1
|
Legault LM, Dupas T, Breton-Larrivée M, Filion-Bienvenue F, Lemieux A, Langford-Avelar A, McGraw S. Sex-specific DNA methylation and gene expression changes in mouse placentas after early preimplantation alcohol exposure. ENVIRONMENT INTERNATIONAL 2024; 192:109014. [PMID: 39321537 DOI: 10.1016/j.envint.2024.109014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/27/2024]
Abstract
During pregnancy, exposure to alcohol represents an environmental insult capable of negatively impacting embryonic development. This influence can stem from disruption of molecular profiles, ultimately leading to manifestation of fetal alcohol spectrum disorder. Despite the central role of the placenta in proper embryonic development and successful pregnancy, studies on the placenta in a prenatal alcohol exposure and fetal alcohol spectrum disorder context are markedly lacking. Here, we employed a well-established model for preimplantation alcohol exposure, specifically targeting embryonic day 2.5, corresponding to the 8-cell stage. The exposure was administered to pregnant C57BL/6 female mice through subcutaneous injection, involving two doses of either 2.5 g/kg 50 % ethanol or an equivalent volume of saline at 2-hour intervals. Morphology, DNA methylation and gene expression patterns were assessed in male and female late-gestation (E18.5) placentas. While overall placental morphology was not altered, we found a significant decrease in male ethanol-exposed embryo weights. When looking at molecular profiles, we uncovered numerous differentially methylated regions (DMRs; 991 in males; 1309 in females) and differentially expressed genes (DEGs; 1046 in males; 340 in females) in the placentas. Remarkably, only 21 DMRs and 54 DEGs were common to both sexes, which were enriched for genes involved in growth factor response pathways. Preimplantation alcohol exposure had a greater impact on imprinted genes expression in male placentas (imprinted DEGs: 18 in males; 1 in females). Finally, by using machine learning model (L1 regularization), we were able to precisely discriminate control and ethanol-exposed placentas based on their specific DNA methylation patterns. This is the first study demonstrating that preimplantation alcohol exposure alters the DNA methylation and transcriptomic profiles of late-gestation placentas in a sex-specific manner. Our findings highlight that the DNA methylation profiles of the placenta could serve as a potent predictive molecular signature for early preimplantation alcohol exposure.
Collapse
Affiliation(s)
- Lisa-Marie Legault
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| | - Thomas Dupas
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| | - Mélanie Breton-Larrivée
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| | - Fannie Filion-Bienvenue
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| | - Anthony Lemieux
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada.
| | - Alexandra Langford-Avelar
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| | - Serge McGraw
- CHU Ste-Justine Azrieli Research Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada; Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
2
|
Zeng X, Cai Y, Wu M, Chen H, Sun M, Yang H. An overview of current advances in perinatal alcohol exposure and pathogenesis of fetal alcohol spectrum disorders. J Neurodev Disord 2024; 16:20. [PMID: 38643092 PMCID: PMC11031898 DOI: 10.1186/s11689-024-09537-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
The adverse use of alcohol is a serious global public health problem. Maternal alcohol consumption during pregnancy usually causes prenatal alcohol exposure (PAE) in the developing fetus, leading to a spectrum of disorders known as fetal alcohol spectrum disorders (FASD) and even fetal alcohol syndrome (FAS) throughout the lifelong sufferers. The prevalence of FASD is approximately 7.7 per 1,000 worldwide, and is even higher in developed regions. Generally, Ethanol in alcoholic beverages can impair embryonic neurological development through multiple pathways leading to FASD. Among them, the leading mechanism of FASDs is attributed to ethanol-induced neuroinflammatory damage to the central nervous system (CNS). Although the underlying molecular mechanisms remain unclear, the remaining multiple pathological mechanisms is likely due to the neurotoxic damage of ethanol and the resultant neuronal loss. Regardless of the molecular pathway, the ultimate outcome of the developing CNS exposed to ethanol is almost always the destruction and apoptosis of neurons, which leads to the reduction of neurons and further the development of FASD. In this review, we systematically summarize the current research progress on the pathogenesis of FASD, which hopefully provides new insights into differential early diagnosis, treatment and prevention for patents with FASD.
Collapse
Affiliation(s)
- Xingdong Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Yongle Cai
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Mengyan Wu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Haonan Chen
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215031, China.
