1
|
Dehghanbanadaki H, Jimbo M, Fendereski K, Kunisaki J, Horns JJ, Ramsay JM, Gross KX, Pastuszak AW, Hotaling JM. Transgenerational effects of paternal exposures: the role of germline de novo mutations. Andrology 2025; 13:101-118. [PMID: 38396220 DOI: 10.1111/andr.13609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/02/2023] [Accepted: 01/23/2024] [Indexed: 02/25/2024]
Abstract
Germline de novo mutations (DNMs) refer to spontaneous mutations arising during gametogenesis, resulting in genetic changes within germ cells that are subsequently transmitted to the next generation. While the impact of maternal exposures on germline DNMs has been extensively studied, more recent studies have begun to highlight the increasing importance of the effects of paternal factors. In this review, we have summarized the existing literature on how various exposures experienced by fathers affect the germline DNM burden in their spermatozoa, as well as their consequences for semen analysis parameters, pregnancy outcomes, and offspring health. A growing body of literature supports the conclusion that advanced paternal age (APA) correlates with a higher germline DNM rate in offspring. Furthermore, lifestyle choices, environmental toxins, assisted reproductive techniques (ART), and chemotherapy are associated with the accumulation of paternal DNMs in spermatozoa, with deleterious consequences for pregnancy outcomes and offspring health. Ultimately, our review highlights the clear importance of the germline DNM mode of inheritance, and the current understanding of how this is affected by various paternal factors. In addition, we explore conflicting reports or gaps of knowledge that should be addressed in future research.
Collapse
Affiliation(s)
- Hojat Dehghanbanadaki
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Masaya Jimbo
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Kiarad Fendereski
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Jason Kunisaki
- Department of Human Genetics, University of Utah, Salt Lake City, Utah, USA
| | - Joshua J Horns
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Joemy M Ramsay
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Kelli X Gross
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - Alexander W Pastuszak
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| | - James M Hotaling
- Division of Urology, Department of Surgery, University of Utah Health, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
Derakhshan M, Kessler NJ, Hellenthal G, Silver MJ. Metastable epialleles in humans. Trends Genet 2024; 40:52-68. [PMID: 38000919 DOI: 10.1016/j.tig.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 11/26/2023]
Abstract
First identified in isogenic mice, metastable epialleles (MEs) are loci where the extent of DNA methylation (DNAm) is variable between individuals but correlates across tissues derived from different germ layers within a given individual. This property, termed systemic interindividual variation (SIV), is attributed to stochastic methylation establishment before germ layer differentiation. Evidence suggests that some putative human MEs are sensitive to environmental exposures in early development. In this review we introduce key concepts pertaining to human MEs, describe methods used to identify MEs in humans, and review their genomic features. We also highlight studies linking DNAm at putative human MEs to early environmental exposures and postnatal (including disease) phenotypes.
Collapse
Affiliation(s)
- Maria Derakhshan
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Noah J Kessler
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | - Matt J Silver
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul, The Gambia.
| |
Collapse
|
3
|
Drzymalla E, Crider KS, Wang A, Marta G, Khoury MJ, Rasooly D. Epigenome-wide association studies of prenatal maternal mental health and infant epigenetic profiles: a systematic review. Transl Psychiatry 2023; 13:377. [PMID: 38062042 PMCID: PMC10703876 DOI: 10.1038/s41398-023-02620-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 10/01/2023] [Accepted: 10/06/2023] [Indexed: 12/18/2023] Open
Abstract
Prenatal stress and poor maternal mental health are associated with adverse offspring outcomes; however, the biological mechanisms are unknown. Epigenetic modification has linked maternal health with offspring development. Epigenome-wide association studies (EWAS) have examined offspring DNA methylation profiles for association with prenatal maternal mental health to elucidate mechanisms of these complex relationships. The objective of this study is to provide a comprehensive, systematic review of EWASs of infant epigenetic profiles and prenatal maternal anxiety, depression, or depression treatment. We conducted a systematic literature search following PRISMA guidelines for EWAS studies between prenatal maternal mental health and infant epigenetics through May 22, 2023. Of 645 identified articles, 20 fulfilled inclusion criteria. We assessed replication of CpG sites among studies, conducted gene enrichment analysis, and evaluated the articles for quality and risk of bias. We found one repeated CpG site among the maternal depression studies; however, nine pairs of overlapping differentially methylatd regions were reported in at least two maternal depression studies. Gene enrichment analysis found significant pathways for maternal depression but not for any other maternal mental health category. We found evidence that these EWAS present a medium to high risk of bias. Exposure to prenatal maternal depression and anxiety or treatment for such was not consistently associated with epigenetic changes in infants in this systematic review and meta-analysis. Small sample size, potential bias due to exposure misclassification and statistical challenges are critical to address in future efforts to explore epigenetic modification as a potential mechanism by which prenatal exposure to maternal mental health disorders leads to adverse infant outcomes.
Collapse
Affiliation(s)
- Emily Drzymalla
- Division of Blood Disorders and Public Health Genomics, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Krista S Crider
- Infant Outcomes Research and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Arick Wang
- Infant Outcomes Research and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Muin J Khoury
- Division of Blood Disorders and Public Health Genomics, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Danielle Rasooly
- Division of Blood Disorders and Public Health Genomics, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
4
|
Marinho LSR, Chiarantin GMD, Ikebara JM, Cardoso DS, de Lima-Vasconcellos TH, Higa GSV, Ferraz MSA, De Pasquale R, Takada SH, Papes F, Muotri AR, Kihara AH. The impact of antidepressants on human neurodevelopment: Brain organoids as experimental tools. Semin Cell Dev Biol 2023; 144:67-76. [PMID: 36115764 DOI: 10.1016/j.semcdb.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/10/2022] [Accepted: 09/10/2022] [Indexed: 11/23/2022]
Abstract
The use of antidepressants during pregnancy benefits the mother's well-being, but the effects of such substances on neurodevelopment remain poorly understood. Moreover, the consequences of early exposure to antidepressants may not be immediately apparent at birth. In utero exposure to selective serotonin reuptake inhibitors (SSRIs) has been related to developmental abnormalities, including a reduced white matter volume. Several reports have observed an increased incidence of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) after prenatal exposure to SSRIs such as sertraline, the most widely prescribed SSRI. The advent of human-induced pluripotent stem cell (hiPSC) methods and assays now offers appropriate tools to test the consequences of such compounds for neurodevelopment in vitro. In particular, hiPSCs can be used to generate cerebral organoids - self-organized structures that recapitulate the morphology and complex physiology of the developing human brain, overcoming the limitations found in 2D cell culture and experimental animal models for testing drug efficacy and side effects. For example, single-cell RNA sequencing (scRNA-seq) and electrophysiological measurements on organoids can be used to evaluate the impact of antidepressants on the transcriptome and neuronal activity signatures in developing neurons. While the analysis of large-scale transcriptomic data depends on dimensionality reduction methods, electrophysiological recordings rely on temporal data series to discriminate statistical characteristics of neuronal activity, allowing for the rigorous analysis of the effects of antidepressants and other molecules that affect the developing nervous system, especially when applied in combination with relevant human cellular models such as brain organoids.
