated and remain in the care of their biological family or are returned to their care by Child Protective Services agencies. The report describes the pediatrician's role in helping to strengthen families' and caregivers' capabilities and competencies and in promoting and maximizing high-quality services for their families in their community. Pediatricians should refer to other reports and policies from the American Academy of Pediatrics for more information about the emotional and behavioral consequences of child maltreatment and the treatment of these consequences.
Collapse
Affiliation(s)
- Emalee Flaherty
- Department of Pediatrics, Northwestern University, Chicago, Illinois
| | - Lori Legano
- Department of Pediatrics, School of Medicine, New York University, New York, New York; and
| | - Sheila Idzerda
- Billings Clinic Bozeman Acorn Pediatrics, Bozeman, Montana
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Karen C, Rajan KE. Social Behaviour and Epigenetic Status in Adolescent and Adult Rats: The Contribution of Early-Life Stressful Social Experience. Cell Mol Neurobiol 2019; 39:371-385. [PMID: 30710320 DOI: 10.1007/s10571-019-00655-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/24/2019] [Indexed: 01/14/2023]
Abstract
Early-life experiences have been linked to individual's epigenetic status and social behaviour. Therefore, the present study aims to test whether the presence of mother suppress the early-life stressful social experience (SSE)-induced effect on social behaviour of adolescent and adult rats, and associated epigenetic changes. To test this, experimental groups [maternally separated pups (MSP)/pups with their mother (M+P)] were allowed to experience the presence of a stranger (ST), and then their social behaviour was compared with the maternal separated (MS) and control (Con) group. We observed that MS, MSP-ST group showed less social interaction with the unknown conspecifics than known conspecifics compared to other groups. Subsequently, we found that SSE elevated the level of DNA methyltransferases (Dnmt3a), ten-eleven translocation (Tet3), methyl-CpG-binding protein-2 (MeCP2) and Repressor Element-1 Silencing Transcription Factor (REST) in amygdala of adolescent and adult MS, MSP-ST groups compared to other groups. As expected, SSE altered the histone (H3) lysine (K14/K9) acetylation (ac) and H3K4/K9 methylation (me2/me3). SSE decreased the level of H3K14ac and H3K9ac in adolescents and then increased in adults. Interestingly, H3K4me2/me3 levels were elevated in adolescent and adults. Whereas H3K9me2/me3 shows contrasting pattern in adolescent, but H3K9me2/me3 levels were increased in adults. In addition, the expression of brain-derived neurotrophic factor (BDNF) was reduced in MS, MSP-ST groups' adolescent and adult rats. Observed correlation between epigenetic changes and social behaviour possibly contributed by early-life SSE in the absence of mother, but mother's presence suppresses the effect of early-life SSE.
Collapse
Affiliation(s)
- Christopher Karen
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India.
| |
Collapse
|
203
|
Tamman AJF, Sippel LM, Han S, Neria Y, Krystal JH, Southwick SM, Gelernter J, Pietrzak RH. Attachment style moderates effects of FKBP5 polymorphisms and childhood abuse on post-traumatic stress symptoms: Results from the National Health and Resilience in Veterans Study. World J Biol Psychiatry 2019; 20:289-300. [PMID: 28891785 DOI: 10.1080/15622975.2017.1376114] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Objectives: To determine the main and interactive effects of four FKBP5 polymorphisms (rs9296158, rs3800373, rs1360780 and rs9470080), childhood abuse and attachment style in predicting severity of PTSD symptoms in two independent, nationally representative samples of US military veterans. Methods: Data were analysed from two independent samples of European-American US military veterans who participated in the National Health and Resilience in Veterans Study (N = 1,585 and 577 respectively). Results: Results revealed that carriage of two FKBP5 minor alleles, childhood abuse and insecure attachment style were associated with greater severity of PTSD symptoms. Gene × environment interactions were also observed, with the interaction of FKBP5 homozygous minor allele carriage and history of childhood abuse associated with greater severity of PTSD symptoms; however, these effects were fully counteracted by secure attachment style. Conclusions: Results of this study build on prior work demonstrating a gene × environment interaction between FKBP5 polymorphisms and childhood abuse in predicting risk for PTSD by suggesting that attachment style may moderate this effect. This study has implications for prevention and treatment efforts designed to promote a secure attachment style in veterans with high-risk FKBP5 genotypes and childhood abuse histories.
Collapse
Affiliation(s)
- Amanda J F Tamman
- a Division of Psychology and Language Sciences , University College London , London , UK.,b The PTSD Research and Treatment Program , Columbia University , New York , NY , USA.,c Anxiety Disorders Clinic , New York State Psychiatric Institute , New York , NY , USA.,d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA
| | - Lauren M Sippel
- d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA.,e Clinical Neurosciences Division , U.S. Department of Veterans Affairs National Center for PTSD, VA Connecticut Healthcare System , West Haven , CT , USA
| | - Shizhong Han
- f Department of Psychiatry , University of Iowa Carver College of Medicine , Iowa City , IA , USA
| | - Yuval Neria
- b The PTSD Research and Treatment Program , Columbia University , New York , NY , USA.,c Anxiety Disorders Clinic , New York State Psychiatric Institute , New York , NY , USA
| | - John H Krystal
- d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA.,e Clinical Neurosciences Division , U.S. Department of Veterans Affairs National Center for PTSD, VA Connecticut Healthcare System , West Haven , CT , USA
| | - Steven M Southwick
- d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA.,e Clinical Neurosciences Division , U.S. Department of Veterans Affairs National Center for PTSD, VA Connecticut Healthcare System , West Haven , CT , USA
| | - Joel Gelernter
- d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA.,e Clinical Neurosciences Division , U.S. Department of Veterans Affairs National Center for PTSD, VA Connecticut Healthcare System , West Haven , CT , USA
| | - Robert H Pietrzak
- d Department of Psychiatry , Yale University School of Medicine , New Haven , CT , USA.,e Clinical Neurosciences Division , U.S. Department of Veterans Affairs National Center for PTSD, VA Connecticut Healthcare System , West Haven , CT , USA
| |
Collapse
|
204
|
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
|
205
|
Wrigglesworth J, Ryan J, Vijayakumar N, Whittle S. Brain-derived neurotrophic factor DNA methylation mediates the association between neighborhood disadvantage and adolescent brain structure. Psychiatry Res Neuroimaging 2019; 285:51-57. [PMID: 30771753 DOI: 10.1016/j.pscychresns.2018.12.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/22/2018] [Accepted: 12/22/2018] [Indexed: 12/13/2022]
Abstract
Prior research indicates that socioeconomic disadvantage is associated with prefrontal cortical (PFC) development in childhood and adolescence, however the mechanisms of this link are unclear. This study investigated whether DNA methylation of the brain-derived neurotrophic factor (BDNF, which plays a key role in synaptic plasticity), mediated the association between neighborhood disadvantage and thickness of the PFC in adolescents. Neighborhood disadvantage was measured in 33 adolescents aged 12-13 years using the Socio-Economic Indexes for Areas. Buccal swabs, collected during mid-adolescence (aged 16-18 years), enabled BDNF DNA methylation of the widely studied exon IV promoter region to be measured. Cortical thickness was assessed during late-adolescence (aged 18-20 years) via T1-weighted magnetic resonance imaging (MRI). A significant negative association between disadvantage and BDNF DNA methylation at a specific site of the exon IV promoter was identified. Lower levels of methylation were also significantly associated with greater thickness of the lateral orbitofrontal cortex (lOFC), and right medial OFC. Lower levels of DNA methylation at this site also mediated associations between higher disadvantage and thinner bilateral lOFC thickness. These novel findings give insight into a potential biological mechanism that could further our understanding as to why brain development is affected by varying environmental exposures.
Collapse
Affiliation(s)
- Jo Wrigglesworth
- Melbourne Neuropsychiatry Center, Department of Psychiatry, The University of Melbourne and Melbourne Health, Parkville, Victoria, Australia; Disease Epigenetics, Murdoch Children's Research Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Joanne Ryan
- Disease Epigenetics, Murdoch Children's Research Institute, The University of Melbourne, Parkville, Victoria, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | | | - Sarah Whittle
- Melbourne Neuropsychiatry Center, Department of Psychiatry, The University of Melbourne and Melbourne Health, Parkville, Victoria, Australia; Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
206
|
Brown A, Fiori LM, Turecki G. Bridging Basic and Clinical Research in Early Life Adversity, DNA Methylation, and Major Depressive Disorder. Front Genet 2019; 10:229. [PMID: 30984237 PMCID: PMC6448008 DOI: 10.3389/fgene.2019.00229] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Early life adversity (ELA)- including childhood physical, emotional, and sexual abuse, as well as childhood neglect- is an important predictive factor for negative psychopathology, including Major Depressive Disorder (MDD). ELA can epigenetically regulate key emotional and behavioral systems in ways that can stably persist into adulthood and contribute to the development of MDD and other psychopathology. DNA methylation has been one of the most investigated forms of epigenetic regulation in ELA to MDD pathway. From these studies, genes and sites associated with ELA/MDD have been identified and should be further investigated in order to identify potential avenues for intervention.
Collapse
Affiliation(s)
- Amanda Brown
- McGill Group for Suicide Studies, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Laura M Fiori
- McGill Group for Suicide Studies, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
207
|
Social isolation and social support at adulthood affect epigenetic mechanisms, brain-derived neurotrophic factor levels and behavior of chronically stressed rats. Behav Brain Res 2019; 366:36-44. [PMID: 30880220 DOI: 10.1016/j.bbr.2019.03.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/20/2022]
Abstract
Epigenetic modulation of brain-derived neurotrophic factor (BDNF) provides one possible explanation for the dysfunctions induced by stress, such as psychiatric disorders and cognitive decline. Interestingly, social support can be protective against some of these effects, but the mechanisms of social buffering are poorly understood. Conversely, early isolation exacerbates the responses to stressors, although its effects in adulthood remain unclear. This study investigated the effects of social isolation and social buffering on hippocampal epigenetic mechanisms, BDNF levels and behavioral responses of chronically stressed young adult rats. Male Wistar rats (3 months) were assigned to accompanied (paired) or isolated housing. After one-month half of each group was submitted to a chronic unpredictable stress (CUS) protocol for 18 days. Among accompanied animals, only one was exposed to stress. Behavioral analysis encompassed the Open field, plus maze and inhibitory avoidance tasks. Hippocampal H3K9 and H4K12 acetylation, HDAC5 expression and BDNF levels were evaluated. Isolated housing increased HDAC5 expression, decreased H3K9 and H4K12 acetylation, reduced BDNF levels, and impaired long-term memory. Stress affected weight gain, induced anxiety-like behavior and decreased AcK9H3 levels. Interactions between housing conditions and social stress were seen only for HDAC5 expression, which showed a further increase in the isolated + CUS group but remained constant in accompanied animals. In conclusion, social isolation at adulthood induced epigenetic alterations and exacerbated the effects of chronic stress on HDAC5. Notwithstanding, social support counteracted the adverse effects of stress on HDAC5 expression.