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Khalifa A, Palu R, Perkins AE, Volz A. Prenatal alcohol exposure alters expression of genes involved in cell adhesion, immune response, and toxin metabolism in adolescent rat hippocampus. PLoS One 2024; 19:e0293425. [PMID: 38271377 PMCID: PMC10810486 DOI: 10.1371/journal.pone.0293425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/11/2023] [Indexed: 01/27/2024] Open
Abstract
Prenatal alcohol exposure (PAE) can result in mild to severe consequences for children throughout their lives, with this range of symptoms referred to as Fetal Alcohol Spectrum Disorders (FASD). These consequences are thought to be linked to changes in gene expression and transcriptional programming in the brain, but the identity of those changes, and how they persist into adolescence are unclear. In this study, we isolated RNA from the hippocampus of adolescent rats exposed to ethanol during prenatal development and compared gene expression to controls. Briefly, dams were either given free access to standard chow ad libitum (AD), pair-fed a liquid diet (PF) or were given a liquid diet with ethanol (6.7% ethanol, ET) throughout gestation (gestational day (GD) 0-20). All dams were given control diet ad libitum beginning on GD 20 and throughout parturition and lactation. Hippocampal tissue was collected from adolescent male and female offspring (postnatal day (PD) 35-36). Exposure to ethanol caused widespread downregulation of many genes as compared to control rats. Gene ontology analysis demonstrated that affected pathways included cell adhesion, toxin metabolism, and immune responses. Interestingly, these differences were not strongly affected by sex. Furthermore, these changes were consistent when comparing ethanol-exposed rats to pair-fed controls provided with a liquid diet and those fed ad libitum on a standard chow diet. We conclude from this study that changes in genetic architecture and the resulting neuronal connectivity after prenatal exposure to alcohol continue through adolescent development. Further research into the consequences of specific gene expression changes on neural and behavioral changes will be vital to our understanding of the FASD spectrum of diseases.
Collapse
Affiliation(s)
- Amal Khalifa
- Department of Computer Science, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Rebecca Palu
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Amy E. Perkins
- Department of Psychology, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| | - Avery Volz
- Department of Biological Sciences, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
- Department of Psychology, Purdue University Fort Wayne, Fort Wayne, IN, United States of America
| |
Collapse
|
4
|
Breuer L, Greenmyer JR, Wilson T. Clinical Diagnosis and Management of Fetal Alcohol Spectrum Disorder and Sensory Processing Disorder in Children. CHILDREN (BASEL, SWITZERLAND) 2024; 11:108. [PMID: 38255421 PMCID: PMC10814837 DOI: 10.3390/children11010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Fetal alcohol spectrum disorder (FASD) is commonly misdiagnosed because of the complexity of presentation and multiple diagnostic criteria. FASD includes four categorical entities (fetal alcohol syndrome, partial fetal alcohol syndrome, alcohol related neurodevelopmental disorder, and alcohol related birth defects). The four FASD diagnostic criteria are facial dysmorphology, growth deficiency, central nervous system dysfunction, and prenatal alcohol exposure. Sensory processing disorders (SPDs) are common in FASD and are observed as inappropriate behavioral responses to environmental stimuli. These can be either a sensory-based motor disorder, sensory discrimination disorder, or sensory modulation disorder. A child with SPD may experience challenges with their fine motor coordination, gross motor coordination, organizational challenges, or behavioral regulation impairments. FASD requires a multidimensional approach to intervention. Although FASD cannot be cured, symptoms can be managed with sleep-based therapies, sensory integration, and cognitive therapies. This paper reviews SPDs in FASD and the interventions that can be used by practitioners to help improve their therapeutic management, although it is unlikely that any single intervention will be the right choice for all patients.