Collapse
Affiliation(s)
| | | | - Juliane Midori Ikebara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | | | - Guilherme Shigueto Vilar Higa
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil; Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | | | - Roberto De Pasquale
- Department of Physiology and Biophysics, Biomedical Sciences Institute I, São Paulo University, São Paulo, SP 05508-000, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil
| | - Fabio Papes
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; Center for Medicinal Chemistry, University of Campinas, Campinas, SP 13083-875, Brazil; Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, Center for Academic Research and Training in Anthropogeny, Kavli Institute for Brain and Mind, Archealization Center (ArchC), La Jolla, CA 92037, USA.
| | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Universidade Federal do ABC, São Bernardo do Campo, SP 09606-045, Brazil.
| |
Collapse
|
5
|
Olstad EW, Nordeng HME, Sandve GK, Lyle R, Gervin K. Effects of prenatal exposure to (es)citalopram and maternal depression during pregnancy on DNA methylation and child neurodevelopment. Transl Psychiatry 2023; 13:149. [PMID: 37147306 PMCID: PMC10163054 DOI: 10.1038/s41398-023-02441-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
Studies assessing associations between prenatal exposure to antidepressants, maternal depression, and offspring DNA methylation (DNAm) have been inconsistent. Here, we investigated whether prenatal exposure to citalopram or escitalopram ((es)citalopram) and maternal depression is associated with differences in DNAm. Then, we examined if there is an interaction effect of (es)citalopram exposure and DNAm on offspring neurodevelopmental outcomes. Finally, we investigated whether DNAm at birth correlates with neurodevelopmental trajectories in childhood. We analyzed DNAm in cord blood from the Norwegian Mother, Father and Child Cohort Study (MoBa) biobank. MoBa contains questionnaire data on maternal (es)citalopram use and depression during pregnancy and information about child neurodevelopmental outcomes assessed by internationally recognized psychometric tests. In addition, we retrieved ADHD diagnoses from the Norwegian Patient Registry and information on pregnancies from the Medical Birth Registry of Norway. In total, 958 newborn cord blood samples were divided into three groups: (1) prenatal (es)citalopram exposed (n = 306), (2) prenatal maternal depression exposed (n = 308), and (3) propensity score-selected controls (n = 344). Among children exposed to (es)citalopram, there were more ADHD diagnoses and symptoms and delayed communication and psychomotor development. We did not identify differential DNAm associated with (es)citalopram or depression, nor any interaction effects on neurodevelopmental outcomes throughout childhood. Trajectory modeling identified subgroups of children following similar developmental patterns. Some of these subgroups were enriched for children exposed to maternal depression, and some subgroups were associated with differences in DNAm at birth. Interestingly, several of the differentially methylated genes are involved in neuronal processes and development. These results suggest DNAm as a potential predictive molecular marker of later abnormal neurodevelopmental outcomes, but we cannot conclude whether DNAm links prenatal (es)citalopram exposure or maternal depression with child neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Emilie Willoch Olstad
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
- UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway.
| | - Hedvig Marie Egeland Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Geir Kjetil Sandve
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Robert Lyle
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristina Gervin
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
6
|
Camerota M, Graw S, Everson TM, McGowan EC, Hofheimer JA, O'Shea TM, Carter BS, Helderman JB, Check J, Neal CR, Pastyrnak SL, Smith LM, Dansereau LM, DellaGrotta SA, Marsit CJ, Lester BM. Prenatal risk factors and neonatal DNA methylation in very preterm infants. Clin Epigenetics 2021; 13:171. [PMID: 34507616 PMCID: PMC8434712 DOI: 10.1186/s13148-021-01164-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Prenatal risk factors are related to poor health and developmental outcomes for infants, potentially via epigenetic mechanisms. We tested associations between person-centered prenatal risk profiles, cumulative prenatal risk models, and epigenome-wide DNA methylation (DNAm) in very preterm neonates. METHODS We studied 542 infants from a multi-center study of infants born < 30 weeks postmenstrual age. We assessed 24 prenatal risk factors via maternal report and medical record review. Latent class analysis was used to define prenatal risk profiles. DNAm was quantified from neonatal buccal cells using the Illumina MethylationEPIC Beadarray. RESULTS We identified three latent profiles of women: a group with few risk factors (61%) and groups with elevated physical (26%) and psychological (13%) risk factors. Neonates born to women in higher risk subgroups had differential DNAm at 2 CpG sites. Higher cumulative prenatal risk was associated with methylation at 15 CpG sites, 12 of which were located in genes previously linked to physical and mental health and neurodevelopment. CONCLUSION We observed associations between prenatal risk factors and DNAm in very preterm infants using both person-centered and cumulative risk approaches. Epigenetics offers a potential biological indicator of prenatal risk exposure.
Collapse
Affiliation(s)
- Marie Camerota
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI, 02905, USA.
| | - Stefan Graw
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Todd M Everson
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Elisabeth C McGowan
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, RI, USA
| | - Julie A Hofheimer
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - T Michael O'Shea
- Department of Pediatrics, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Brian S Carter
- Department of Pediatrics-Neonatology, Children's Mercy Hospital, Kansas City, MO, USA
| | - Jennifer B Helderman
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jennifer Check
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Charles R Neal
- Department of Pediatrics, University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | - Steven L Pastyrnak
- Department of Pediatrics, Spectrum Health-Helen DeVos Hospital, Grand Rapids, MI, USA
| | - Lynne M Smith
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lynne M Dansereau
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI, 02905, USA
| | - Sheri A DellaGrotta
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI, 02905, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Barry M Lester
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, 101 Dudley Street, Providence, RI, 02905, USA
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
7
|
Burstein O, Simon N, Simchon-Tenenbaum Y, Rehavi M, Franko M, Shamir A, Doron R. Moderation of the transgenerational transference of antenatal stress-induced anxiety. Transl Psychiatry 2021; 11:268. [PMID: 33947833 PMCID: PMC8094124 DOI: 10.1038/s41398-021-01383-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/10/2021] [Accepted: 03/29/2021] [Indexed: 01/08/2023] Open
Abstract
Maternal stress has debilitating implications for both mother and child, including increased risk for anxiety. The current COVID-19 pandemic escalates these phenomena, thus, urging the need to further explore and validate feasible therapeutic options. Unlike the protracted nature of clinical studies, animal models could offer swift evidence. Prominent candidates for treatment are selective serotonin reuptake inhibitors (SSRIs) to the mother, that putatively accommodate maternal functioning, and, thereby, also protect the child. However, SSRIs might have deleterious effects. It is important to assess whether SSRIs and other pharmacotherapies can moderate the transference of anxiety by soothing maternal anxiety and to examine the extent of offspring's exposure to the drugs via lactation. To our knowledge, the possibility that antenatal stress exacerbates lactation-driven exposure to SSRIs has not been tested yet. Thirty ICR-outbred female mice were exposed to stress during gestation and subsequently administered with either the SSRI, escitalopram, or the novel herbal candidate, shan-zha, during lactation. Upon weaning, both dams' and pups' anxiety-like behavior and serum escitalopram levels were assessed. The major findings of the current study show that both agents moderated the antenatal stress-induced transgenerational transference of anxiety by ameliorating dams' anxiety. Interestingly though, pups' exposure to escitalopram via lactation was exacerbated by antenatal stress. The latter finding provides a significant insight into the mechanism of lactation-driven exposure to xenobiotics and calls for a further consideration vis-à-vis the administration of other drugs during breastfeeding.