Collapse
|
208
|
Cortes LR, Cisternas CD, Forger NG. Does Gender Leave an Epigenetic Imprint on the Brain? Front Neurosci 2019; 13:173. [PMID: 30872999 PMCID: PMC6400866 DOI: 10.3389/fnins.2019.00173] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/13/2019] [Indexed: 01/21/2023] Open
Abstract
The words “sex” and “gender” are often used interchangeably in common usage. In fact, the Merriam-Webster dictionary offers “sex” as the definition of gender. The authors of this review are neuroscientists, and the words “sex” and “gender” mean very different things to us: sex is based on biological factors such as sex chromosomes and gonads, whereas gender has a social component and involves differential expectations or treatment by conspecifics, based on an individual’s perceived sex. While we are accustomed to thinking about “sex” and differences between males and females in epigenetic marks in the brain, we are much less used to thinking about the biological implications of gender. Nonetheless, careful consideration of the field of epigenetics leads us to conclude that gender must also leave an epigenetic imprint on the brain. Indeed, it would be strange if this were not the case, because all environmental influences of any import can epigenetically change the brain. In the following pages, we explain why there is now sufficient evidence to suggest that an epigenetic imprint for gender is a logical conclusion. We define our terms for sex, gender, and epigenetics, and describe research demonstrating sex differences in epigenetic mechanisms in the brain which, to date, is mainly based on work in non-human animals. We then give several examples of how gender, rather than sex, may cause the brain epigenome to differ in males and females, and finally consider the myriad of ways that sex and gender interact to shape gene expression in the brain.
Collapse
Affiliation(s)
- Laura R Cortes
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Carla D Cisternas
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
209
|
Van Wert M, Anreiter I, Fallon BA, Sokolowski MB. Intergenerational Transmission of Child Abuse and Neglect: A Transdisciplinary Analysis. GENDER AND THE GENOME 2019. [DOI: 10.1177/2470289719826101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Melissa Van Wert
- Centre for Research on Children and Families, McGill University, Montreal, Quebec, Canada
- Melissa Van Wert and Ina Anreiter contributed equally to the preparation of this manuscript
| | - Ina Anreiter
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
- Melissa Van Wert and Ina Anreiter contributed equally to the preparation of this manuscript
| | - Barbara A. Fallon
- Factor Inwentash, Faculty of Social Work, University of Toronto, Toronto, Ontario, Canada
| | - Marla B. Sokolowski
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
- Child and Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada
| |
Collapse
|
210
|
Andersen SL. Stress, sensitive periods, and substance abuse. Neurobiol Stress 2019; 10:100140. [PMID: 30569003 PMCID: PMC6288983 DOI: 10.1016/j.ynstr.2018.100140] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/18/2018] [Accepted: 11/26/2018] [Indexed: 12/14/2022] Open
Abstract
Research on the inter-relationship between drug abuse and social stress has primarily focused on the role of stress exposure during adulthood and more recently, adolescence. Adolescence is a time of heightened reward sensitivity, but it is also a time when earlier life experiences are expressed. Exposure to stress early in postnatal life is associated with an accelerated age of onset for drug use. Lifelong addiction is significantly greater if drug use is initiated during early adolescence. Understanding how developmental changes following stress exposure interact with sensitive periods to unfold over the course of maturation is integral to reducing their later impact on substance use. Arousal levels, gender/sex, inflammation, and the timing of stress exposure play a role in the vulnerability of these circuits. The current review focuses on how early postnatal stress impacts brain development during a sensitive period to increase externalizing and internalizing behaviors in adolescence that include social interactions (aggression; sexual activity), working memory impairment, and depression. How stress effects the developmental trajectories of brain circuits that are associated with addiction are discussed for both clinical and preclinical studies.
Collapse
|
211
|
Toma S, Fiksenbaum L, Omrin D, Goldstein BI. Elevated Familial Cardiovascular Burden Among Adolescents With Familial Bipolar Disorder. Front Psychiatry 2019; 10:8. [PMID: 30761021 PMCID: PMC6361809 DOI: 10.3389/fpsyt.2019.00008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/08/2019] [Indexed: 02/01/2023] Open
Abstract
Background: Bipolar disorder (BD) is one of the most heritable medical conditions, and certain phenotypic characteristics are especially familial in BD. BD is also strongly associated with elevated and premature cardiovascular disease (CVD) morbidity and mortality. Thus, far, little is known regarding the familiality of cardiovascular risk in BD. We therefore examined the extent of CVD-related conditions among relatives of: adolescents with BD with a family history of BD (familial BD), adolescents with BD without a family history of BD (non-familial BD) and healthy controls (HC). Materials and Methods: The sample included 372 adolescents; 75 with familial BD, 96 with non-familial BD, and 201 HC. Parents of the adolescents completed the CARDIA Family Medical History interview regarding the adolescents' first- and second- degree adult relatives. We computed a "cardiovascular risk score" (CRS) for each relative, based on the sum of the presence of diabetes, hypertension, obesity, dyslipidemia, stroke, angina, and myocardial infarction (range 0-7). Primary analyses examined for group differences in mean overall CRS scores among first and second- degree relatives combined, controlling for age, sex, and race. Secondary analyses examined first- and second-degree relatives separately, controlling for age, sex, and race. Results: There were significant between-group differences in CRS in first- and second- degree relatives combined, following the hypothesized ordering: CRS was highest among adolescents with familial BD (1.14 ± 0.78), intermediate among adolescents with non-familial BD (0.92 ± 0.79) and lowest in HC (0.76 ± 0.79; F = 6.23, df = 2, p = 0.002, ηp 2 = 0.03). There was a significant pairwise difference between adolescents with familial BD and HC (p = 0.002, Cohen's d = 0.49). A similar pattern of between-group differences was identified when first-degree and second-degree relatives were examined separately. Limitations: familial cardiovascular burden was determined based on parent interview, not evaluated directly. Conclusions: Adolescents with BD with a family history of BD have elevated rates of CVD-related conditions among their relatives. This may be related to genetic overlap between BD and CVD-related conditions, shared environmental factors that contribute to both BD and CVD-related conditions, or a combination of these factors. More research is warranted to better understand the interaction between familial risk for BD and CVD, and to address this risk using family-wide preventive approaches.
Collapse
Affiliation(s)
- Simina Toma
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Lisa Fiksenbaum
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Danielle Omrin
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Benjamin I. Goldstein
- Centre for Youth Bipolar Disorder, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Pharmacology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
212
|
Phillips NLH, Roth TL. Animal Models and Their Contribution to Our Understanding of the Relationship Between Environments, Epigenetic Modifications, and Behavior. Genes (Basel) 2019; 10:genes10010047. [PMID: 30650619 PMCID: PMC6357183 DOI: 10.3390/genes10010047] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/29/2018] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
The use of non-human animals in research is a longstanding practice to help us understand and improve human biology and health. Animal models allow researchers, for example, to carefully manipulate environmental factors in order to understand how they contribute to development, behavior, and health. In the field of behavioral epigenetics such approaches have contributed novel findings of how the environment physically interacts with our genes, leading to changes in behavior and health. This review highlights some of this research, focused on prenatal immune challenges, environmental toxicants, diet, and early-life stress. In conjunction, we also discuss why animal models were integral to these discoveries and the translational relevance of these discoveries.
Collapse
Affiliation(s)
- Natalia Ledo Husby Phillips
- Department of Psychological and Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, USA.
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, 108 Wolf Hall, Newark, DE 19716, USA.
| |
Collapse
|
213
|
Jaffee SR. Lead exposure and child maltreatment as models for how to conceptualize early-in-life risk factors for violence. Infant Ment Health J 2019; 40:23-38. [DOI: 10.1002/imhj.21756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
214
|
Abstract
Our social environment, from the microscopic to the macro-social, affects us for the entirety of our lives. One integral line of research to examine how interpersonal and societal environments can get "under the skin" is through the lens of epigenetics. Epigenetic mechanisms are adaptations made to our genome in response to our environment which include tags placed on and removed from the DNA itself to how our DNA is packaged, affecting how our genes are read, transcribed, and interact. These tags are affected by social environments and can persist over time; this may aid us in responding to experiences and exposures, both the enriched and the disadvantageous. From memory formation to immune function, the experience-dependent plasticity of epigenetic modifications to micro- and macro-social environments may contribute to the process of learning from comfort, pain, and stress to better survive in whatever circumstances life has in store.
Collapse
Affiliation(s)
- Sarah M Merrill
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Gladish
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- Human Early Learning Partnership, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
215
|
Rustad SR, Papale LA, Alisch RS. DNA Methylation and Hydroxymethylation and Behavior. Curr Top Behav Neurosci 2019; 42:51-82. [PMID: 31392630 DOI: 10.1007/7854_2019_104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Environmentally sensitive molecular mechanisms in the brain, such as DNA methylation, have become a significant focus of neuroscience research because of mounting evidence indicating that they are critical in response to social situations, stress, threats, and behavior. The recent identification of 5-hydroxymethylcytosine (5hmC), which is enriched in the brain (tenfold over peripheral tissues), raises new questions as to the role of this base in mediating epigenetic effects in the brain. The development of genome-wide methods capable of distinguishing 5-methylcytosine (5mC) from 5hmC has revealed that a growing number of behaviors are linked to independent disruptions of 5mC and 5hmC levels, further emphasizing the unique importance of both of these modifications in the brain. Here, we review the recent links that indicate DNA methylation (both 5mC and 5hmC) is highly dynamic and that perturbations in this modification may contribute to behaviors related to psychiatric disorders and hold clinical relevance.