Collapse
Affiliation(s)
- Lorel Breuer
- Department of Biology, Winona State University, Winona, MN 55987, USA;
| | - Jacob R. Greenmyer
- Pediatric Hematology and Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Ted Wilson
- Department of Biology, Winona State University, Winona, MN 55987, USA;
| |
Collapse
|
5
|
Tooley KB, Chucair-Elliott AJ, Ocañas SR, Machalinski AH, Pham KD, Hoolehan W, Kulpa AM, Stanford DR, Freeman WM. Differential usage of DNA modifications in neurons, astrocytes, and microglia. Epigenetics Chromatin 2023; 16:45. [PMID: 37953264 PMCID: PMC10642035 DOI: 10.1186/s13072-023-00522-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Cellular identity is determined partly by cell type-specific epigenomic profiles that regulate gene expression. In neuroscience, there is a pressing need to isolate and characterize the epigenomes of specific CNS cell types in health and disease. In this study, we developed an in vivo tagging mouse model (Camk2a-NuTRAP) for paired isolation of neuronal DNA and RNA without cell sorting and then used this model to assess epigenomic regulation, DNA modifications in particular, of gene expression between neurons and glia. RESULTS After validating the cell-specificity of the Camk2a-NuTRAP model, we performed TRAP-RNA-Seq and INTACT-whole genome oxidative bisulfite sequencing (WGoxBS) to assess the neuronal translatome and epigenome in the hippocampus of young mice (4 months old). WGoxBS findings were validated with enzymatic methyl-Seq (EM-Seq) and nanopore sequencing. Comparing neuronal data to microglial and astrocytic data from NuTRAP models, microglia had the highest global mCG levels followed by astrocytes and then neurons, with the opposite pattern observed for hmCG and mCH. Differentially modified regions between cell types were predominantly found within gene bodies and distal intergenic regions, rather than proximal promoters. Across cell types there was a negative correlation between DNA modifications (mCG, mCH, hmCG) and gene expression at proximal promoters. In contrast, a negative correlation of gene body mCG and a positive relationship between distal promoter and gene body hmCG with gene expression was observed. Furthermore, we identified a neuron-specific inverse relationship between mCH and gene expression across promoter and gene body regions. CONCLUSIONS Neurons, astrocytes, and microglia demonstrate different genome-wide levels of mCG, hmCG, and mCH that are reproducible across analytical methods. However, modification-gene expression relationships are conserved across cell types. Enrichment of differential modifications across cell types in gene bodies and distal regulatory elements, but not proximal promoters, highlights epigenomic patterning in these regions as potentially greater determinants of cell identity. These findings also demonstrate the importance of differentiating between mC and hmC in neuroepigenomic analyses, as up to 30% of what is conventionally interpreted as mCG can be hmCG, which often has a different relationship to gene expression than mCG.
Collapse
Affiliation(s)
- Kyla B Tooley
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Sarah R Ocañas
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Adeline H Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Kevin D Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Walker Hoolehan
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Adam M Kulpa
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - David R Stanford
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
6
|
Tooley KB, Chucair-Elliott AJ, Ocañas SR, Machalinski AH, Pham KD, Stanford DR, Freeman WM. Differential usage of DNA modifications in neurons, astrocytes, and microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.05.543497. [PMID: 37333391 PMCID: PMC10274634 DOI: 10.1101/2023.06.05.543497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Cellular identity is determined partly by cell type-specific epigenomic profiles that regulate gene expression. In neuroscience, there is a pressing need to isolate and characterize the epigenomes of specific CNS cell types in health and disease. This is especially true as for DNA modifications where most data are derived from bisulfite sequencing that cannot differentiate between DNA methylation and hydroxymethylation. In this study, we developed an in vivo tagging mouse model (Camk2a-NuTRAP) for paired isolation of neuronal DNA and RNA without cell sorting and then used this model to assess epigenomic regulation of gene expression between neurons and glia. Results After validating the cell-specificity of the Camk2a-NuTRAP model, we performed TRAP-RNA-Seq and INTACT whole genome oxidative bisulfite sequencing to assess the neuronal translatome and epigenome in the hippocampus of young mice (3 months old). These data were then compared to microglial and astrocytic data from NuTRAP models. When comparing the different cell types, microglia had the highest global mCG levels followed by astrocytes and then neurons, with the opposite pattern observed for hmCG and mCH. Differentially modified regions between cell types were predominantly found within gene bodies and distal intergenic regions, with limited differences occurring within proximal promoters. Across cell types there was a negative correlation between DNA modifications (mCG, mCH, hmCG) and gene expression at proximal promoters. In contrast, a negative correlation of mCG with gene expression within the gene body while a positive relationship between distal promoter and gene body hmCG and gene expression was observed. Furthermore, we identified a neuron-specific inverse relationship between mCH and gene expression across promoter and gene body regions. Conclusions In this study, we identified differential usage of DNA modifications across CNS cell types, and assessed the relationship between DNA modifications and gene expression in neurons and glia. Despite having different global levels, the general modification-gene expression relationship was conserved across cell types. The enrichment of differential modifications in gene bodies and distal regulatory elements, but not proximal promoters, across cell types highlights epigenomic patterning in these regions as potentially greater determinants of cell identity.