Collapse
Affiliation(s)
- Or Burstein
- Department of Psychology, Bar Ilan University, Ramat Gan, Israel
| | - Noam Simon
- School of Behavioral Science, The Academic College of Tel Aviv-Yaffo, Tel Aviv-Yaffo, Israel
| | - Yaarit Simchon-Tenenbaum
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Dr. Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv, Israel
| | - Moshe Rehavi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Dr. Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv, Israel
| | - Motty Franko
- Department of Psychology, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- Department of Education and Psychology, The Open University of Israel, Raanana, Israel
| | - Alon Shamir
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Mazor Mental Health Center, Akko, Israel
| | - Ravid Doron
- Department of Education and Psychology, The Open University of Israel, Raanana, Israel.
| |
Collapse
|
8
|
Olstad EW, Nordeng HME, Gervin K. Prenatal medication exposure and epigenetic outcomes: a systematic literature review and recommendations for prenatal pharmacoepigenetic studies. Epigenetics 2021; 17:357-380. [PMID: 33926354 PMCID: PMC8993058 DOI: 10.1080/15592294.2021.1903376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
When used during pregnancy, analgesics and psychotropics pass the placenta to enter the foetal circulation and may induce epigenetic modifications. Where such modifications occur and whether they disrupt normal foetal developme nt, are currently unanswered questions. This field of prenatal pharmacoepigenetics has received increasing attention, with several studies reporting associations between in utero medication exposure and offspring epigenetic outcomes. Nevertheless, no recent systematic review of the literature is available. Therefore, the objectives of this review were to (i) provide an overview of the literature on the association of prenatal exposure to psychotropics a nd analgesics with epigenetic outcomes, and (ii) suggest recommendations for future studies within prenatal pharmacoepigenetics. We performed systematic literature searches in five databases. The eligible studies assessed human prenatal exposure to psychotropics or analgesics, with epigenetic analyses of offspring tissue as an outcome. We identified 18 eligible studies including 4,419 neonates exposed to either antidepressants, antiepileptic drugs, paracetamol, acetylsalicylic acid, or methadone. The epigenetic outcome in all studies was DNA methylation in cord blood, placental tissue or buccal cells. Although most studies found significant differences in DNA methylation upon medication exposure, almost no differences were persistent across studies for similar medications and sequencing methods. The reviewed studies were challenging to compare due to poor transparency in reporting, and heterogeneous methodology, design, genome coverage, and statistical modelling. We propose 10 recommendations for future prenatal pharmacoepigenetic studies considering both epidemiological and epigenetic perspectives. These recommendations may improve the quality, comparability, and clinical relevance of such studies. PROSPERO registration ID: CRD42020166675.
Collapse
Affiliation(s)
- Emilie Willoch Olstad
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Hedvig Marie Egeland Nordeng
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Kristina Gervin
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.,Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Letourneau N, Ntanda H, Jong VL, Mahinpey N, Giesbrecht G, Ross KM. Prenatal maternal distress and immune cell epigenetic profiles at 3-months of age. Dev Psychobiol 2021; 63:973-984. [PMID: 33569773 DOI: 10.1002/dev.22103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Prenatal maternal distress predicts altered offspring immune outcomes, potentially via altered epigenetics. The role of different kinds of prenatal maternal distress on DNA methylation profiles is not understood. METHODS A sample of 117 women (APrON cohort) were followed from pregnancy to the postpartum period. Maternal distress (depressive symptoms, pregnancy-specific anxiety, stressful life events) were assessed mid-pregnancy, late-pregnancy, and 3-months postpartum. DNA methylation profiles were obtained from 3-month-old blood samples. Principal component analysis identified two epigenetic components, characterized as Immune Signaling and DNA Transcription through gene network analysis. Covariates were maternal demographics, pre-pregnancy body mass index, child sex, birth gestational age, and postpartum maternal distress. Penalized regression (LASSO) models were used. RESULTS Late-pregnancy stressful life events, b = 0.006, early-pregnancy depressive symptoms, b = 0.027, late-pregnancy depressive symptoms, b = 0.014, and pregnancy-specific anxiety during late pregnancy, b = -0.631, were predictive of the Immune Signaling component, suggesting that these aspects of maternal distress could affect methylation in offspring immune signaling pathways. Only early-pregnancy depressive symptoms was predictive of the DNA Transcription component, b = -0.0004, suggesting that this aspect of maternal distress is implicated in methylation of offspring DNA transcription pathways. CONCLUSIONS Exposure timing and kind of prenatal maternal distress could matter in the prediction of infant immune epigenetic profiles.
Collapse
Affiliation(s)
| | | | - Victor L Jong
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
10
|
Gaining a deeper understanding of social determinants of preterm birth by integrating multi-omics data. Pediatr Res 2021; 89:336-343. [PMID: 33188285 PMCID: PMC7898277 DOI: 10.1038/s41390-020-01266-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
In the US, high rates of preterm birth (PTB) and profound Black-White disparities in PTB have persisted for decades. This review focuses on the role of social determinants of health (SDH), with an emphasis on maternal stress, in PTB disparity and biological embedding. It covers: (1) PTB disparity in US Black women and possible contributors; (2) the role of SDH, highlighting maternal stress, in the persistent racial disparity of PTB; (3) epigenetics at the interface between genes and environment; (4) the role of the genome in modifying maternal stress-PTB associations; (5) recent advances in multi-omics studies of PTB; and (6) future perspectives on integrating multi-omics with SDH to elucidate the Black-White disparity in PTB. Available studies have indicated that neither environmental exposures nor genetics alone can adequately explain the Black-White PTB disparity. Preliminary yet promising findings of epigenetic and gene-environment interaction studies underscore the value of integrating SDH with multi-omics in prospective birth cohort studies, especially among high-risk Black women. In an era of rapid advancements in biomedical sciences and technologies and a growing number of prospective birth cohort studies, we have unprecedented opportunities to advance this field and finally address the long history of health disparities in PTB. IMPACT: This review provides an overview of social determinants of health (SDH) with a focus on maternal stress and its role on Black-White disparity in preterm birth (PTB). It summarizes the available literature on the interplay of maternal stress with key biological layers (e.g., individual genome and epigenome in response to environmental stressors) and significant knowledge gaps. It offers perspectives that such knowledge may provide deeper insight into how SDH affects PTB and why some women are more vulnerable than others and underscores the critical need for integrating SDH with multi-omics in prospective birth cohort studies, especially among high-risk Black women.
Collapse
|
11
|
Wang Y, Qian M, Tang D, Herbstman J, Perera F, Wang S. A powerful and flexible weighted distance-based method incorporating interactions between DNA methylation and environmental factors on health outcomes. Bioinformatics 2020; 36:653-659. [PMID: 31504174 DOI: 10.1093/bioinformatics/btz630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 08/19/2019] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Deoxyribonucleic acid (DNA) methylation plays a crucial role in human health. Studies have demonstrated associations between DNA methylation and environmental factors with evidence also supporting the idea that DNA methylation may modify the risk of environmental factors on health outcomes. However, due to high dimensionality and low study power, current studies usually focus on finding differential methylation on health outcomes at CpG level or gene level combining multiple CpGs and/or finding environmental effects on health outcomes but ignoring their interactions on health outcomes. Here we introduce the idea of a pseudo-data matrix constructed with cross-product terms between CpGs and environmental factors that are able to capture their interactions. We then develop a powerful and flexible weighted distance-based method with the pseudo-data matrix where association strength was used as weights on CpGs, environmental factors and their interactions to up-weight signals and down-weight noises in distance calculations. RESULTS We compared the power of this novel approach and several comparison methods in simulated datasets and the Mothers and Newborns birth cohort of the Columbia Center for Children's Environmental Health to determine whether prenatal polycyclic aromatic hydrocarbons interacts with DNA methylation in association with Attention Deficit Hyperactivity Disorder and Mental Development Index at age 3. AVAILABILITY AND IMPLEMENTATION An R code for the proposed method Dw-M-E-int together with a tutorial and a sample dataset is available for downloading from http://www.columbia.edu/∼sw2206/softwares.htm. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ya Wang
- Department of Biostatistics, New York, NY 10032, USA
| | - Min Qian
- Department of Biostatistics, New York, NY 10032, USA
| | - Deliang Tang
- Columbia Center for Children's Environmental Health, Department of Environmental Health Science, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Julie Herbstman
- Columbia Center for Children's Environmental Health, Department of Environmental Health Science, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Frederica Perera
- Columbia Center for Children's Environmental Health, Department of Environmental Health Science, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Shuang Wang
- Department of Biostatistics, New York, NY 10032, USA
| |
Collapse
|
12
|
Stress During Pregnancy and Epigenetic Modifications to Offspring DNA: A Systematic Review of Associations and Implications for Preterm Birth. J Perinat Neonatal Nurs 2020; 34:134-145. [PMID: 32332443 PMCID: PMC7185032 DOI: 10.1097/jpn.0000000000000471] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Offspring born preterm (ie, before 37 weeks of gestation) are more likely to die or experience long-standing illness than full-term offspring. Maternal genetic variants (ie, heritable, stable variations in the genetic code) and epigenetic modifications (ie, chemical modifications to the genetic code that can affect which genes are turned on or off) in response to stress have been implicated in preterm birth. Fetal genetic variants have been linked to preterm birth though the role of offspring epigenetics in preterm birth remains understudied. This systematic review synthesizes the literature examining associations among stress during pregnancy and epigenetic modifications to offspring DNA, with 25 reports identified. Ten reports examined DNA methylation (ie, addition/removal of methyl groups to/from DNA) across the epigenome. The remainder examined DNA methylation near genes of interest, primarily genes linked to hypothalamic-pituitary-adrenal axis function (NR3C1, FKBP51), growth/immune function (IGF2), and socioemotional regulation (SLC6A4, OXTR). The majority of reports noted associations among stress and offspring DNA methylation, primarily when perceived stress, anxiety, or depression served as the predictor. Findings suggest that differences in offspring epigenetic patterns may play a role in stress-associated preterm birth and serve as targets for novel interventions.