Collapse
Affiliation(s)
| | - Ligia A Papale
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA. .,Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
216
|
Abstract
Stress is an adaptive response to environment aversive stimuli and a common life experience of one's daily life. Chronic or excessive stress especially that happened in early life is found to be deleterious to individual's physical and mental health, which is highly related to depressive disorders onset. Stressful life events are consistently considered to be the high-risk factors of environment for predisposing depressive disorders. In linking stressful life events with depressive disorder onset, dysregulated HPA axis activity is supposed to play an important role in mediating aversive impacts of life stress on brain structure and function. Increasing evidence have indicated the strong association of stress, especially the chronic stress and early life stress, with depressive disorders development, while the association of stress with depression is moderated by genetic risk factors, including polymorphism of SERT, BDNF, GR, FKBP5, MR, and CRHR1. Meanwhile, stressful life experience particularly early life stress will exert epigenetic modification in these risk genes via DNA methylation and miRNA regulation to generate long-lasting effects on these genes expression, which in turn cause brain structural and functional alteration, and finally increase the vulnerability to depressive disorders. Therefore, the interaction of environment with gene, in which stressful life exposure interplay with genetic risk factors and epigenetic modification, is essential in predicting depressive disorders development. As the mediator of environmental risk factors, stress will function together with genetic and epigenetic mechanism to influence brain structure and function, physiology and psychology, and finally the vulnerability to depressive disorders.
Collapse
|
217
|
Agorastos A, Pervanidou P, Chrousos GP, Baker DG. Developmental Trajectories of Early Life Stress and Trauma: A Narrative Review on Neurobiological Aspects Beyond Stress System Dysregulation. Front Psychiatry 2019; 10:118. [PMID: 30914979 PMCID: PMC6421311 DOI: 10.3389/fpsyt.2019.00118] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 02/15/2019] [Indexed: 12/21/2022] Open
Abstract
Early life stressors display a high universal prevalence and constitute a major public health problem. Prolonged psychoneurobiological alterations as sequelae of early life stress (ELS) could represent a developmental risk factor and mediate risk for disease, leading to higher physical and mental morbidity rates in later life. ELS could exert a programming effect on sensitive neuronal brain networks related to the stress response during critical periods of development and thus lead to enduring hyper- or hypo-activation of the stress system and altered glucocorticoid signaling. In addition, alterations in emotional and autonomic reactivity, circadian rhythm disruption, functional and structural changes in the brain, as well as immune and metabolic dysregulation have been lately identified as important risk factors for a chronically impaired homeostatic balance after ELS. Furthermore, human genetic background and epigenetic modifications through stress-related gene expression could interact with these alterations and explain inter-individual variation in vulnerability or resilience to stress. This narrative review presents relevant evidence from mainly human research on the ten most acknowledged neurobiological allostatic pathways exerting enduring adverse effects of ELS even decades later (hypothalamic-pituitary-adrenal axis, autonomic nervous system, immune system and inflammation, oxidative stress, cardiovascular system, gut microbiome, sleep and circadian system, genetics, epigenetics, structural, and functional brain correlates). Although most findings back a causal relation between ELS and psychobiological maladjustment in later life, the precise developmental trajectories and their temporal coincidence has not been elucidated as yet. Future studies should prospectively investigate putative mediators and their temporal sequence, while considering the potentially delayed time-frame for their phenotypical expression. Better screening strategies for ELS are needed for a better individual prevention and treatment.
Collapse
Affiliation(s)
- Agorastos Agorastos
- II. Department of Psychiatry, Division of Neurosciences, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Panagiota Pervanidou
- Unit of Developmental and Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- Unit of Developmental and Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dewleen G Baker
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States.,VA Center of Excellence for Stress and Mental Health, San Diego, La Jolla, CA, United States
| |
Collapse
|
218
|
Changes in neuroplasticity following early-life social adversities: the possible role of brain-derived neurotrophic factor. Pediatr Res 2019; 85:225-233. [PMID: 30341412 DOI: 10.1038/s41390-018-0205-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 02/06/2023]
Abstract
Social adversities experienced in childhood can have a profound impact on the developing brain, leading to the emergence of psychopathologies in adulthood. Despite the burden this places on both the individual and society, the neurobiological aspects mediating this transition remain unclear. Recent advances in preclinical and clinical research have begun examining neuroplasticity-the nervous system's ability to form adaptive changes in response to new experience-in the context of early-life vulnerability to social adversities and plasticity-related alterations following such traumatic events. A key mediator of plasticity-related molecular processes is the brain-derived neurotrophic factor (BDNF), which has also been implicated in various psychiatric disorders related to childhood social adversities. Preclinical and clinical data suggest early-life social adversities (ELSA) might be associated with accelerated maturation of social network circuitry, a possible ontogenic adaptation to the adverse environment. Neural plasticity decreases by adulthood, lessening the efficacy of treatment in ELSA-related psychiatric disorders. However, literature data suggest that by increasing BDNF/TrkB signalling through antidepressant treatment a juvenile-like plasticity state can be induced, which allows for reorganization of the social circuitry when guided by psychotherapy and surrounded by a safe and positive environment.
Collapse
|
219
|
Chang L, Kigar SL, Ho JH, Cuarenta A, Gunderson HC, Baldo BA, Bakshi VP, Auger AP. Early life stress alters opioid receptor mRNA levels within the nucleus accumbens in a sex-dependent manner. Brain Res 2018; 1710:102-108. [PMID: 30594547 DOI: 10.1016/j.brainres.2018.12.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/10/2018] [Accepted: 12/26/2018] [Indexed: 01/24/2023]
Abstract
Early life stress (ELS) strongly impacts mental health, but little is known about its interaction with biological sex and postnatal development to influence risk and resilience to psychopathologies. A number of psychiatric disorders, such as social anhedonia and drug addiction, involve dysfunctional opioid signaling; moreover, there is evidence for differential central opioid function in males vs. females. The present study examined opioid receptor gene expression in the nucleus accumbens (NAc) and amygdala of male and female rats subjected to a neonatal predator odor exposure (POE) paradigm to model ELS. Brain tissue was collected at two developmental time points: neonatal and juvenile. Results showed that, following the neonatal POE experience, opioid receptor mRNA levels in the NAc were differentially regulated at the neonatal and juvenile time points. POE downregulated neonatal mu- and kappa-opioid receptor mRNA levels in neonatal females, but upregulated mu- and delta-opioid receptor mRNA levels in juvenile females. Intriguingly, POE had no significant effect on NAc opioid receptor mRNA levels in males at either time point, indicating that the impact of POE on opioid system development is sex-dependent. Finally, POE failed to alter amygdalar opioid receptor gene expression in either sex at either time-point. The spatiotemporally- and sex-specific impact of ELS within the developing brain may confer differential risk or resilience for males and females to develop atypical opioid-regulated behaviors associated with conditions such as depression and addiction.
Collapse
Affiliation(s)
- Liza Chang
- Department of Psychology, University of Wisconsin-Madison, United States.
| | - Stacey L Kigar
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, United States
| | - Jasmine H Ho
- Department of Psychology, University of Wisconsin-Madison, United States
| | - Amelia Cuarenta
- Department of Psychology, University of Wisconsin-Madison, United States
| | - Haley C Gunderson
- Department of Psychology, University of Wisconsin-Madison, United States
| | - Brian A Baldo
- Department of Psychiatry, University of Wisconsin-Madison, United States; Neuroscience Training Program, University of Wisconsin-Madison, United States
| | - Vaishali P Bakshi
- Department of Psychiatry, University of Wisconsin-Madison, United States; Neuroscience Training Program, University of Wisconsin-Madison, United States
| | - Anthony P Auger
- Department of Psychology, University of Wisconsin-Madison, United States; Neuroscience Training Program, University of Wisconsin-Madison, United States.
| |
Collapse
|
220
|
Lucero I. Written in the Body?: Healing the Epigenetic Molecular Wounds of Complex Trauma Through Empathy and Kindness. JOURNAL OF CHILD & ADOLESCENT TRAUMA 2018; 11:443-455. [PMID: 32318167 PMCID: PMC7163842 DOI: 10.1007/s40653-018-0205-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
While research on the biological/genetic approach to trauma transmission has become increasingly abundant over the past 15 years, this research has been primarily limited to scientific research journals and has not yet significantly appeared in practice journals, graduate programs, clinical settings, or policy decisions. This paper aims to develop a bridge across disciplines, integrating a review of biological science literature with mental health literature to provide a multidisciplinary overview of the role of epigenetic mechanisms in the transmission of complex trauma. Such a multidisciplinary overview is important in allowing professionals across disciplines to approach their work with a more complete understanding of the way in which ecological systems shape trauma transmission and healing. While encouraging collaboration between researchers and providers across fields, this paper argues that to heal the person, one must first work to heal the environment.