Collapse
Affiliation(s)
- Kyla B. Tooley
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Ana J. Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Sarah R. Ocañas
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Adeline H. Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Kevin D. Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - David R. Stanford
- Center for Biomedical Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Willard M. Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Department of Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| |
Collapse
|
7
|
Sennsfelder L, Guilly S, Leruste S, Hoareau L, Léocadie W, Beuvain P, Nekaa M, Bagard M, Robin S, Lanneaux J, Etchebarren L, Tallot M, Spodenkiewicz M, Alessandri JL, Morel G, Blanluet M, Gueguen P, Roy-Doray B. Description of Copy Number Variations in a Series of Children and Adolescents with FASD in Reunion Island. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10040694. [PMID: 37189943 DOI: 10.3390/children10040694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Fetal Alcohol Spectrum Disorders (FASD) are the most common cause of neurocognitive impairment and social inadaptation, affecting 1 birth in 100. Despite the existence of precise diagnostic criteria, the diagnosis remains difficult, often confounded with other genetic syndromes or neurodevelopmental disorders. Since 2016, Reunion Island has been a pilot region for the identification, diagnosis, and care of FASD in France. OBJECTIVE To evaluate the prevalence and the types of Copy Number Variations (CNV) in FASD patients. METHODS A retrospective chart review of 101 patients diagnosed with FASD in the Reference Center for developmental anomalies and in the FASD Diagnostic Center of the University Hospital was performed. Records of all patients were reviewed to obtain their medical history, family history, clinical phenotype, and investigations, including genetic testing (CGH- or SNP-array). RESULTS A rate of 20.8% (n = 21) of CNVs was found including 57% (12/21) of pathogenic variants and 29% (6/21) of variants of uncertain signification (VUS). CONCLUSION A particularly high number of CNVs was found in children and adolescents with FASD. It reinforces the plea for a multidisciplinary approach for developmental disorders to explore both environmental factors, such as avoidable teratogens and intrinsic vulnerabilities, especially genetic determinants.
Collapse
Affiliation(s)
- Laëtitia Sennsfelder
- Laboratoire EPI (Etudes pharmaco-immunologiques), UFR Santé, Université de La Réunion, CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Susie Guilly
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Sébastien Leruste
- CIC 1410 (Centre d'Investigation Clinique), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
- UFR Santé, Université de La Réunion, 97410 Saint-Pierre, France
| | - Ludovic Hoareau
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Willy Léocadie
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Pauline Beuvain
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Meïssa Nekaa
- Centre Ressources TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), Fondation Père Favron, CHU (Centre Hospitalier Universitaire) de La Réunion, 97546 Saint-Pierre, France
| | - Maïté Bagard
- Centre Ressources TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), Fondation Père Favron, CHU (Centre Hospitalier Universitaire) de La Réunion, 97546 Saint-Pierre, France
| | - Stéphanie Robin
- Centre Diagnostic TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
| | - Justine Lanneaux
- Centre Diagnostic TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
| | - Léa Etchebarren
- Centre Diagnostic TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
| | - Marilyn Tallot
- Centre Diagnostic TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
| | - Michel Spodenkiewicz
- CIC 1410 (Centre d'Investigation Clinique), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
- Pôle de Santé Mentale, CHU (Centre Hospitalier Universitaire) de La Réunion, 97448 Saint-Pierre, France
| | - Jean-Luc Alessandri
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs Sud-Ouest Occitanie Réunion, Site Constitutif de La Réunion, 97400 Saint-Denis, France
| | - Godelieve Morel
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs Sud-Ouest Occitanie Réunion, Site Constitutif de La Réunion, 97400 Saint-Denis, France
| | - Maud Blanluet
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Paul Gueguen
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
| | - Bérénice Roy-Doray
- Laboratoire EPI (Etudes pharmaco-immunologiques), UFR Santé, Université de La Réunion, CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
- Service de Génétique, CHU (Centre Hospitalier Universitaire) de La Réunion, La Réunion, 97400 Saint-Denis, France
- CIC 1410 (Centre d'Investigation Clinique), CHU (Centre Hospitalier Universitaire) de La Réunion, 97400 Saint-Denis, France
- Centre Ressources TSAF (Troubles du Spectre de l'Alcoolisation Fœtale), Fondation Père Favron, CHU (Centre Hospitalier Universitaire) de La Réunion, 97546 Saint-Pierre, France
- Centre de Référence Anomalies du Développement et Syndromes Malformatifs Sud-Ouest Occitanie Réunion, Site Constitutif de La Réunion, 97400 Saint-Denis, France
| |
Collapse
|
8
|
Adams JW, Negraes PD, Truong J, Tran T, Szeto RA, Guerra BS, Herai RH, Teodorof-Diedrich C, Spector SA, Del Campo M, Jones KL, Muotri AR, Trujillo CA. Impact of alcohol exposure on neural development and network formation in human cortical organoids. Mol Psychiatry 2023; 28:1571-1584. [PMID: 36385168 PMCID: PMC10208963 DOI: 10.1038/s41380-022-01862-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Prenatal alcohol exposure is the foremost preventable etiology of intellectual disability and leads to a collection of diagnoses known as Fetal Alcohol Spectrum Disorders (FASD). Alcohol (EtOH) impacts diverse neural cell types and activity, but the precise functional pathophysiological effects on the human fetal cerebral cortex are unclear. Here, we used human cortical organoids to study the effects of EtOH on neurogenesis and validated our findings in primary human fetal neurons. EtOH exposure produced temporally dependent cellular effects on proliferation, cell cycle, and apoptosis. In addition, we identified EtOH-induced alterations in post-translational histone modifications and chromatin accessibility, leading to impairment of cAMP and calcium signaling, glutamatergic synaptic development, and astrocytic function. Proteomic spatial profiling of cortical organoids showed region-specific, EtOH-induced alterations linked to changes in cytoskeleton, gliogenesis, and impaired synaptogenesis. Finally, multi-electrode array electrophysiology recordings confirmed the deleterious impact of EtOH on neural network formation and activity in cortical organoids, which was validated in primary human fetal tissues. Our findings demonstrate progress in defining the human molecular and cellular phenotypic signatures of prenatal alcohol exposure on functional neurodevelopment, increasing our knowledge for potential therapeutic interventions targeting FASD symptoms.