Collapse
|
13
|
SSRIs and SNRIs (SRI) in Pregnancy: Effects on the Course of Pregnancy and the Offspring: How Far Are We from Having All the Answers? Int J Mol Sci 2019; 20:ijms20102370. [PMID: 31091646 PMCID: PMC6567187 DOI: 10.3390/ijms20102370] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/31/2022] Open
Abstract
Serotonin has important roles in the development of the brain and other organs. Manipulations of synaptic serotonin by drugs such as serotonin reuptake inhibitors (SRI) or serotonin norepinephrine reuptake inhibitors (SNRI) might alter their development and function. Of interest, most studies on the outcome of prenatal exposure to SRI in human have not found significant embryonic or fetal damage, except for a possible, slight increase in cardiac malformations. In up to a third of newborns exposed to SRI, exposure may induce transient neonatal behavioral changes (poor neonatal adaptation) and increased rate of persistent pulmonary hypertension. Prenatal SRI may also cause slight motor delay and language impairment but these are transient. The data on the possible association of prenatal SRIs with autism spectrum disorder (ASD) are inconsistent, and seem to be related to pre-pregnancy treatment or to maternal depression. Prenatal SRIs also appear to affect the hypothalamic hypophyseal adrenal (HPA) axis inducing epigenetic changes, but the long-term consequences of these effects on humans are as yet unknown. SRIs are metabolized in the liver by several cytochrome P450 (CYP) enzymes. Faster metabolism of most SRIs in late pregnancy leads to lower maternal concentrations, and thus potentially to decreased efficacy which is more prominent in women that are rapid metabolizers. Studies suggest that the serotonin transporter SLC6A4 promoter is associated with adverse neonatal outcomes after SRI exposure. Since maternal depression may adversely affect the child's development, one has to consider the risk of SRI discontinuation on the fetus and the child. As with any drug treatment in pregnancy, the benefits to the mother should be considered versus the possible hazards to the developing embryo/fetus.
Collapse
|
14
|
Cardenas A, Faleschini S, Cortes Hidalgo A, Rifas-Shiman SL, Baccarelli AA, DeMeo DL, Litonjua AA, Neumann A, Felix JF, Jaddoe VWV, El Marroun H, Tiemeier H, Oken E, Hivert MF, Burris HH. Prenatal maternal antidepressants, anxiety, and depression and offspring DNA methylation: epigenome-wide associations at birth and persistence into early childhood. Clin Epigenetics 2019; 11:56. [PMID: 30925934 PMCID: PMC6441191 DOI: 10.1186/s13148-019-0653-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Maternal mood disorders and their treatment during pregnancy may have effects on the offspring epigenome. We aim to evaluate associations of maternal prenatal antidepressant use, anxiety, and depression with cord blood DNA methylation across the genome at birth and test for persistence of associations in early and mid-childhood blood DNA. METHODS A discovery phase was conducted in Project Viva, a prospective pre-birth cohort study with external replication in an independent cohort, the Generation R Study. In Project Viva, pregnant women were recruited between 1999 and 2002 in Eastern Massachusetts, USA. In the Generation R Study, pregnant women were recruited between 2002 and 2006 in Rotterdam, the Netherlands. In Project Viva, 479 infants had data on maternal antidepressant use, anxiety, depression, and cord blood DNA methylation, 120 children had DNA methylation measured in early childhood (~ 3 years), and 460 in mid-childhood (~ 7 years). In the Generation R Study, 999 infants had data on maternal antidepressants and cord blood DNA methylation. The prenatal antidepressant prescription was obtained from medical records. At-mid pregnancy, symptoms of anxiety and depression were assessed with the Pregnancy-Related Anxiety Scale and the Edinburgh Postnatal Depression Scale in Project Viva and with the Brief Symptom Inventory in the Generation R Study. Genome-wide DNA methylation was measured using the Infinium HumanMethylation450 BeadChip in both cohorts. RESULTS In Project Viva, 2.9% (14/479) pregnant women were prescribed antidepressants, 9.0% (40/445) experienced high pregnancy-related anxiety, and 8.2% (33/402) reported symptoms consistent with depression. Newborns exposed to antidepressants in pregnancy had 7.2% lower DNA methylation (95% CI, - 10.4, - 4.1; P = 1.03 × 10-8) at cg22159528 located in the gene body of ZNF575, and this association replicated in the Generation R Study (β = - 2.5%; 95% CI - 4.2, - 0.7; P = 0.006). In Project Viva, the association persisted in early (β = - 6.2%; 95% CI - 10.7, - 1.6) but not mid-childhood. We observed cohort-specific associations for maternal anxiety and depression in Project Viva that did not replicate. CONCLUSIONS The ZNF575 gene is involved in transcriptional regulation but specific functions are largely unknown. Given the widespread use of antidepressants in pregnancy, as well as the effects of exposure to anxiety and depression, implications of potential fetal epigenetic programming by these risk factors and their impacts on development merit further investigation.
Collapse
Affiliation(s)
- Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | | | - Andrea Cortes Hidalgo
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Sheryl L. Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY USA
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, NY USA
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
| | - Janine F. Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Vincent W. V. Jaddoe
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Hanan El Marroun
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC – Sophia Children’s Hospital, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Emily Oken
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA USA
| | - Heather H. Burris
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
15
|
Genome-wide epigenetic signatures of childhood adversity in early life: Opportunities and challenges. J Dev Orig Health Dis 2019; 10:65-72. [PMID: 30744719 DOI: 10.1017/s2040174418000843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Maternal adversity and fetal glucocorticoid exposure has long-term effects on cardiovascular, metabolic and behavioral systems in offspring that can persist throughout the lifespan. These data, along with other environmental exposure data, implicate epigenetic modifications as potential mechanisms for long-term effects of maternal exposures on adverse health outcomes in offspring. Advances in microarray, sequencing and bioinformatic approaches have enabled recent studies to examine the genome-wide epigenetic response to maternal adversity. Studies of maternal exposures to xenobiotics such as arsenic and smoking have been performed at birth to examine fetal epigenomic signatures in cord blood relating to adult health outcomes. However, there have been no epigenomic studies examining these effects in animal models. On the other hand, to date, only a few studies of the effects of maternal psychosocial stress have been performed in human infants, and the majority of animal studies have examined epigenomic outcomes in adulthood. In terms of maternal exposure to excess glucocorticoids by synthetic glucocorticoid treatment, there has been no epigenetic study performed in humans and only a few studies undertaken in animal models. This review emphasizes the importance of examining biomarkers of exposure to adversity throughout development to identify individuals at risk and to target interventions. Thus, research performed at birth will be reviewed. In addition, potential subject characteristics associated with epigenetic modifications, technical considerations, the selection of target tissues and combining human studies with animal models will be discussed in relation to the design of experiments in this field of study.