Collapse
Affiliation(s)
- Ivana Lucero
- School of Social Work, Simmons College, Boston, MA 02115 USA
| |
Collapse
|
221
|
Bearer EL, Mulligan BS. Epigenetic Changes Associated with Early Life Experiences: Saliva, A Biospecimen for DNA Methylation Signatures. Curr Genomics 2018; 19:676-698. [PMID: 30532647 PMCID: PMC6225450 DOI: 10.2174/1389202919666180307150508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/21/2017] [Accepted: 03/04/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Adverse Childhood Experiences (ACEs), which include traumatic injury, are associated with poor health outcomes in later life, yet the biological mechanisms mediating this association are unknown. Neurocircuitry, immune system and hormone regulation differ from normal in adults reporting ACEs. These systems could be affected by epigenetic changes, including methylation of cytosine (5mC) in genomic DNA, activated by ACEs. Since 5mC levels influence gene expression and can be long-lasting, altered 5mC status at specific sites or throughout the genome is hypothesized to influence mental and physical outcomes after ACE(s). Human and animal studies support this, with animal models allowing experiments for attributing causality. Here we provide a lengthy introduction and background on 5mC and the impact of early life adversity. OBJECTIVE Next we address the issue of a mixture of cell types in saliva, the most accessible biospecimen for 5mC analysis. Typical human bio-specimens for 5mC analysis include saliva or buccal swabs, whole blood or types of blood cells, tumors and post-mortem brain. In children saliva is the most accessible biospecimen, but contains a mixture of keratinocytes and white blood cells, as do buccal swabs. Even in saliva from the same individual at different time points, cell composition may differ widely. Similar issues affect analysis in blood, where nucleated cells represent a wide array of white blood cell types. Unless variations in ratios of these cells between each sample are included in the analysis, results can be unreliable. METHODS Several different biochemical assays are available to test for site-specific methylation levels genome-wide, each producing different information, with high-density arrays being the easiest to use, and bisulfite whole genome sequencing the most comprehensive. We compare results from different assays and use high-throughput computational processing to deconvolve cell composition in saliva samples. RESULTS Here we present examples demonstrating the critical importance of determining the relative contribution of blood cells versus keratinocytes to the 5mC profile found in saliva. We further describe a strategy to perform a reference-based computational correction for cell composition, and therefore to identify differential methylation patterns due to experience, or for the diagnosis of phenotypes that correlate between traits, such as hormone levels, trauma status and various mental health outcomes. CONCLUSION Specific sites that respond to adversity with altered methylation levels in either blood cells, keratinocytes or both can be identified by this rigorous approach, which will then be useful as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Elaine L. Bearer
- Address correspondence to this author at the Department of Pathology MSC 08-4640, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131, USA; Tel: 505-272-2404; Fax: 505-272-8084; E-mails: ;
| | | |
Collapse
|
222
|
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]
|
223
|
Lux V. Epigenetic Programming Effects of Early Life Stress: A Dual-Activation Hypothesis. Curr Genomics 2018; 19:638-652. [PMID: 30532644 PMCID: PMC6225448 DOI: 10.2174/1389202919666180307151358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/04/2017] [Accepted: 08/04/2017] [Indexed: 12/30/2022] Open
Abstract
Epigenetic processes during early brain development can function as 'developmental switches' that contribute to the stability of long-term effects of early environmental influences by programming central feedback mechanisms of the HPA axis and other neural networks. In this thematic review, we summarize accumulated evidence for a dual-activation of stress-related and sensory networks underlying the epigenetic programming effects of early life stress. We discuss findings indicating epigenetic programming of stress-related genes with impact on HPA axis function, the interaction of epigenetic mechanisms with neural activity in stress-related neural networks, epigenetic effects of glucocorticoid exposure, and the impact of stress on sensory development. Based on these findings, we propose that the combined activation of stress-related neural networks and stressor-specific sensory networks leads to both neural and hormonal priming of the epigenetic machinery, which sensitizes these networks for developmental programming effects. This allows stressor-specific adaptations later in life, but may also lead to functional mal-adaptations, depending on timing and intensity of the stressor. Finally, we discuss methodological and clinical implications of the dual-activation hypothesis. We emphasize that, in addition to modifications in stress-related networks, we need to account for functional modifications in sensory networks and their epigenetic underpinnings to elucidate the long-term effects of early life stress.
Collapse
Affiliation(s)
- Vanessa Lux
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
224
|
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
|
225
|
Abstract
Prenatal adversity shapes child neurodevelopment and risk for later mental health problems. The quality of the early care environment can buffer some of the negative effects of prenatal adversity on child development. Retrospective studies, in adult samples, highlight epigenetic modifications as sentinel markers of the quality of the early care environment; however, comparable data from pediatric cohorts are lacking. Participants were drawn from the Maternal Adversity Vulnerability and Neurodevelopment (MAVAN) study, a longitudinal cohort with measures of infant attachment, infant development, and child mental health. Children provided buccal epithelial samples (mean age = 6.99, SD = 1.33 years, n = 226), which were used for analyses of genome-wide DNA methylation and genetic variation. We used a series of linear models to describe the association between infant attachment and (a) measures of child outcome and (b) DNA methylation across the genome. Paired genetic data was used to determine the genetic contribution to DNA methylation at attachment-associated sites. Infant attachment style was associated with infant cognitive development (Mental Development Index) and behavior (Behavior Rating Scale) assessed with the Bayley Scales of Infant Development at 36 months. Infant attachment style moderated the effects of prenatal adversity on Behavior Rating Scale scores at 36 months. Infant attachment was also significantly associated with a principal component that accounted for 11.9% of the variation in genome-wide DNA methylation. These effects were most apparent when comparing children with a secure versus a disorganized attachment style and most pronounced in females. The availability of paired genetic data revealed that DNA methylation at approximately half of all infant attachment-associated sites was best explained by considering both infant attachment and child genetic variation. This study provides further evidence that infant attachment can buffer some of the negative effects of early adversity on measures of infant behavior. We also highlight the interplay between infant attachment and child genotype in shaping variation in DNA methylation. Such findings provide preliminary evidence for a molecular signature of infant attachment and may help inform attachment-focused early intervention programs.
Collapse
|
226
|
Brancato A, Cannizzaro C. Mothering under the influence: how perinatal drugs of abuse alter the mother-infant interaction. Rev Neurosci 2018; 29:283-294. [PMID: 29194045 DOI: 10.1515/revneuro-2017-0052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/16/2017] [Indexed: 12/26/2022]
Abstract
Although drug-abusing women try to moderate their drug and alcohol use during pregnancy, they often relapse at a time when childcare needs are high and maternal bonding is critical to an infant's development. In the clinical setting, the search for the neural basis of drug-induced caregiving deficits is complex due to several intervening variables. Rather, the preclinical studies that control for drug dose and regimen, as well as for gestational and postpartum environment, allow a precise determination of the effects of drugs on maternal behaviour. Given the relevance of the issue, this review will gather reports on the phenotypic correlates of maternal behaviour in preclinical studies, and focus on the detrimental consequences on the mother-infant interaction exerted by the perinatal use of alcohol, nicotine, cannabis, cocaine and stimulants and opiates. The drug-induced disruptions of this maternal repertoire are associated with adverse maternal and infant outcomes. A comprehensive overview will help promote the refinement of the treatment approaches toward maternal drug use disorders and maternal misbehaviour, in favour of augmented parenting resiliency.
Collapse
Affiliation(s)
- Anna Brancato
- Department of Sciences for Health Promotion and Mother and Child Care 'G. D'Alessandro', University of Palermo, via del Vespro 129, I-90127 Palermo, Italy
| | - Carla Cannizzaro
- Department of Sciences for Health Promotion and Mother and Child Care 'G. D'Alessandro', University of Palermo, via del Vespro 129, I-90127 Palermo, Italy
| |
Collapse
|
227
|
Wearick-Silva LE, Orso R, Martins LA, Creutzberg KC, Centeno-Silva A, Xavier LL, Grassi-Oliveira R, Mestriner RG. Dual influences of early life stress induced by limited bedding on walking adaptability and Bdnf/TrkB and Drd1/Drd2 gene expression in different mouse brain regions. Behav Brain Res 2018; 359:66-72. [PMID: 30347225 DOI: 10.1016/j.bbr.2018.10.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/10/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Introduction Evidence suggests early life stress impairs development, quality of life and increases vulnerability to disease. One important aspect of the stress experience is its impact on cognitive-motor performance, which includes the ability to adapt walking according to the environmental conditions. This study aimed to investigate how early-life stress affects walking adaptability of mice, while investigating BDNF/TrkB and Drd1/Drd2 expression in different brain regions. Methods Briefly, we exposed male C56BL/6 to the limited bedding protocol (LB) from post-natal day (PND) 2 to PND9 and then tested animals in the ladder walking task at PND60. RT-qPCR was used to investigate gene expression in the mPFC, hippocampus, motor cortex and cerebellum 2 h after the task Results LB induced a wide range of variability and therefore two distinct subgroups of animals within the LB group were established: a) superior performance (LB-SP); and b) inferior performance (LB-IP), compared to controls. Additionally, Drd1 gene expression was increased in the mPFC of LB-IP animals and in the cerebellum of LB-SP animals, while Drd2 expression was reduced in the hippocampus of the LB-IP group. BDNF exon IV gene expression in the mPFC and motor cortex was increased in both the LB-IP and LB-SP subgroups. TrkB gene expression in the hippocampus was reduced in the LB-IP group. A strong negative correlation was found between walking adaptability performance and BDNF exon IV gene expression in the motor cortex. Conversely, a positive correlation was found between walking adaptability performance and TrkB expression in the mPFC and a negative correlation in the hippocampus. Both Drd1 and Drd2 gene expression were negatively correlated with the ability to adapt walking. Conclusions Overall, our findings suggest exposure to early life stress leads to distinct walking adaptability phenotypes, which may be related to Drd1, Drd2, Bdnf exon IV and TrkB gene expression in brain regions that influence walking adaptability.
Collapse
Affiliation(s)
- L E Wearick-Silva
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - R Orso
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - L A Martins
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil
| | - K C Creutzberg
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - A Centeno-Silva
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - L L Xavier
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil
| | - R Grassi-Oliveira
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Porto Alegre, RS, Brazil; Developmental Cognitive Neuroscience Laboratory, Porto Alegre, RS, Brazil
| | - R G Mestriner
- Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Neurorehabilitation and Neural Repair Research Group, Porto Alegre, RS, Brazil.
| |
Collapse
|
228
|
Kang HJ, Bae KY, Kim SW, Shin IS, Hong YJ, Ahn Y, Jeong MH, Yoon JS, Kim JM. BDNF Methylation and Suicidal Ideation in Patients with Acute Coronary Syndrome. Psychiatry Investig 2018; 15:1094-1097. [PMID: 30380818 PMCID: PMC6259000 DOI: 10.30773/pi.2018.09.20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/20/2018] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Patients with acute coronary syndrome (ACS) are at an increased risk of suicide. It is well known that epigenetic mechanisms may explain the pathophysiology of suicidal behavior including suicidal ideation (SI), but no study has explored these mechanisms in ACS populations. METHODS In total, 969 patients were initially recruited within 2 weeks of the acute coronary event and, 711 patients were successfully followed up 1 year after ACS. SI was evaluated using the relevant items on the Montgomery-Åsberg Depression Rating Scale and covariates potentially affecting SI were estimated. RESULTS Brain-derived neurotrophic factor (BDNF) hypermethylation was associated with SI in both the acute and chronic phases of ACS, although the association was not statistically significant in the acute phase after applying Bonferroni's correction. CONCLUSION These results suggested that BDNF hypermethylation may have played a role in an epigenetic predisposition for SI in ACS patients, particularly during the chronic phase.