Collapse
Affiliation(s)
- Jason W Adams
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
- Department of Neurosciences, University of California San Diego, School of Medicine, La Jolla, CA, 92093, USA
- Center for Academic Research and Training in Anthropogeny, University of California San Diego, La Jolla, CA, 92093, USA
| | - Priscilla D Negraes
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
| | - Justin Truong
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
| | - Timothy Tran
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
| | - Ryan A Szeto
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
| | - Bruno S Guerra
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
- Experimental Multiuser Laboratory, Pontifícia Universidade Católica do Paraná, Curitiba, PR, 80215-901, Brazil
| | - Roberto H Herai
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA
- Experimental Multiuser Laboratory, Pontifícia Universidade Católica do Paraná, Curitiba, PR, 80215-901, Brazil
| | - Carmen Teodorof-Diedrich
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, CA, 92093, USA
| | - Stephen A Spector
- Department of Pediatrics, Division of Infectious Diseases, University of California San Diego, La Jolla, CA, 92093, USA
| | - Miguel Del Campo
- Department of Pediatrics, Division of Dysmorphology and Teratology, University of California, La Jolla, CA, 92093, USA
| | - Kenneth L Jones
- Department of Pediatrics, Division of Dysmorphology and Teratology, University of California, La Jolla, CA, 92093, USA
| | - Alysson R Muotri
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA.
- Center for Academic Research and Training in Anthropogeny, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Cleber A Trujillo
- Department of Pediatrics/Rady Children's Hospital, Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
9
|
Breton-Larrivée M, Elder E, Legault LM, Langford-Avelar A, MacFarlane AJ, McGraw S. Mitigating the detrimental developmental impact of early fetal alcohol exposure using a maternal methyl donor-enriched diet. FASEB J 2023; 37:e22829. [PMID: 36856720 DOI: 10.1096/fj.202201564r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/23/2023] [Accepted: 02/03/2023] [Indexed: 03/02/2023]
Abstract
Fetal alcohol exposure at any stage of pregnancy can lead to fetal alcohol spectrum disorder (FASD), a group of life-long conditions characterized by congenital malformations, as well as cognitive, behavioral, and emotional impairments. The teratogenic effects of alcohol have long been publicized; yet fetal alcohol exposure is one of the most common preventable causes of birth defects. Currently, alcohol abstinence during pregnancy is the best and only way to prevent FASD. However, alcohol consumption remains astoundingly prevalent among pregnant women; therefore, additional measures need to be made available to help protect the developing embryo before irreparable damage is done. Maternal nutritional interventions using methyl donors have been investigated as potential preventative measures to mitigate the adverse effects of fetal alcohol exposure. Here, we show that a single acute preimplantation (E2.5; 8-cell stage) fetal alcohol exposure (2 × 2.5 g/kg ethanol with a 2h interval) in mice leads to long-term FASD-like morphological phenotypes (e.g. growth restriction, brain malformations, skeletal delays) in late-gestation embryos (E18.5) and demonstrate that supplementing the maternal diet with a combination of four methyl donor nutrients, folic acid, choline, betaine, and vitamin B12, prior to conception and throughout gestation effectively reduces the incidence and severity of alcohol-induced morphological defects without altering DNA methylation status of imprinting control regions and regulation of associated imprinted genes. This study clearly supports that preimplantation embryos are vulnerable to the teratogenic effects of alcohol, emphasizes the dangers of maternal alcohol consumption during early gestation, and provides a potential proactive maternal nutritional intervention to minimize FASD progression, reinforcing the importance of adequate preconception and prenatal nutrition.