Collapse
|
16
|
Epigenetic age acceleration is associated with allergy and asthma in children in Project Viva. J Allergy Clin Immunol 2019; 143:2263-2270.e14. [PMID: 30738172 DOI: 10.1016/j.jaci.2019.01.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Epigenetic clocks have been suggested to capture one feature of the complexity between aging and the epigenome. However, little is known about the epigenetic clock in childhood allergy and asthma. OBJECTIVE We sought to examine associations of DNA methylation age (DNAmAge) and epigenetic age acceleration with childhood allergy and asthma. METHODS We calculated DNAmAge and age acceleration at birth, early childhood, and midchildhood based on the IlluminaHumanMethylation450BeadChip in Project Viva. We evaluated epigenetic clock associations with allergy and asthma using covariate-adjusted linear and logistic regressions. We attempted to replicate our findings in the Genetics of Asthma in Costa Rica Study. RESULTS At midchildhood (mean age, 7.8 years) in Project Viva, DNAmAge and age acceleration were cross-sectionally associated with greater total serum IgE levels and greater odds of atopic sensitization. Every 1-year increase in intrinsic epigenetic age acceleration was associated with a 1.22 (95% CI, 1.07-1.39), 1.17 (95% CI, 1.03-1.34), and 1.29 (95% CI, 1.12-1.49) greater odds of atopic sensitization and environmental and food allergen sensitization. DNAmAge and extrinsic epigenetic age acceleration were also cross-sectionally associated with current asthma at midchildhood. DNAmAge and age acceleration at birth and early childhood were not associated with midchildhood allergy or asthma. The midchildhood association between age acceleration and atopic sensitization were replicated in an independent data set. CONCLUSIONS Because the epigenetic clock might reflect immune and developmental components of biological aging, our study suggests pathways through which molecular epigenetic mechanisms of immunity, development, and maturation can interact along the age axis and associate with childhood allergy and asthma by midchildhood.
Collapse
|
17
|
Lupu D, Varshney MK, Mucs D, Inzunza J, Norinder U, Loghin F, Nalvarte I, Rüegg J. Fluoxetine Affects Differentiation of Midbrain Dopaminergic Neurons In Vitro. Mol Pharmacol 2018; 94:1220-1231. [PMID: 30115672 DOI: 10.1124/mol.118.112342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/11/2018] [Indexed: 01/12/2023] Open
Abstract
Recent meta-analyses found an association between prenatal exposure to the antidepressant fluoxetine (FLX) and an increased risk of autism in children. This developmental disorder has been related to dysfunctions in the brains' rewards circuitry, which, in turn, has been linked to dysfunctions in dopaminergic (DA) signaling. The present study investigated if FLX affects processes involved in dopaminergic neuronal differentiation. Mouse neuronal precursors were differentiated into midbrain dopaminergic precursor cells (mDPCs) and concomitantly exposed to clinically relevant doses of FLX. Subsequently, dopaminergic precursors were evaluated for expression of differentiation and stemness markers using quantitative polymerase chain reaction. FLX treatment led to increases in early regional specification markers orthodenticle homeobox 2 (Otx2) and homeobox engrailed-1 and -2 (En1 and En2). On the other hand, two transcription factors essential for midbrain dopaminergic (mDA) neurogenesis, LIM homeobox transcription factor 1 α (Lmx1a) and paired-like homeodomain transcription factor 3 (Pitx3) were downregulated by FLX treatment. The stemness marker nestin (Nes) was increased, whereas the neuronal differentiation marker β3-tubulin (Tubb3) decreased. Additionally, we observed that FLX modulates the expression of several genes associated with autism spectrum disorder and downregulates the estrogen receptors (ERs) α and β Further investigations using ERβ knockout (BERKO) mDPCs showed that FLX had no or even opposite effects on several of the genes analyzed. These findings suggest that FLX affects differentiation of the dopaminergic system by increasing production of dopaminergic precursors, yet decreasing their maturation, partly via interference with the estrogen system.
Collapse
Affiliation(s)
- Diana Lupu
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Mukesh K Varshney
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Daniel Mucs
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - José Inzunza
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Ulf Norinder
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Felicia Loghin
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Ivan Nalvarte
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| | - Joëlle Rüegg
- Department of Toxicology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (D.L., F.L.); Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden (M.K.V., J.I., I.N.); Unit of Work Environment Toxicology, Institute of Environmental Medicine (D.M.) and Department of Clinical Neuroscience (J.R.), Karolinska Institutet, Stockholm, Sweden; Department Computer and Systems Sciences, Stockholm University, Kista, Sweden (U.N.); and Swetox, Unit of Toxicology Sciences, Karolinska Institutet, Södertälje, Sweden (D.L., D.M., U.N., J.R.)
| |
Collapse
|
18
|
Gartstein MA, Skinner MK. Prenatal influences on temperament development: The role of environmental epigenetics. Dev Psychopathol 2018; 30:1269-1303. [PMID: 29229018 PMCID: PMC5997513 DOI: 10.1017/s0954579417001730] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This review summarizes current knowledge and outlines future directions relevant to questions concerning environmental epigenetics and the processes that contribute to temperament development. Links between prenatal adversity, epigenetic programming, and early manifestations of temperament are important in their own right, also informing our understanding of biological foundations for social-emotional development. In addition, infant temperament attributes represent key etiological factors in the onset of developmental psychopathology, and studies elucidating their prenatal foundations expand our understanding of developmental origins of health and disease. Prenatal adversity can take many forms, and this overview is focused on the environmental effects of stress, toxicants, substance use/psychotropic medication, and nutrition. Dysregulation associated with attention-deficit/hyperactivity-disruptive disorders was noted in the context of maternal substance use and toxicant exposures during gestation, as well as stress. Although these links can be made based on the existing literature, currently few studies directly connect environmental influences, epigenetic programming, and changes in brain development/behavior. The chain of events starting with environmental inputs and resulting in alterations to gene expression, physiology, and behavior of the organism is driven by epigenetics. Epigenetics provides the molecular mechanism of how environmental factors impact development and subsequent health and disease, including early brain and temperament development.
Collapse
Affiliation(s)
- Maria A. Gartstein
- Department of Psychology, Washington State University, Pullman, WA-99164-4820, USA
| | - Michael K. Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA-99164-4236, USA
| |
Collapse
|
19
|
Hannon E, Schendel D, Ladd-Acosta C, Grove J, Hansen CS, Andrews SV, Hougaard DM, Bresnahan M, Mors O, Hollegaard MV, Bækvad-Hansen M, Hornig M, Mortensen PB, Børglum AD, Werge T, Pedersen MG, Nordentoft M, Buxbaum J, Daniele Fallin M, Bybjerg-Grauholm J, Reichenberg A, Mill J. Elevated polygenic burden for autism is associated with differential DNA methylation at birth. Genome Med 2018; 10:19. [PMID: 29587883 PMCID: PMC5872584 DOI: 10.1186/s13073-018-0527-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/20/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a severe neurodevelopmental disorder characterized by deficits in social communication and restricted, repetitive behaviors, interests, or activities. The etiology of ASD involves both inherited and environmental risk factors, with epigenetic processes hypothesized as one mechanism by which both genetic and non-genetic variation influence gene regulation and pathogenesis. The aim of this study was to identify DNA methylation biomarkers of ASD detectable at birth. METHODS We quantified neonatal methylomic variation in 1263 infants-of whom ~ 50% went on to subsequently develop ASD-using DNA isolated from archived blood spots taken shortly after birth. We used matched genotype data from the same individuals to examine the molecular consequences of ASD-associated genetic risk variants, identifying methylomic variation associated with elevated polygenic burden for ASD. In addition, we performed DNA methylation quantitative trait loci (mQTL) mapping to prioritize target genes from ASD GWAS findings. RESULTS We identified robust epigenetic signatures of gestational age and prenatal tobacco exposure, confirming the utility of DNA methylation data generated from neonatal blood spots. Although we did not identify specific loci showing robust differences in neonatal DNA methylation associated with later ASD, there was a significant association between increased polygenic burden for autism and methylomic variation at specific loci. Each unit of elevated ASD polygenic risk score was associated with a mean increase in DNA methylation of - 0.14% at two CpG sites located proximal to a robust GWAS signal for ASD on chromosome 8. CONCLUSIONS This study is the largest analysis of DNA methylation in ASD undertaken and the first to integrate genetic and epigenetic variation at birth. We demonstrate the utility of using a polygenic risk score to identify molecular variation associated with disease, and of using mQTL to refine the functional and regulatory variation associated with ASD risk variants.