Collapse
Affiliation(s)
- Hee-Ju Kang
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kyung-Yeol Bae
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Il-Seon Shin
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young Joon Hong
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jin-Sang Yoon
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae-Min Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
229
|
Ihara K, Fuchikami M, Hashizume M, Okada S, Kawai H, Obuchi S, Hirano H, Fujiwara Y, Hachisu M, Hongyong K, Morinobu S. The influence of aging on the methylation status of brain-derived neurotrophic factor gene in blood. Int J Geriatr Psychiatry 2018; 33:1312-1318. [PMID: 29953671 DOI: 10.1002/gps.4927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/08/2018] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Brain-derived neurotrophic factor (BDNF) is involved in the pathophysiology of psychiatric disorders in adults and elderly individuals, and as a result, the DNA methylation (DNAm) of the BDNF gene in peripheral tissues including blood has been extensively examined to develop a useful biomarker for psychiatric disorders. However, studies to date have not previously investigated the effect of age on DNAm of the BDNF gene in blood. In this context, we measured DNAm of 39 CpG units in the CpG island at the promoter of exon I of the BDNF gene. METHODS We analyzed genomic DNA from peripheral blood of 105 health Japanese women 20 to 80 years of age to identify aging-associated change in DNAm of the BDNF gene. In addition, we examined the relationship between total MMSE scores, numbers of stressful life events, and serum BDNF levels on DNAm of the BDNF gene. The DNAm rate at each CpG unit was measured using a MassArray® system (Agena Bioscience), and serum BDNF levels were measured by ELISA. RESULTS There was a significant correlation between DNAm and age in 13 CpGs. However, there was no significant correlation between DNAm and total MMSE scores, numbers of life events, or serum BDNF levels. CONCLUSION Despite the small number of subjects and the inclusion of only female subjects, our results suggest that DNAm of 13 CpGs of the BDNF gene may be an appropriate biomarker for aging and useful for predicting increased susceptibility to age-related psychiatric disorders.
Collapse
Affiliation(s)
- Kazushige Ihara
- Hirosaki University Graduate School of Medicine, Department of Social Medicine, Aomori, Japan
| | | | - Masahiro Hashizume
- Toho University Faculty of Medicine, Department of Psychosomatic Medicine, Tokyo, Japan
| | - Satoshi Okada
- Hiroshima University, Department of Psychiatry and Neurosciences, Division of Frontier Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Hisashi Kawai
- Tokyo Metropolitan Institute of Gerontology, Human Care Research Team, Tokyo, Japan
| | - Shuichi Obuchi
- Tokyo Metropolitan Institute of Gerontology, Human Care Research Team, Tokyo, Japan
| | - Hirohiko Hirano
- Tokyo Metropolitan Geriatric Hospital, Department of Dentistry, Tokyo, Japan
| | - Yoshinori Fujiwara
- Tokyo Metropolitan Institute of Gerontology, Research Team for Social Participation and Community Health, Tokyo, Japan
| | - Mitsugu Hachisu
- Showa University, Department of Pharmaceutical therapeutics, Division of Clinical Pharmacy, Pharmacy School, Tokyo, Japan
| | - Kim Hongyong
- Tokyo Metropolitan Institute of Gerontology, Research Team for Promoting Independence of the Elderly, Tokyo, Japan
| | - Shigeru Morinobu
- Kochi University, Department of Neuropsychiatry, Kochi Medical School, Nankoku, Japan.,Kibi International University, Department of Occupational Therapy, School of Health Science and Social Welfare, Takahashi, Japan
| |
Collapse
|
230
|
Matt SM, Zimmerman JD, Lawson MA, Bustamante AC, Uddin M, Johnson RW. Inhibition of DNA Methylation With Zebularine Alters Lipopolysaccharide-Induced Sickness Behavior and Neuroinflammation in Mice. Front Neurosci 2018; 12:636. [PMID: 30279646 PMCID: PMC6153314 DOI: 10.3389/fnins.2018.00636] [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: 01/19/2018] [Accepted: 08/24/2018] [Indexed: 12/24/2022] Open
Abstract
Activity of DNA methyltransferases (DNMTs), the enzymes that catalyze DNA methylation, is dynamically regulated in the brain. DNMT inhibitors alter DNA methylation globally in the brain and at individual neural plasticity-associated genes, but how DNMT inhibitors centrally influence lipopolysaccharide (LPS)-induced neuroinflammation is not known. We investigated whether the DMNT inhibitor, zebularine, would alter sickness behavior, DNA methylation of the Il-1β promoter and expression of inflammatory genes in hippocampus and microglia. Contrary to our hypothesis that zebularine may exaggerate LPS-induced sickness response and neuroinflammation, adult mice treated with an intracerebroventricular (ICV) injection of zebularine prior to LPS had surprisingly faster recovery of burrowing behavior compared to mice treated with LPS. Further, genes of inflammatory markers, epigenetic regulators, and the microglial sensory apparatus (i.e., the sensome) were differentially expressed by zebularine alone or in combination with LPS. Bisulfite pyrosequencing revealed that ICV zebularine led to decreased DNA methylation of two CpG sites near the Il-1β proximal promoter alone or in combination with LPS. Zebularine treated mice still exhibited decreased DNA methylation 48 h after treatment when LPS-induced sickness behavior as well as hippocampal and microglial gene expression were similar to control mice. Taken together, these data suggest that decreased DNA methylation, specifically of the Il-1β promoter region, with a DNMT inhibitor in the brain disrupts molecular mechanisms of neuroinflammation.
Collapse
Affiliation(s)
- Stephanie M Matt
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Animal Sciences Laboratory, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Jalisa D Zimmerman
- Animal Sciences Laboratory, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Marcus A Lawson
- Animal Sciences Laboratory, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Angela C Bustamante
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Monica Uddin
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Rodney W Johnson
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Animal Sciences Laboratory, Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.,Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
231
|
Toth M. The other side of the coin: Hypersociability. GENES BRAIN AND BEHAVIOR 2018; 18:e12512. [PMID: 30101538 DOI: 10.1111/gbb.12512] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/18/2018] [Accepted: 08/07/2018] [Indexed: 12/29/2022]
Abstract
Affiliative social motivation and behavior, that is, sociability that includes attachment, prosocial behavior (sharing, caring and helping) and empathy (the ability to understand and share the feelings of others), has high variability in the human population, with a portion of people outside of the normal range. While psychiatric disorders and autism spectrum disorders are typically associated with a deficit in social behavior, the opposite trait of hypersociability and indiscriminate friendliness are exhibited by individual with specific neurodevelopmental disorders and following early adverse care. Here we discuss both genetic and environmental factors that cause or increase the risk for developing pathological hypersociability from human to rodent models.
Collapse
Affiliation(s)
- Miklos Toth
- Department of Pharmacology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
232
|
Yogman M, Garner A, Hutchinson J, Hirsh-Pasek K, Golinkoff RM, Baum R, Gambon T, Lavin A, Mattson G, Wissow L, Hill DL, Ameenuddin N, Chassiakos Y(LR, Cross C, Boyd R, Mendelson R, Moreno MA, Radesky J, Swanson WS, Hutchinson J, Smith J. The Power of Play: A Pediatric Role in Enhancing Development in Young Children. Pediatrics 2018; 142:peds.2018-2058. [PMID: 30126932 DOI: 10.1542/peds.2018-2058] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Children need to develop a variety of skill sets to optimize their development and manage toxic stress. Research demonstrates that developmentally appropriate play with parents and peers is a singular opportunity to promote the social-emotional, cognitive, language, and self-regulation skills that build executive function and a prosocial brain. Furthermore, play supports the formation of the safe, stable, and nurturing relationships with all caregivers that children need to thrive.Play is not frivolous: it enhances brain structure and function and promotes executive function (ie, the process of learning, rather than the content), which allow us to pursue goals and ignore distractions.When play and safe, stable, nurturing relationships are missing in a child's life, toxic stress can disrupt the development of executive function and the learning of prosocial behavior; in the presence of childhood adversity, play becomes even more important. The mutual joy and shared communication and attunement (harmonious serve and return interactions) that parents and children can experience during play regulate the body's stress response. This clinical report provides pediatric providers with the information they need to promote the benefits of play and and to write a prescription for play at well visits to complement reach out and read. At a time when early childhood programs are pressured to add more didactic components and less playful learning, pediatricians can play an important role in emphasizing the role of a balanced curriculum that includes the importance of playful learning for the promotion of healthy child development.
Collapse
Affiliation(s)
- Michael Yogman
- Department of Pediatrics, Harvard Medical School, Harvard University and Mount Auburn Hospital, Cambridge, Massachusetts
| | - Andrew Garner
- Department of Pediatrics, School of Medicine, Case Western Reserve University and University Hospitals Medical Practices, Cleveland, Ohio
| | - Jeffrey Hutchinson
- Department of Pediatrics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland
| | - Kathy Hirsh-Pasek
- Department of Psychology, Brookings Institution and Temple University, Philadelphia, Pennsylvania; and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Wang RH, Chen YF, Chen S, Hao B, Xue L, Wang XG, Shi YW, Zhao H. Maternal Deprivation Enhances Contextual Fear Memory via Epigenetically Programming Second-Hit Stress-Induced Reelin Expression in Adult Rats. Int J Neuropsychopharmacol 2018; 21:1037-1048. [PMID: 30169690 PMCID: PMC6209857 DOI: 10.1093/ijnp/pyy078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 08/29/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Early-life stress increases the risk for posttraumatic stress disorder. However, the epigenetic mechanism of early-life stress-induced susceptibility to posttraumatic stress disorder in adulthood remains unclear. METHODS Rat pups were exposed to maternal deprivation during postnatal days 1 to 14 for 3 hours daily and treated with the DNA methyltransferase inhibitor zebularine, L-methionine, or vehicle 7 days before contextual fear conditioning, which was used as a second stress and to mimic the reexperiencing symptom of posttraumatic stress disorder in adulthood. Long-term potentiation, dendritic spine density, DNA methyltransferase mRNA, Reelin gene methylation, and Reelin protein expression in the hippocampal CA1 were measured. RESULTS Maternal deprivation enhanced contextual fear memory in adulthood. Meanwhile, maternal deprivation decreased DNA methyltransferase mRNA and Reelin gene methylation in the hippocampal CA1 on postnatal days 22 and 90. Reelin protein expression was increased in the hippocampal CA1 following contextual fear conditioning in adulthood. Furthermore, compared with rats that experienced maternal deprivation alone, rats also exposed to contextual fear conditioning showed an enhanced induction of hippocampal long-term potentiation and increased dendritic spine density in the hippocampal CA1 following contextual fear conditioning in adulthood. Zebularine pretreatment led to an enhancement of contextual fear memory, hypomethylation of the Reelin gene, and increased Reelin protein expression in adult rats, while L-methionine had the opposite effects. CONCLUSIONS Maternal deprivation can epigenetically program second-hit stress-induced Reelin expression and enhance the susceptibility to contextual fear memory in adulthood. These findings provide a new framework for understanding the cumulative stress hypothesis.