Collapse
Affiliation(s)
- Mélanie Breton-Larrivée
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada
| | - Elizabeth Elder
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada
| | - Lisa-Marie Legault
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada
| | - Alexandra Langford-Avelar
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada
| | - Amanda J MacFarlane
- Agriculture, Food, and Nutrition Evidence Center, Texas A&M University, Texas, Fort Worth, USA.,Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Serge McGraw
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Canada.,Department of Obstetrics and Gynecology, Université de Montréal, Montreal, Canada
| |
Collapse
|
10
|
Auvinen P, Vehviläinen J, Marjonen H, Modhukur V, Sokka J, Wallén E, Rämö K, Ahola L, Salumets A, Otonkoski T, Skottman H, Ollikainen M, Trokovic R, Kahila H, Kaminen-Ahola N. Chromatin modifier developmental pluripotency associated factor 4 (DPPA4) is a candidate gene for alcohol-induced developmental disorders. BMC Med 2022; 20:495. [PMID: 36581877 PMCID: PMC9801659 DOI: 10.1186/s12916-022-02699-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/07/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) affects embryonic development, causing a variable fetal alcohol spectrum disorder (FASD) phenotype with neuronal disorders and birth defects. We hypothesize that early alcohol-induced epigenetic changes disrupt the accurate developmental programming of embryo and consequently cause the complex phenotype of developmental disorders. To explore the etiology of FASD, we collected unique biological samples of 80 severely alcohol-exposed and 100 control newborns at birth. METHODS We performed genome-wide DNA methylation (DNAm) and gene expression analyses of placentas by using microarrays (EPIC, Illumina) and mRNA sequencing, respectively. To test the manifestation of observed PAE-associated DNAm changes in embryonic tissues as well as potential biomarkers for PAE, we examined if the changes can be detected also in white blood cells or buccal epithelial cells of the same newborns by EpiTYPER. To explore the early effects of alcohol on extraembryonic placental tissue, we selected 27 newborns whose mothers had consumed alcohol up to gestational week 7 at maximum to the separate analyses. Furthermore, to explore the effects of early alcohol exposure on embryonic cells, human embryonic stem cells (hESCs) as well as hESCs during differentiation into endodermal, mesodermal, and ectodermal cells were exposed to alcohol in vitro. RESULTS DPPA4, FOXP2, and TACR3 with significantly decreased DNAm were discovered-particularly the regulatory region of DPPA4 in the early alcohol-exposed placentas. When hESCs were exposed to alcohol in vitro, significantly altered regulation of DPPA2, a closely linked heterodimer of DPPA4, was observed. While the regulatory region of DPPA4 was unmethylated in both control and alcohol-exposed hESCs, alcohol-induced decreased DNAm similar to placenta was seen in in vitro differentiated mesodermal and ectodermal cells. Furthermore, common genes with alcohol-associated DNAm changes in placenta and hESCs were linked exclusively to the neurodevelopmental pathways in the enrichment analysis, which emphasizes the value of placental tissue when analyzing the effects of prenatal environment on human development. CONCLUSIONS Our study shows the effects of early alcohol exposure on human embryonic and extraembryonic cells, introduces candidate genes for alcohol-induced developmental disorders, and reveals potential biomarkers for prenatal alcohol exposure.
Collapse
Affiliation(s)
- P Auvinen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - J Vehviläinen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - H Marjonen
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - V Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre on Health Technologies, 50411, Tartu, Estonia
| | - J Sokka
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - E Wallén
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - K Rämö
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - L Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland
| | - A Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre on Health Technologies, 50411, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, S-171 76, Stockholm, Sweden
| | - T Otonkoski
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
- Children's Hospital, Helsinki University Central Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - H Skottman
- Faculty of Medicine and Health Technology, Tampere University, 33520, Tampere, Finland
| | - M Ollikainen
- Institute for Molecular Medicine, Finland, FIMM, HiLIFE, University of Helsinki, 00290, Helsinki, Finland
| | - R Trokovic
- Research Programs Unit, Stem cells and Metabolism and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - H Kahila
- Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, 00290, Helsinki, Finland
| | - N Kaminen-Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, 00290, Helsinki, Finland.
| |
Collapse
|
11
|
Costa TJ, De Oliveira JC, Giachini FR, Lima VV, Tostes RC, Bomfim GF. Programming of Vascular Dysfunction by Maternal Stress: Immune System Implications. Front Physiol 2022; 13:787617. [PMID: 35360231 PMCID: PMC8961444 DOI: 10.3389/fphys.2022.787617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
A growing body of evidence highlights that several insults during pregnancy impact the vascular function and immune response of the male and female offspring. Overactivation of the immune system negatively influences cardiovascular function and contributes to cardiovascular disease. In this review, we propose that modulation of the immune system is a potential link between prenatal stress and offspring vascular dysfunction. Glucocorticoids are key mediators of stress and modulate the inflammatory response. The potential mechanisms whereby prenatal stress negatively impacts vascular function in the offspring, including poor hypothalamic–pituitary–adrenal axis regulation of inflammatory response, activation of Th17 cells, renin–angiotensin–aldosterone system hyperactivation, reactive oxygen species imbalance, generation of neoantigens and TLR4 activation, are discussed. Alterations in the immune system by maternal stress during pregnancy have broad relevance for vascular dysfunction and immune-mediated diseases, such as cardiovascular disease.