Collapse
Affiliation(s)
- Eilis Hannon
- University of Exeter Medical School, University of Exeter, RILD Building, Level 4, Barrack Rd, Exeter, EX2 5DW UK
| | - Diana Schendel
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Jakob Grove
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Christine Søholm Hansen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
| | - Shan V. Andrews
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - David Michael Hougaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Michaeline Bresnahan
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, New York, USA
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Mads Vilhelm Hollegaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Mady Hornig
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, New York, USA
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, USA
| | - Preben Bo Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University; Aarhus University Hospital, Risskov, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Anders D. Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Centre for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Giørtz Pedersen
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
- Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Merete Nordentoft
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Mental Health Services in the Capital Region of Denmark, Mental Health Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Joseph Buxbaum
- Department of Psychiatry, Mount Sinai School of Medicine, New York City, USA
| | - M. Daniele Fallin
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, New York, USA
- Department of Psychiatry, Columbia University, New York, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD USA
| | - Jonas Bybjerg-Grauholm
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- Center for Neonatal Screening, Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Abraham Reichenberg
- Department of Psychiatry, Mount Sinai School of Medicine, New York City, USA
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, RILD Building, Level 4, Barrack Rd, Exeter, EX2 5DW UK
| |
Collapse
|
20
|
Dou J, Schmidt RJ, Benke KS, Newschaffer C, Hertz-Picciotto I, Croen LA, Iosif AM, LaSalle JM, Fallin MD, Bakulski KM. Cord blood buffy coat DNA methylation is comparable to whole cord blood methylation. Epigenetics 2018; 13:108-116. [PMID: 29451060 PMCID: PMC5836975 DOI: 10.1080/15592294.2017.1417710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/29/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
Cord blood DNA methylation is associated with numerous health outcomes and environmental exposures. Whole cord blood DNA reflects all nucleated blood cell types, while centrifuging whole blood separates red blood cells, generating a white blood cell buffy coat. Both sample types are used in DNA methylation studies. Cell types have unique methylation patterns and processing can impact cell distributions, which may influence comparability. We evaluated differences in cell composition and DNA methylation between cord blood buffy coat and whole cord blood samples. Cord blood DNA methylation was measured with the Infinium EPIC BeadChip (Illumina) in eight individuals, each contributing buffy coat and whole blood samples. We analyzed principal components (PC) of methylation, performed hierarchical clustering, and computed correlations of mean-centered methylation between pairs. We conducted moderated t-tests on single sites and estimated cell composition. DNA methylation PCs were associated with individual (PPC1 = 1.4 × 10-9; PPC2 = 2.9 × 10-5; PPC3 = 3.8 × 10-5; PPC4 = 4.2 × 10-6; PPC5 = 9.9 × 10-13, PPC6 = 1.3 × 10-11) and not with sample type (PPC1-6>0.7). Samples hierarchically clustered by individual. Pearson correlations of mean-centered methylation between paired samples ranged from r = 0.66 to r = 0.87. No individual site significantly differed between buffy coat and whole cord blood when adjusting for multiple comparisons (five sites had unadjusted P<10-5). Estimated cell type proportions did not differ by sample type (P = 0.46), and estimated proportions were highly correlated between paired samples (r = 0.99). Differences in methylation and cell composition between buffy coat and whole cord blood are much lower than inter-individual variation, demonstrating that both sample preparation types can be analytically combined and compared.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Rebecca J. Schmidt
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | - Kelly S. Benke
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University
| | - Craig Newschaffer
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University
- A.J. Drexel Autism Institute, Drexel University
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | | | - Ana-Maria Iosif
- Department of Public Health Sciences, University of California Davis
- University of California Davis, MIND Institute
| | - Janine M. LaSalle
- University of California Davis, MIND Institute
- Department of Medical Microbiology and Immunology, Genome Center, University of California Davis
| | - M. Daniele Fallin
- Department of Mental Health, Bloomberg School of Public Health, Johns Hopkins University
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins University
| | - Kelly M. Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan
| |
Collapse
|
21
|
Gemmel M, Bögi E, Ragan C, Hazlett M, Dubovicky M, van den Hove DL, Oberlander TF, Charlier TD, Pawluski JL. Perinatal selective serotonin reuptake inhibitor medication (SSRI) effects on social behaviors, neurodevelopment and the epigenome. Neurosci Biobehav Rev 2018; 85:102-116. [DOI: 10.1016/j.neubiorev.2017.04.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/10/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
|
22
|
McLaughlin P, Mactier H, Gillis C, Hickish T, Parker A, Liang WJ, Osselton MD. Increased DNA Methylation of ABCB1, CYP2D6, and OPRM1 Genes in Newborn Infants of Methadone-Maintained Opioid-Dependent Mothers. J Pediatr 2017; 190:180-184.e1. [PMID: 28867064 DOI: 10.1016/j.jpeds.2017.07.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/05/2017] [Accepted: 07/13/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To investigate whether in utero opioid exposure, which has been linked to adverse neurodevelopmental and social outcomes, is associated with altered DNA methylation of opioid-related genes at birth. STUDY DESIGN Observational cohort study of 21 healthy methadone-maintained opioid-dependent mother-infant dyads consecutively delivered at >36 weeks of gestation, and 2 comparator groups: smoking, "deprived" opioid-naïve mother-infant dyads (n = 17) and nonsmoking, "affluent" opioid-naïve mother-infant dyads (n = 15). DNA methylation of ABCB1, CYP2D6, and OPRM1 genes for mothers and babies was determined from buccal swabs. Plasma methadone concentrations were additionally measured for methadone-maintained opioid-dependent mothers. RESULTS DNA methylation for ABCB1 and CYP2D6 was similar in opioid-naïve infants compared with their mothers, but was less for OPRM1 (3 ± 1.6% vs 8 ± 1%, P < .0005). Opioid-exposed newborns had similar DNA methylation to their mothers for all genes studied and greater methylation of ABCB1 (18 ± 4.8% vs 3 ± 0.5%), CYP2D6 (92 ± 1.2% vs 89 ± 2.4%), and OPRM1 (8 ± 0.3% vs 3 ± 1.6%) compared with opioid-naïve newborns (P < .0005 for all 3 genes). Infant DNA methylation was not related to birth weight, length of hospital stay, maternal smoking, dose or plasma concentration of methadone at delivery, or postcode of residence. CONCLUSIONS In utero exposure to opioids is associated with increased methylation of opioid-related genes in the newborn infant. It is not clear whether these findings are due to opioid exposure per se or other associated lifestyle factors.