Collapse
Affiliation(s)
- Run-Hua Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
| | - Ye-Fei Chen
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
| | - Si Chen
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China,Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bo Hao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
| | - Li Xue
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Guang Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan-Wei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Correspondence: Hu Zhao, PhD, MD, Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China (); and Yan-Wei Shi, PhD, Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China ()
| | - Hu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China,Correspondence: Hu Zhao, PhD, MD, Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China (); and Yan-Wei Shi, PhD, Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China ()
| |
Collapse
|
234
|
Thomas M, Knoblich N, Wallisch A, Glowacz K, Becker-Sadzio J, Gundel F, Brückmann C, Nieratschker V. Increased BDNF methylation in saliva, but not blood, of patients with borderline personality disorder. Clin Epigenetics 2018; 10:109. [PMID: 30134995 PMCID: PMC6106893 DOI: 10.1186/s13148-018-0544-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Background The importance of epigenetic alterations in psychiatric disorders is increasingly acknowledged and the use of DNA methylation patterns as markers of disease is a topic of ongoing investigation. Recent studies suggest that patients suffering from Borderline Personality Disorder (BPD) display differential DNA methylation of various genes relevant for neuropsychiatric conditions. For example, several studies report differential methylation in the promoter region of the brain-derived neurotrophic factor gene (BDNF) in blood. However, little is known about BDNF methylation in other tissues. Results In the present study, we analyzed DNA methylation of the BDNF IV promoter in saliva and blood of 41 BPD patients and 41 matched healthy controls and found significant hypermethylation in the BPD patient’s saliva, but not blood. Further, we report that BDNF methylation in saliva of BPD patients significantly decreased after a 12-week psychotherapeutic intervention. Conclusions Providing a direct comparison of BDNF methylation in blood and saliva of the same individuals, our results demonstrate the importance of choice of tissue for the study of DNA methylation. In addition, they indicate a better suitability of saliva for the study of differential BDNF methylation in BPD patients. Further, our data appear to indicate a reversal of disease-specific alterations in BDNF methylation in response to psychotherapy, though further experiments are necessary to validate these results and determine the specificity of the effect. Electronic supplementary material The online version of this article (10.1186/s13148-018-0544-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mara Thomas
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany.,Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Nora Knoblich
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Annalena Wallisch
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Katarzyna Glowacz
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Julia Becker-Sadzio
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Friederike Gundel
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Christof Brückmann
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany
| | - Vanessa Nieratschker
- Department of Psychiatry and Psychotherapy, University Hospital Tübingen, Calwerstr. 14, 72076, Tübingen, Germany.
| |
Collapse
|
235
|
Shepard RD, Gouty S, Kassis H, Berenji A, Zhu W, Cox BM, Nugent FS. Targeting histone deacetylation for recovery of maternal deprivation-induced changes in BDNF and AKAP150 expression in the VTA. Exp Neurol 2018; 309:160-168. [PMID: 30102916 DOI: 10.1016/j.expneurol.2018.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/17/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Severe early life stressors increase the probability of developing psychiatric disorders later in life through modifications in neuronal circuits controlling brain monoaminergic signaling. Our previous work demonstrated that 24 h maternal deprivation (MD) in male Sprague Dawley rats modifies dopamine (DA) signaling from the ventral tegmental area (VTA) through changes at GABAergic synapses that were reversible by in vitro histone deacetylase (HDAC) inhibition which led to restoration of the scaffold A-kinase anchoring protein (AKAP150) signaling and subsequently recovered GABAergic plasticity (Authement et al., 2015). Using a combination of in situ hybridization, Western blots and immunohistochemistry, we confirmed that MD-induced epigenetic modifications at the level of histone acetylation were associated with an upregulation of HDAC2. MD also increased Akap5 mRNA levels in the VTA. Western blot analysis of AKAP150 protein expression showed an increase in synaptic levels of AKAP150 protein in the VTA with an accompanying decrease in synaptic levels of protein kinase A (PKA). Moreover, the abundance of mature brain-derived neurotrophic factor (BDNF) protein of VTA tissues from MD rats was significantly lower than in control groups. In vivo systemic injection with a selective class I HDAC inhibitor (CI-994) was sufficient to reverse MD-induced histone hypoacetylation in the VTA for 24 h after the injection. Furthermore, HDAC inhibition normalized the levels of mBDNF and AKAP150 proteins at 24 h. Our data suggest that HDAC-mediated targeting of BDNF and AKAP-dependent local signaling within VTA could provide novel therapeutics for prevention of later-life psychopathology.
Collapse
Affiliation(s)
- Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Haifa Kassis
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Aylar Berenji
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - William Zhu
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, MD 20814, USA.
| |
Collapse
|
236
|
Abstract
PURPOSE OF REVIEW We review the existing literature on gene-environment interactions (G×E) and epigenetic changes primarily in borderline personality disorder (BPD) but also in antisocial, schizotypal, and avoidant personality disorders. RECENT FINDINGS Research supports that susceptibility genes to BPD or its underlying traits may be expressed under certain environmental conditions such as physical or childhood sexual abuse. Epigenetic modifications of neurodevelopment- and stress-related genes are suggested to underlie the relationship between early life adversary and borderline personality disorder. Only limited studies have investigated the role of gene-environment interactions and epigenetic changes in the genesis of antisocial, schizotypal, and avoidant personality disorders. Considering the lack of pharmacological treatment for most personality disorders, the emerging evidence on the critical role of G×E and epigenetic changes in the genesis of personality disorders could help develop more biologically oriented therapeutic approaches. Future studies should explore the potential of this new therapeutic dimension.
Collapse
|
237
|
Karatekin C, Almy B, Mason SM, Borowsky I, Barnes A. Documentation of Child Maltreatment in Electronic Health Records. Clin Pediatr (Phila) 2018; 57:1041-1052. [PMID: 29168394 DOI: 10.1177/0009922817743571] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
International Classification of Diseases codes for child maltreatment can aid surveillance and research, but the extent to which they are used is not well established. We documented prevalence of the use of maltreatment-related codes, examined demographic characteristics of youth assigned these codes, and compared results with previous studies. Data were extracted from electronic health records of 0- to 21-year-olds assigned 1 of 15 maltreatment-related International Classification of Diseases, Ninth Revision, codes who had encounters in a large medical system over a 4-year period. Only 0.02% of approximately 2.5 million youth had a maltreatment-related code, replicating other studies. Results provide a dramatic contrast to much higher rates based on self-report or informant-report and referrals to Child Protective Services. Lack of documentation of maltreatment in electronic health records can lead to missed chances at early intervention, inadequate coordination of health care, insufficient allocation of resources to addressing problems related to maltreatment, and flawed public health data.
Collapse
|
238
|
Abstract
Attachment theory was developed by John Bowlby in the 1950s. He defined attachment as a specific neurobiological system that resulted in the infant connecting to the primary caretaker in such a way to create an inner working model of relationships that continues throughout life and affects the future mental health and physical health of the infant. Given the significance of this inner working model, there has been a tremendous amount of research done in animals as well as humans to better understand the neurobiology. In this article the neurobiology of early development will be outlined with respect to the formation of attachment. This article will review what we have begun to understand as the neurobiology of attachment and will describe how the relationship with the primary caretaker affects the infant in a way leading to neurobiological changes that later in life affect emotional responses, reward, and perception difficulties that we recognize as psychiatric illness and medical morbidity.
Collapse
Affiliation(s)
- Joanna Chambers
- Associate Professor of Clinical Psychiatry, Indiana University School of Medicine; Chair of Scientific Programs, American Academy of Psychoanalysis and Dynamic Psychiatry
| |
Collapse
|
239
|
Jiménez JP, Botto A, Herrera L, Leighton C, Rossi JL, Quevedo Y, Silva JR, Martínez F, Assar R, Salazar LA, Ortiz M, Ríos U, Barros P, Jaramillo K, Luyten P. Psychotherapy and Genetic Neuroscience: An Emerging Dialog. Front Genet 2018; 9:257. [PMID: 30065751 PMCID: PMC6056612 DOI: 10.3389/fgene.2018.00257] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022] Open
Abstract
Recent research in psychiatric genetics has led to a move away from simple diathesis-stress models to more complex models of psychopathology incorporating a focus on gene–environment interactions and epigenetics. Our increased understanding of the way biology encodes the impact of life events on organisms has also generated more sophisticated theoretical models concerning the molecular processes at the interface between “nature” and “nurture.” There is also increasing consensus that psychotherapy entails a specific type of learning in the context of an emotional relationship (i.e., the therapeutic relationship) that may also lead to epigenetic modifications across different therapeutic treatment modalities. This paper provides a systematic review of this emerging body of research. It is concluded that, although the evidence is still limited at this stage, extant research does indeed suggest that psychotherapy may be associated with epigenetic changes. Furthermore, it is argued that epigenetic studies may play a key role in the identification of biomarkers implicated in vulnerability for psychopathology, and thus may improve diagnosis and open up future research opportunities regarding the mechanism of action of psychotropic drugs as well as psychotherapy. We review evidence suggesting there may be important individual differences in susceptibility to environmental input, including psychotherapy. In addition, given that there is increasing evidence for the transgenerational transmission of epigenetic modifications in animals and humans exposed to trauma and adversity, epigenetic changes produced by psychotherapy may also potentially be passed on to the next generation, which opens up new perspective for prevention science. We conclude this paper stressing the limitations of current research and by proposing a set of recommendations for future research in this area.