Collapse
Affiliation(s)
- Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlio Cezar De Oliveira
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Rita C. Tostes
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
| | - Gisele Facholi Bomfim
- Health Education Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil
- *Correspondence: Gisele Facholi Bomfim,
| |
Collapse
|
12
|
Hong L, Li N, Gasque V, Mehta S, Ye L, Wu Y, Li J, Gewies A, Ruland J, Hirschi KK, Eichmann A, Hendry C, van Dijk D, Mani A. Prdm6 controls heart development by regulating neural crest cell differentiation and migration. JCI Insight 2022; 7:156046. [PMID: 35108221 PMCID: PMC8876496 DOI: 10.1172/jci.insight.156046] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/13/2022] [Indexed: 11/22/2022] Open
Abstract
The molecular mechanisms that drive the acquisition of distinct neural crest cell (NCC) fates is still poorly understood. Here, we identified Prdm6 as an epigenetic modifier that temporally and spatially regulates the expression of NCC specifiers and determines the fate of a subset of migrating cardiac NCCs (CNCCs). Using transcriptomic analysis and genetic and fate mapping approaches in transgenic mice, we showed that disruption of Prdm6 was associated with impaired CNCC differentiation, delamination, and migration and led to patent ductus arteriosus (DA) and ventricular noncompaction. Bulk and single-cell RNA-Seq analyses of the DA and CNCCs identified Prdm6 as a regulator of a network of CNCC specification genes, including Wnt1, Tfap2b, and Sox9. Loss of Prdm6 in CNCCs diminished its expression in the pre-epithelial–mesenchymal transition (pre-EMT) cluster, resulting in the retention of NCCs in the dorsal neural tube. This defect was associated with diminished H4K20 monomethylation and G1-S progression and augmented Wnt1 transcript levels in pre-EMT and neural tube clusters, which we showed was the major driver of the impaired CNCC migration. Altogether, these findings revealed Prdm6 as a key regulator of CNCC differentiation and migration and identified Prdm6 and its regulated network as potential targets for the treatment of congenital heart diseases.
Collapse
Affiliation(s)
- Lingjuan Hong
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | - Na Li
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | - Victor Gasque
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | - Sameet Mehta
- Yale Center for Genome Analysis, Yale University School of Medicine, New Haven, United States of America
| | - Lupeng Ye
- Department of Genetics, Yale University School of Medicine, New Haven, United States of America
| | - Yinyu Wu
- Department of Genetics, Yale University School of Medicine, New Haven, United States of America
| | - Jinyu Li
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | | | | | - Karen K Hirschi
- University of Virginia School of Medicine, Charlottesville, United States of America
| | - Anne Eichmann
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | - Caroline Hendry
- Department of Genetics, Yale University School of Medicine, New Haven, United States of America
| | - David van Dijk
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| | - Arya Mani
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, United States of America
| |
Collapse
|
13
|
Hutchinson D, Spry EA, Mohamad Husin H, Middleton M, Hearps S, Moreno-Betancur M, Elliott EJ, Ryan J, Olsson CA, Patton GC. Longitudinal prediction of periconception alcohol use: a 20-year prospective cohort study across adolescence, young adulthood and pregnancy. Addiction 2022; 117:343-353. [PMID: 34495562 DOI: 10.1111/add.15632] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/17/2021] [Accepted: 06/16/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Alcohol consumption is common in adolescence and young adulthood and may continue into pregnancy, posing serious risk to early fetal development. We examine the frequency of periconception alcohol use (prior to pregnancy awareness) and the extent to which adolescent and young adult alcohol use prospectively predict periconception use. DESIGN A longitudinal, population-based study. SETTING Victoria, Australia. PARTICIPANTS A total of 289 women in trimester three of pregnancy (age 29-35 years; 388 pregnancies). MEASURES The main exposures were binge [≥ 4.0 standard drinks (SDs)/day] and frequent (≥ 3 days/week) drinking in adolescence (mean age = 14.9-17.4 years) and young adulthood (mean age 20.7-29.1 years). Outcomes were frequency (≥ 3 days/week, ≥ monthly, never) and quantity (≥ 4.0 SDs, ≥ 0.5 and < 4.0 SDs, none) of periconception drinking. FINDINGS Alcohol use was common in young adulthood prior to pregnancy (72%) and in the early weeks of pregnancy (76%). The proportions drinking on most days and binge drinking were similar at both points. Reflecting a high degree of continuity in alcohol use behaviours, most women who drank periconceptionally had an earlier history of frequent (77%) and/or binge (85%) drinking throughout the adolescent or young adult years. Young adult binge drinking prospectively predicted periconception drinking quantity [odds ratio (OR) = 3.7, 95% confidence interval (CI) = 1.9-7.4], compared with women with no prior history. Similarly, frequent young adult drinking prospectively predicted frequent periconception drinking (OR = 30.7, 95% CI = 12.3-76.7). CONCLUSIONS Women who engage in risky (i.e. frequent and binge) drinking in their adolescent and young adult years are more likely to report risky drinking in early pregnancy prior to pregnancy recognition than women with no prior history of risky drinking.