Collapse
Affiliation(s)
- Poppy McLaughlin
- Department of Archaeology, Anthropology, and Forensic Science, Bournemouth University, Poole, United Kingdom
| | - Helen Mactier
- Neonatal Unit, Princess Royal Maternity, Glasgow, United Kingdom.
| | - Cheryl Gillis
- Neonatal Unit, Princess Royal Maternity, Glasgow, United Kingdom
| | - Tamas Hickish
- Department of Molecular Pathology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Anton Parker
- Department of Molecular Pathology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Wei-Jun Liang
- Department of Archaeology, Anthropology, and Forensic Science, Bournemouth University, Poole, United Kingdom
| | - M David Osselton
- Department of Archaeology, Anthropology, and Forensic Science, Bournemouth University, Poole, United Kingdom
| |
Collapse
|
23
|
Neurobehavioral risks of SSRIs in pregnancy: Comparing human and animal data. Reprod Toxicol 2017; 72:191-200. [DOI: 10.1016/j.reprotox.2017.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 04/20/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022]
|
24
|
Descalzi G, Mitsi V, Purushothaman I, Gaspari S, Avrampou K, Loh YHE, Shen L, Zachariou V. Neuropathic pain promotes adaptive changes in gene expression in brain networks involved in stress and depression. Sci Signal 2017; 10:10/471/eaaj1549. [PMID: 28325815 DOI: 10.1126/scisignal.aaj1549] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuropathic pain is a complex chronic condition characterized by various sensory, cognitive, and affective symptoms. A large percentage of patients with neuropathic pain are also afflicted with depression and anxiety disorders, a pattern that is also seen in animal models. Furthermore, clinical and preclinical studies indicate that chronic pain corresponds with adaptations in several brain networks involved in mood, motivation, and reward. Chronic stress is also a major risk factor for depression. We investigated whether chronic pain and stress affect similar molecular mechanisms and whether chronic pain can affect gene expression patterns that are involved in depression. Using two mouse models of neuropathic pain and depression [spared nerve injury (SNI) and chronic unpredictable stress (CUS)], we performed next-generation RNA sequencing and pathway analysis to monitor changes in gene expression in the nucleus accumbens (NAc), the medial prefrontal cortex (mPFC), and the periaqueductal gray (PAG). In addition to finding unique transcriptome profiles across these regions, we identified a substantial number of signaling pathway-associated genes with similar changes in expression in both SNI and CUS mice. Many of these genes have been implicated in depression, anxiety, and chronic pain in patients. Our study provides a resource of the changes in gene expression induced by long-term neuropathic pain in three distinct brain regions and reveals molecular connections between pain and chronic stress.
Collapse
Affiliation(s)
- Giannina Descalzi
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vasiliki Mitsi
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sevasti Gaspari
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kleopatra Avrampou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yong-Hwee Eddie Loh
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Li Shen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venetia Zachariou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
25
|
Ryan J, Mansell T, Fransquet P, Saffery R. Does maternal mental well-being in pregnancy impact the early human epigenome? Epigenomics 2017; 9:313-332. [PMID: 28140666 DOI: 10.2217/epi-2016-0118] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is considerable interest in the potential nongenetic transmission of a suite of mental health conditions across generations, with epigenetics emerging as a candidate mediator of such effects. This review summarizes findings from 22 studies measuring candidate gene DNA methylation and seven epigenome-wide association studies of offspring epigenetic profile in women with adverse mental wellbeing measures (stress, depression or anxiety) in pregnancy. Despite some compelling evidence to suggest an association, there is a lack of reproducible findings, potentially linked to a number of limitations to this research and the field more broadly. Large cohorts with well characterized exposures across pregnancy are now needed. There is exciting potential that epigenetics may help explain some of the link between maternal wellbeing and child health outcomes, thereby informing novel interventions, but future studies must address current limitations to advance translational knowledge in this area.
Collapse
Affiliation(s)
- Joanne Ryan
- Department of Cancer & Disease Epigenetics, Murdoch Childrens Research Institute, Royal Childrens Hospital, & Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.,Department of Epidemiology & Preventive Medicine, School of Public Health & Preventive Medicine, Monash University, Prahran, Victoria, Australia.,Inserm U1061, Hopital La Colombiere, University Montpellier, Montpellier, France
| | - Toby Mansell
- Department of Cancer & Disease Epigenetics, Murdoch Childrens Research Institute, Royal Childrens Hospital, & Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Fransquet
- Department of Cancer & Disease Epigenetics, Murdoch Childrens Research Institute, Royal Childrens Hospital, & Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Richard Saffery
- Department of Cancer & Disease Epigenetics, Murdoch Childrens Research Institute, Royal Childrens Hospital, & Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
26
|
Pishva E, Rutten BPF, van den Hove D. DNA Methylation in Major Depressive Disorder. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 978:185-196. [PMID: 28523547 DOI: 10.1007/978-3-319-53889-1_10] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epigenetic mechanisms regulate gene expression, influencing protein levels and ultimately shaping phenotypes during life. However, both stochastic epigenetic variations and environmental reprogramming of the epigenome might influence neurodevelopment and ageing, and this may contribute to the origins of mental ill-health. Studying the role of epigenetic mechanisms is challenging, as genotype-, tissue- and cell type-dependent epigenetic changes have to be taken into account, while the nature of mental disorders also poses significant challenges for linking them with biological profiles. In this chapter, we summarise the current evidence suggesting the role of DNA methylation as a key epigenetic mechanism in major depressive disorder.
Collapse
Affiliation(s)
- Ehsan Pishva
- Complex Disease Epigenetic Group, University of Exeter Medical School, RILD Building, RD&E Hospital Wonford, Barrack Road, Exeter, EX2 5DW, UK. .,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, The Netherlands.
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, The Netherlands
| | - Daniel van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, The Netherlands.,Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, Wurzburg, 97080, Germany
| |
Collapse
|
27
|
Viuff ACF, Pedersen LH, Kyng K, Staunstrup NH, Børglum A, Henriksen TB. Antidepressant medication during pregnancy and epigenetic changes in umbilical cord blood: a systematic review. Clin Epigenetics 2016; 8:94. [PMID: 27610205 PMCID: PMC5015265 DOI: 10.1186/s13148-016-0262-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/30/2016] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION Epigenetic mechanisms are important for the regulation of gene expression and differentiation in the fetus and the newborn child. Symptoms of maternal depression and antidepressant use affects up to 20 % of pregnant women, and may lead to epigenetic changes with life-long impact on child health. The aim of this review is to investigate whether there is an association between exposure to maternal antidepressants during pregnancy and epigenetic changes in the newborn. MATERIAL AND METHODS Systematic literature searches were performed in MEDLINE and EMBASE combining MeSH terms covering epigenetic changes, use of antidepressant medication, pregnancy and newborns. A keyword search was also performed. We included studies on pregnant women and their children where there was a history of maternal depressed mood or anxiety, a reported use of antidepressant medication, and measurements of epigenetic changes in umbilical cord blood. Studies using genome-wide or candidate-based epigenetic analyses were included. Citations and references from the included articles were investigated to locate further relevant articles. The completeness of reporting as well as the risk of bias and confounding was assessed. RESULTS Six studies were included. They all investigated methylation changes. Genome-wide methylation changes were examined in 184 children and methylation status in specific genes was examined in 96 children exposed to antidepressant medication. Three of the studies found an association between use of antidepressant medication during pregnancy and methylation status at various CpG sites measured in cord blood of the newborn. One of these studies found an association in African-Americans, but not Caucasians. The remaining three studies found associations between maternal mood and epigenetic changes in umbilical cord blood but no association between epigenetic changes and maternal use of antidepressant medication. CONCLUSION The included studies have not established a clear association between use of antidepressant medication during pregnancy and epigenetic changes in the cord blood. Future studies using newer, more wide-ranging epigenetic methods could discover possible new differentially methylated sites. Larger sample sizes and good validity of exposures are warranted in order to adjust for level of maternal depression, other maternal illness, maternal use of other types of medication, and maternal ethnicity. PROSPERO registration number: CRD42015026575.