Collapse
Affiliation(s)
- Juan P Jiménez
- Department of Psychiatry and Mental Health - East, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alberto Botto
- Department of Psychiatry and Mental Health - East, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Luisa Herrera
- Human Genetics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Caroline Leighton
- Department of Psychiatry and Mental Health - East, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José L Rossi
- Department of Psychology, Faculty of Social Sciences, Universidad de Chile, Santiago, Chile
| | - Yamil Quevedo
- Department of Psychiatry and Mental Health - East, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jaime R Silva
- Center for Attachment and Emotional Regulation (CARE), Faculty of Psychology, Universidad del Desarrollo, Santiago, Chile
| | - Felipe Martínez
- Center for Intercultural and Indigenous Research, Anthropology Program, Institute of Sociology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Assar
- ICBM Human Genetics Program, Centre for Medical Informatics and Telemedicine, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Manuel Ortiz
- Department of Psychology, Faculty of Education, Social Sciences and Humanities, Universidad de La Frontera, Temuco, Chile
| | - Ulises Ríos
- Department of Psychiatry, Universidad de Valparaíso, Valparaíso, Chile
| | - Paulina Barros
- Department of Psychiatry and Mental Health - East, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Karina Jaramillo
- Ph.D. Program in Public Health, School of Public Health, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Patrick Luyten
- Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium.,Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom
| |
Collapse
|
240
|
Frías-Lasserre D, Villagra CA, Guerrero-Bosagna C. Stress in the Educational System as a Potential Source of Epigenetic Influences on Children's Development and Behavior. Front Behav Neurosci 2018; 12:143. [PMID: 30057532 PMCID: PMC6053942 DOI: 10.3389/fnbeh.2018.00143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/25/2018] [Indexed: 11/24/2022] Open
Abstract
Despite current advances on the relevance of environmental cues and epigenetic mechanisms in biological processes, including behavior, little attention has been paid to the potential link between epigenetic influences and educational sciences. For instance, could the learning environment and stress determine epigenetic marking, affecting students' behavior development? Could this have consequences on educational outcomes? So far, it has been shown that environmental stress influences neurological processes and behavior both in humans and rats. Through epigenetic mechanisms, offspring from stressed individuals develop altered behavior without any exposure to traumatizing experiences. Methylated DNA and noncoding RNAs regulate neurological processes such as synaptic plasticity and brain cortex development in children. The malfunctioning of these processes is associated with several neurological disorders, and these findings open up new avenues for the design of enriched environments for education and therapy. In this article, we discuss current cases of stress and behavioral disorders found in youngsters, and highlight the importance of considering epigenetic processes affecting the development of cognitive abilities and learning within the educational environment and for the development of teaching methodologies.
Collapse
Affiliation(s)
- Daniel Frías-Lasserre
- Instituto de Entomología, Universidad Metropolitana de Ciencias de la Educación, Santiago, Chile
| | - Cristian A. Villagra
- Instituto de Entomología, Universidad Metropolitana de Ciencias de la Educación, Santiago, Chile
| | | |
Collapse
|
241
|
A paternal methyl donor depleted diet leads to increased anxiety- and depression-like behavior in adult rat offspring. Biosci Rep 2018; 38:BSR20180730. [PMID: 29945927 PMCID: PMC6153370 DOI: 10.1042/bsr20180730] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
Epigenetic mechanisms such as DNA methylation elicit lasting changes in gene expression and likely mediate gene-environment interactions that shape brain development, behavior, and emotional health. Myriad environmental factors influence DNA methylation, including methyl donor content in the paternal diet, could influence methylation in offspring via changes in the paternal germ line. The present study examines the effects of paternal methyl donor dietary deficiency on offspring's emotional behaviors, including anxiety, social interaction, and depression-like behavior. We previously found that rats bred to display high levels of anxiety- and depression-like behavior exhibit diminished DNA methylation in the amygdala. We also observed that depleting dietary methyl donor content exacerbated the rats' already high levels of anxiety- and depression-like behavior. Here we sought to determine whether paternal dietary methyl donor depletion elicits intergenerational effects on first generation (F1) offspring's behavior (potentially triggering a similar increase in anxiety- and/or depression-like behavior). Thus, adult male rats prone to high anxiety/depression-like behavior, were fed either a methyl donor depleted (DEP) or control (CON) diet for 5 weeks prior to mating. They were paired with females and resultant F1 male offspring were subjected to a behavioral test battery in adulthood. F1-DEP offspring showed a similar behavioral profile to the F0 males, including greater depression-like behavior in the forced swim test (FST) and increased anxiety-like behavior in the open field test (OFT). Future work will interrogate molecular changes in the brains of F1 offspring that mediate these intergenerational effects of paternal methyl donor dietary content on offspring emotional behavior.
Collapse
|
242
|
Fuentes S, Daviu N, Gagliano H, Belda X, Armario A, Nadal R. Early life stress in rats sex-dependently affects remote endocrine rather than behavioral consequences of adult exposure to contextual fear conditioning. Horm Behav 2018; 103:7-18. [PMID: 29802874 DOI: 10.1016/j.yhbeh.2018.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/09/2018] [Accepted: 05/22/2018] [Indexed: 01/07/2023]
Abstract
Exposure to electric foot-shocks can induce in rodents contextual fear conditioning, generalization of fear to other contexts and sensitization of the hypothalamic-pituitary-adrenal (HPA) axis to further stressors. All these aspects are relevant for the study of post-traumatic stress disorder. In the present work we evaluated in rats the sex differences and the role of early life stress (ELS) in fear memories, generalization and sensitization. During the first postnatal days subjects were exposed to restriction of nesting material along with exposure to a "substitute" mother. In the adulthood they were exposed to (i) a contextual fear conditioning to evaluate long-term memory and extinction and (ii) to a novel environment to study cognitive fear generalization and HPA axis heterotypic sensitization. ELS did not alter acquisition, expression or extinction of context fear conditioned behavior (freezing) in either sex, but reduced activity in novel environments only in males. Fear conditioning associated hypoactivity in novel environments (cognitive generalization) was greater in males than females but was not specifically affected by ELS. Although overall females showed greater basal and stress-induced levels of ACTH and corticosterone, an interaction between ELS, shock exposure and sex was found regarding HPA hormones. In males, ELS did not affect ACTH response in any situation, whereas in females, ELS reduced both shock-induced sensitization of ACTH and its conditioned response to the shock context. Also, shock-induced sensitization of corticosterone was only observed in males and ELS specifically reduced corticosterone response to stressors in males but not females. In conclusion, ELS seems to have only a minor impact on shock-induced behavioral conditioning, while affecting the unconditioned and conditioned responses of HPA hormones in a sex-dependent manner.
Collapse
Affiliation(s)
- Sílvia Fuentes
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Spain
| | - Núria Daviu
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Humberto Gagliano
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Xavier Belda
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Antonio Armario
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain.
| | - Roser Nadal
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
243
|
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
|
244
|
Patton GC, Olsson CA, Skirbekk V, Saffery R, Wlodek ME, Azzopardi PS, Stonawski M, Rasmussen B, Spry E, Francis K, Bhutta ZA, Kassebaum NJ, Mokdad AH, Murray CJL, Prentice AM, Reavley N, Sheehan P, Sweeny K, Viner RM, Sawyer SM. Adolescence and the next generation. Nature 2018; 554:458-466. [PMID: 29469095 DOI: 10.1038/nature25759] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/18/2018] [Indexed: 12/30/2022]
Abstract
Adolescent growth and social development shape the early development of offspring from preconception through to the post-partum period through distinct processes in males and females. At a time of great change in the forces shaping adolescence, including the timing of parenthood, investments in today's adolescents, the largest cohort in human history, will yield great dividends for future generations.
Collapse
Affiliation(s)
- George C Patton
- The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Craig A Olsson
- The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria 3052, Australia.,Deakin University Geelong, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria 3220, Australia
| | - Vegard Skirbekk
- Centre for Fertility and Health, Norwegian Institute of Public Health, Nydalen, Oslo 0403, Norway.,Columbia University, New York, New York 10032, USA
| | - Richard Saffery
- The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
| | - Mary E Wlodek
- The University of Melbourne, Department of Physiology, Parkville, Victoria 3010, Australia
| | - Peter S Azzopardi
- The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Maternal and Child Health Program, International Development Discipline, Burnet Institute, Melbourne, Victoria 3004, Australia.,Wardliparingga Aboriginal Research Unit, South Australian Health and Medical Research Institute, Adelaide, South Australia 5000, Australia
| | - Marcin Stonawski
- Department of Demography, Cracow University of Economics, Cracow 31-510, Poland.,European Commission, Joint Research Centre, Centre for Advanced Studies, Ispra, Varese 21027, Italy
| | - Bruce Rasmussen
- Victoria Institute of Strategic Economic Studies, Victoria University, Melbourne, Victoria 3000, Australia
| | - Elizabeth Spry
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria 3052, Australia.,Deakin University Geelong, Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Geelong, Victoria 3220, Australia
| | - Kate Francis
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Zulfiqar A Bhutta
- SickKids Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Centre of Excellence in Women and Child Health, Aga Khan University, Karachi 74800, Pakistan
| | - Nicholas J Kassebaum
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington 98121, USA.,Division of Pediatric Anesthesiology & Pain Medicine, Seattle Children's Hospital, Seattle, Washington 98105, USA
| | - Ali H Mokdad
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington 98121, USA
| | - Christopher J L Murray
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington 98121, USA
| | - Andrew M Prentice
- MRC Unit The Gambia, Fajara, Gambia.,MRC International Nutrition Group, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Nicola Reavley
- The University of Melbourne, Melbourne School of Population and Global Health, Parkville, Victoria 3010, Australia
| | - Peter Sheehan
- Victoria Institute of Strategic Economic Studies, Victoria University, Melbourne, Victoria 3000, Australia
| | - Kim Sweeny
- Victoria Institute of Strategic Economic Studies, Victoria University, Melbourne, Victoria 3000, Australia
| | - Russell M Viner
- UCL Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Susan M Sawyer
- The University of Melbourne, Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, Parkville, Victoria 3010, Australia.,Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| |
Collapse
|
245
|
Post RM, Altshuler LL, Kupka R, McElroy SL, Frye MA, Rowe M, Grunze H, Suppes T, Keck PE, Leverich GS, Nolen WA. Multigenerational transmission of liability to psychiatric illness in offspring of parents with bipolar disorder. Bipolar Disord 2018; 20:432-440. [PMID: 29926532 DOI: 10.1111/bdi.12668] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Instead of the typical assessment of risk of illness in the offspring based on a parent with bipolar disorder, we explored the potential multigenerational conveyance across several disorders of the vulnerability to illness in the offspring of a patient with bipolar disorder. METHODS A total of 968 outpatients (average age 41 years) with bipolar illness gave informed consent and filled out a detailed questionnaire about a family history in their parents, grandparents, and offspring of: depression; bipolar disorder; alcohol abuse; substance abuse; suicide attempt; or "other" illness. Of those with children, 346 were from the USA and 132 were from Europe. Amount and type of illness in progenitors in two and three previous generations were related to offspring illness. RESULTS The type of illness seen in both prior generations was associated with the same type of illness in the offspring of a bipolar patient, including depression, bipolar disorder, alcohol and substance abuse and "other" illness, but not suicide attempts. There was an impact of multiple generations, such that depression in grandparents and/or great-grandparents increased the risk of depression in the offspring from 12.6% to 41.4%. CONCLUSIONS A family history of illness burden in prior generations was previously related to an earlier age of onset of bipolar illness in our adult patients with bipolar disorder and is now also found to be related to the incidence of multiple psychiatric illnesses in their offspring. Genetic and epigenetic mechanisms deserve consideration for this multigenerational conveyance of illness vulnerability, and clinical and public health attempts to prevent or slow this transmission are indicated.
Collapse
Affiliation(s)
- Robert M Post
- Bipolar Collaborative Network, Bethesda, Maryland
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, District of Columbia
| | - Lori L Altshuler
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California
- Department of Psychiatry, VA Greater Los Angeles Healthcare System, West Los Angeles Healthcare Center, Los Angeles, California
| | - Ralph Kupka
- Department of Psychiatry and Amsterdam Public Health Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Susan L McElroy
- Lindner Center of HOPE, Mason, Ohio
- Biological Psychiatry Program, University of Cincinnati Medical College, Cincinnati, Ohio
| | - Mark A Frye
- Department of Psychiatry, Mayo Clinic, Rochester, Minnesota
| | - Michael Rowe
- Bipolar Collaborative Network, Bethesda, Maryland
| | - Heinz Grunze
- Klinikum am Weissenhof, Weinsberg Germany & Paracelsus Medical University, Nuremberg, Germany
| | - Trisha Suppes
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California
- V.A. Palo Alto Health Care System, Palo Alto, California
| | - Paul E Keck
- Lindner Center of HOPE, Mason, Ohio
- Department of Psychiatry & Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Willem A Nolen
- University Medical Center, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
246
|
Youssef MM, Underwood MD, Huang YY, Hsiung SC, Liu Y, Simpson NR, Bakalian MJ, Rosoklija GB, Dwork AJ, Arango V, Mann JJ. Association of BDNF Val66Met Polymorphism and Brain BDNF Levels with Major Depression and Suicide. Int J Neuropsychopharmacol 2018; 21:528-538. [PMID: 29432620 PMCID: PMC6007393 DOI: 10.1093/ijnp/pyy008] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/11/2018] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Background Brain-derived neurotrophic factor is implicated in the pathophysiology of major depressive disorder and suicide. Both are partly caused by early life adversity, which reduces brain-derived neurotrophic factor protein levels. This study examines the association of brain-derived neurotrophic factor Val66Met polymorphism and brain brain-derived neurotrophic factor levels with depression and suicide. We hypothesized that both major depressive disorder and early life adversity would be associated with the Met allele and lower brain brain-derived neurotrophic factor levels. Such an association would be consistent with low brain-derived neurotrophic factor mediating the effect of early life adversity on adulthood suicide and major depressive disorder. Methods Brain-derived neurotrophic factor Val66Met polymorphism was genotyped in postmortem brains of 37 suicide decedents and 53 nonsuicides. Additionally, brain-derived neurotrophic factor protein levels were determined by Western blot in dorsolateral prefrontal cortex (Brodmann area 9), anterior cingulate cortex (Brodmann area 24), caudal brainstem, and rostral brainstem. The relationships between these measures and major depressive disorder, death by suicide, and reported early life adversity were examined. Results Subjects with the Met allele had an increased risk for depression. Depressed patients also have lower brain-derived neurotrophic factor levels in anterior cingulate cortex and caudal brainstem compared with nondepressed subjects. No effect of history of suicide death or early life adversity was observed with genotype, but lower brain-derived neurotrophic factor levels in the anterior cingulate cortex were found in subjects who had been exposed to early life adversity and/or died by suicide compared with nonsuicide decedents and no reported early life adversity. Conclusions This study provides further evidence implicating low brain brain-derived neurotrophic factor and the brain-derived neurotrophic factor Met allele in major depression risk. Future studies should seek to determine how altered brain-derived neurotrophic factor expression contributes to depression and suicide.
Collapse
Affiliation(s)
- Mariam M Youssef
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Mark D Underwood
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
- Department of Psychiatry, Columbia University, New York, New York
| | - Yung-Yu Huang
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Shu-chi Hsiung
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Yan Liu
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Norman R Simpson
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Mihran J Bakalian
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | - Gorazd B Rosoklija
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
- Macedonian Academy of Sciences & Arts, Republic of Macedonia
| | - Andrew J Dwork
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
- Department of Psychiatry, Columbia University, New York, New York
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Victoria Arango
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
- Department of Psychiatry, Columbia University, New York, New York
| | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
- Department of Psychiatry, Columbia University, New York, New York
- Department of Radiology, Columbia University, New York, New York
| |
Collapse
|
247
|
Dong BE, Xue Y, Sakata K. The effect of enriched environment across ages: A study of anhedonia and BDNF gene induction. GENES BRAIN AND BEHAVIOR 2018; 17:e12485. [PMID: 29717802 DOI: 10.1111/gbb.12485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/19/2022]
Abstract
Enriched environment treatment (EET) is a potential intervention for depression by inducing brain-derived neurotrophic factor (BDNF). However, its age dependency remains unclear. We recently found that EET during early-life development (ED) was effective in increasing exploratory activity and anti-despair behavior, particularly in promoter IV-driven BDNF deficient mice (KIV), with the largest BDNF protein induction in the hippocampus and frontal cortex. Here, we further determined age dependency of EET effects on anhedonia and promoter-specific BDNF transcription, by using the sucrose preference test and qRT-PCR. Wild-type (WT) and KIV mice received 2 months of EET during ED, young-adulthood and old-adulthood (0-2, 2-4 and 12-14 months, respectively). All KIV groups showed reduced sucrose preference, which EET equally reversed regardless of age. EET increased hippocampal BDNF mRNA levels for all ages and genotypes, but increased frontal cortex BDNF mRNA levels only in ED KIV and old WT mice. Transcription by promoters I and IV was age-dependent in the hippocampus of WT mice: more effective induction of exon IV or I during ED or old-adulthood, respectively. Transcription by almost all 9 promoters was age-specific in the frontal cortex, mostly observed in ED KIV mice. After discontinuance of EET, the EET effects on anti-anhedonia and BDNF transcription in both regions persisted only in ED KIV mice. These results suggested that EET was equally effective in reversing anhedonia and inducing hippocampal BDNF transcription, but was more effective during ED in inducing frontal cortex BDNF transcription and for lasting anti-anhedonic and BDNF effects particularly in promoter IV-BDNF deficiency.
Collapse
Affiliation(s)
- B E Dong
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Y Xue
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - K Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
248
|
Hacimusalar Y, Eşel E. Suggested Biomarkers for Major Depressive Disorder. ACTA ACUST UNITED AC 2018; 55:280-290. [PMID: 30224877 DOI: 10.5152/npa.2017.19482] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 06/08/2017] [Indexed: 12/21/2022]
Abstract
Currently, the diagnosis of major depressive disorder (MDD) mainly relies on clinical examination and subjective evaluation of depressive symptoms. There is no non-invasive, quantitative test available today for the diagnosis of MDD. In MDD, exploration of biomarkers will be helpful in diagnosing the disorder as well as in choosing a treatment, and predicting the treatment response. In this article, it is aimed to review the findings of suggested biomarkers such as growth factors, cytokines and other inflammatory markers, oxidative stress markers, endocrine markers, energy balance hormones, genetic and epigenetic features, and neuroimaging in MDD and to evaluate how these findings contribute to the pathophysiology of MDD, the prediction of treatment response, severity of the disorder, and identification of subtypes. Among these, the findings related to the brain-derived neurotrophic factor, the hypothalamo-pituitary-adrenal axis, cytokines, and neuroimaging may be strong candidates for being biomarkers MDD, and may provide critical information in understanding biological etiology of depression. Although the findings are not sufficient yet, we think that the results of epigenetic studies will also provide very important contributions to the biomarker research in MDD. The availability of biomarkers in MDD will be an advancement that will facilitate the diagnosis of the disorder, treatment choices in the early stages, and prediction of the course of the disorder.
Collapse
Affiliation(s)
- Yunus Hacimusalar
- Department of Psychiatry, Kayseri Training and Research Hospital, Kayseri, Turkey
| | - Ertuğrul Eşel
- Department of Psychiatry, Erciyes University Faculty of Medicine, Kayseri, Turkey
| |
Collapse
|
249
|
Ye D, Zhang L, Fan W, Zhang X, Dong E. Genipin normalizes depression-like behavior induced by prenatal stress through inhibiting DNMT1. Epigenetics 2018. [PMID: 29522357 DOI: 10.1080/15592294.2018.1450033] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Synthetic antidepressants in current use for the complex etiopathogeneses of depression have slow response and remission as well as various unpleasant side effects. As a result, it is imperative to develop new antidepressants with more effectiveness and less severe side effects. Recent studies demonstrated that genipin, the aglycon of geniposide, extracted from Gardenia jasminoides Ellis has antidepressive effects. However, knowledge regarding the molecular mechanisms of its antidepressant effects remains limited. Employing a depression-like mouse model, we confirmed that genipin is capable of correcting depressions-like behaviors induced by prenatal stress in offspring from prenatally stressed dams (defined as PRS mice). In further experiments, we found that the effect of genipin on PRS mice occurs through DNA demethylation by inhibiting DNA methyltransferase 1 (DNMT1), normalizing the expression of reduced brain-derived neurotrophic factor (BDNF) in the hippocampus.
Collapse
Affiliation(s)
- Di Ye
- a Oncology Department, The Second Affiliated Hospital , Chongqing Medical University , Yuzhong District , Chongqing , China
| | - Li Zhang
- a Oncology Department, The Second Affiliated Hospital , Chongqing Medical University , Yuzhong District , Chongqing , China
| | - Weidong Fan
- a Oncology Department, The Second Affiliated Hospital , Chongqing Medical University , Yuzhong District , Chongqing , China
| | - Xianquan Zhang
- a Oncology Department, The Second Affiliated Hospital , Chongqing Medical University , Yuzhong District , Chongqing , China
| | - Erbo Dong
- a Oncology Department, The Second Affiliated Hospital , Chongqing Medical University , Yuzhong District , Chongqing , China
| |
Collapse
|
250
|
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]
|