Collapse
Affiliation(s)
- Anne-Cathrine F. Viuff
- Perinatal Epidemiology Research Unit, Pediatric Department, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Lars Henning Pedersen
- Department of Obstetrics and Gynecology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Kasper Kyng
- Perinatal Epidemiology Research Unit, Pediatric Department, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Nicklas Heine Staunstrup
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Risskov, Denmark
- Department of Biomedicine and Centre for Integrative Sequencing, iSEQ, University of Aarhus, Aarhus C, 8000 Denmark
- Translational Neuropsychiatric Unit, Aarhus University Hospital, Risskov, 8240 Denmark
| | - Anders Børglum
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Risskov, Denmark
- Department of Biomedicine and Centre for Integrative Sequencing, iSEQ, University of Aarhus, Aarhus C, 8000 Denmark
| | - Tine Brink Henriksen
- Perinatal Epidemiology Research Unit, Pediatric Department, Aarhus University Hospital Skejby, Aarhus, Denmark
| |
Collapse
|
28
|
Gentile S, Fusco ML. Placental and fetal effects of antenatal exposure to antidepressants or untreated maternal depression. J Matern Fetal Neonatal Med 2016; 30:1189-1199. [PMID: 27379818 DOI: 10.1080/14767058.2016.1209184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To assess systematically the effects of antidepressants and untreated maternal depression on human placenta and the developing fetus. METHODS Pertinent medical literature information was identified using MEDLINE/PubMed, SCOPUS and EMBASE. Electronic searches, limited to human studies published in English, provided 21 studies reporting primary data on placental and fetal effects of antidepressant exposure or untreated gestational depression. RESULTS The impact of antidepressants and non-medicated maternal depression on placental functioning and fetal biochemical architecture seems to be demonstrated, although its clinical significance remains unclear. More robust data seem to indicate that exposure to either antidepressants or untreated maternal depression may induce epigenetic changes and interfere with the physiological fetal behavior. Two cases of iatrogenic fetal tachyarrhythmia have also been reported. CONCLUSIONS Future research should clarify the clinical relevance of the impact of antidepressant and untreated maternal depression exposure on placental functioning. Moreover, ultrasound studies investigating fetal responses to antidepressants or maternal depressive symptoms are mandatory. This assessment should be performed during the whole duration of gestational period, when different fetal behavioral patterns become progressively detectable. Analyses of biochemical and epigenetic modifications associated with maternal mood symptoms and antidepressant treatment should also be implemented.
Collapse
Affiliation(s)
- Salvatore Gentile
- a ASL Salerno, Department of Mental Health, Mental Health Center Cava de' Tirreni, Vietri sul Mare , Salerno , Italy.,b Department of Neurosciences , Division of Perinatal Psychiatry, Medical School "Federico II", University of Naples , Naples , Italy
| | - Maria Luigia Fusco
- c Mental Health Institute, Torre Annunziata , Naples , Italy , and.,d Department of Developmental Psychology , Post-Graduate School of Psychotherapy (SIPGI), Torre Annunziata, Naples , Italy
| |
Collapse
|
29
|
Kumsta R, Marzi SJ, Viana J, Dempster EL, Crawford B, Rutter M, Mill J, Sonuga-Barke EJS. Severe psychosocial deprivation in early childhood is associated with increased DNA methylation across a region spanning the transcription start site of CYP2E1. Transl Psychiatry 2016; 6:e830. [PMID: 27271856 PMCID: PMC4931613 DOI: 10.1038/tp.2016.95] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/22/2016] [Accepted: 03/31/2016] [Indexed: 01/09/2023] Open
Abstract
Exposure to adverse rearing environments including institutional deprivation and severe childhood abuse is associated with an increased risk for mental and physical health problems across the lifespan. Although the mechanisms mediating these effects are not known, recent work in rodent models suggests that epigenetic processes may be involved. We studied the impact of severe early-life adversity on epigenetic variation in a sample of adolescents adopted from the severely depriving orphanages of the Romanian communist era in the 1980s. We quantified buccal cell DNA methylation at ~400 000 sites across the genome in Romanian adoptees exposed to either extended (6-43 months; n=16) or limited duration (<6 months; n=17) of severe early-life deprivation, in addition to a matched sample of UK adoptees (n=16) not exposed to severe deprivation. Although no probe-wise differences remained significant after controlling for the number of probes tested, we identified an exposure-associated differentially methylated region (DMR) spanning nine sequential CpG sites in the promoter-regulatory region of the cytochrome P450 2E1 gene (CYP2E1) on chromosome 10 (corrected P=2.98 × 10(-5)). Elevated DNA methylation across this region was also associated with deprivation-related clinical markers of impaired social cognition. Our data suggest that environmental insults of sufficient biological impact during early development are associated with long-lasting epigenetic changes, potentially reflecting a biological mechanism linking the effects of early-life adversity to cognitive and neurobiological phenotypes.
Collapse
Affiliation(s)
- R Kumsta
- Department of Genetic Psychology, Faculty of Psychology, Ruhr-University Bochum, Bochum, Germany
| | - S J Marzi
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - J Viana
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - E L Dempster
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - B Crawford
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - M Rutter
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - J Mill
- MRC Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - E J S Sonuga-Barke
- Department of Psychology, Institute for Disorders of Impulse and Attention, Developmental Brain-Behavior Laboratory, University of Southampton, Southampton, UK
- Department of Experimental Clinical and Health Psychology, Ghent University, Ghent, Belgium
| |
Collapse
|
30
|
Gartstein MA, Hookenson KV, Brain U, Devlin AM, Grunau RE, Oberlander TF. Sculpting infant soothability: the role of prenatal SSRI antidepressant exposure and neonatalSLC6A4methylation status. Dev Psychobiol 2016; 58:745-58. [DOI: 10.1002/dev.21414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/12/2016] [Indexed: 01/27/2023]
Affiliation(s)
- Maria A. Gartstein
- Department of Psychology; Washington State University; Pullman Washington
| | - Kaia V. Hookenson
- Department of Pediatrics; University of British Columbia, Child and Family Research Institute; Vancouver British Columbia Canada
| | - Ursula Brain
- Department of Pediatrics; University of British Columbia, Child and Family Research Institute; Vancouver British Columbia Canada
| | - Angela M. Devlin
- Department of Pediatrics; University of British Columbia, Child and Family Research Institute; Vancouver British Columbia Canada
| | - Ruth E. Grunau
- Department of Pediatrics; University of British Columbia, Child and Family Research Institute; Vancouver British Columbia Canada
| | - Tim F. Oberlander
- Department of Pediatrics; University of British Columbia, Child and Family Research Institute; Vancouver British Columbia Canada
| |
Collapse
|
31
|
Cacabelos R, Torrellas C. Epigenetics of Aging and Alzheimer's Disease: Implications for Pharmacogenomics and Drug Response. Int J Mol Sci 2015; 16:30483-543. [PMID: 26703582 PMCID: PMC4691177 DOI: 10.3390/ijms161226236] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/16/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
Epigenetic variability (DNA methylation/demethylation, histone modifications, microRNA regulation) is common in physiological and pathological conditions. Epigenetic alterations are present in different tissues along the aging process and in neurodegenerative disorders, such as Alzheimer’s disease (AD). Epigenetics affect life span and longevity. AD-related genes exhibit epigenetic changes, indicating that epigenetics might exert a pathogenic role in dementia. Epigenetic modifications are reversible and can potentially be targeted by pharmacological intervention. Epigenetic drugs may be useful for the treatment of major problems of health (e.g., cancer, cardiovascular disorders, brain disorders). The efficacy and safety of these and other medications depend upon the efficiency of the pharmacogenetic process in which different clusters of genes (pathogenic, mechanistic, metabolic, transporter, pleiotropic) are involved. Most of these genes are also under the influence of the epigenetic machinery. The information available on the pharmacoepigenomics of most drugs is very limited; however, growing evidence indicates that epigenetic changes are determinant in the pathogenesis of many medical conditions and in drug response and drug resistance. Consequently, pharmacoepigenetic studies should be incorporated in drug development and personalized treatments.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| | - Clara Torrellas
- EuroEspes Biomedical Research Center, Institute of Medical Science and Genomic Medicine, 15165-Bergondo, Corunna, Spain.
- Chair of Genomic Medicine, Camilo José Cela University, 28692-Madrid, Spain.
| |
Collapse
|