Collapse
Affiliation(s)
- Delyse Hutchinson
- Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Deakin University, Geelong, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Elizabeth A Spry
- Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Deakin University, Geelong, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Hanafi Mohamad Husin
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Melissa Middleton
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen Hearps
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Margarita Moreno-Betancur
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Elizabeth J Elliott
- Faculty of Medicine and Health, Discipline of Child and Adolescent Health, The University of Sydney, Sydney, NSW, Australia.,The Sydney Children's Hospital Network, Westmead, Kid's Research, Sydney, NSW, Australia
| | - Joanne Ryan
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia.,Department of Epidemiology, Monash University, Prahran, VIC, Australia
| | - Craig A Olsson
- Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Deakin University, Geelong, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - George C Patton
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Centre for Adolescent Health, Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Association of prenatal alcohol exposure with offspring DNA methylation in mammals: a systematic review of the evidence. Clin Epigenetics 2022; 14:12. [PMID: 35073992 PMCID: PMC8785586 DOI: 10.1186/s13148-022-01231-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
Prenatal alcohol exposure (PAE) is associated with a range of adverse offspring neurodevelopmental outcomes. Several studies suggest that PAE modifies DNA methylation in offspring cells and tissues, providing evidence for a potential mechanistic link to Fetal Alcohol Spectrum Disorder (FASD). We systematically reviewed existing evidence on the extent to which maternal alcohol use during pregnancy is associated with offspring DNA methylation.
Methods
A systematic literature search was conducted across five online databases according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. PubMed, Web of Science, EMBASE, Google Scholar and CINAHL Databases were searched for articles relating to PAE in placental mammals. Data were extracted from each study and the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I) was used to assess the potential for bias in human studies.
Results
Forty-three articles were identified for inclusion. Twenty-six animal studies and 16 human studies measured offspring DNA methylation in various tissues using candidate gene analysis, methylome-wide association studies (MWAS), or total nuclear DNA methylation content. PAE dose and timing varied between studies. Risk of bias was deemed high in nearly all human studies. There was insufficient evidence in human and animal studies to support global disruption of DNA methylation from PAE. Inconclusive evidence was found for hypomethylation at IGF2/H19 regions within somatic tissues. MWAS assessing PAE effects on offspring DNA methylation showed inconsistent evidence. There was some consistency in the relatively small number of MWAS conducted in populations with FASD. Meta-analyses could not be conducted due to significant heterogeneity between studies.
Conclusion
Considering heterogeneity in study design and potential for bias, evidence for an association between PAE and offspring DNA methylation was inconclusive. Some reproducible associations were observed in populations with FASD although the limited number of these studies warrants further research.
Trail Registration: This review is registered with PROSPERO (registration number: CRD42020167686).
Collapse
|
15
|
Lussier AA, Bodnar TS, Weinberg J. Intersection of Epigenetic and Immune Alterations: Implications for Fetal Alcohol Spectrum Disorder and Mental Health. Front Neurosci 2021; 15:788630. [PMID: 34924946 PMCID: PMC8680672 DOI: 10.3389/fnins.2021.788630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Prenatal alcohol exposure can impact virtually all body systems, resulting in a host of structural, neurocognitive, and behavioral abnormalities. Among the adverse impacts associated with prenatal alcohol exposure are alterations in immune function, including an increased incidence of infections and alterations in immune/neuroimmune parameters that last throughout the life-course. Epigenetic patterns are also highly sensitive to prenatal alcohol exposure, with widespread alcohol-related alterations to epigenetic profiles, including changes in DNA methylation, histone modifications, and miRNA expression. Importantly, epigenetic programs are crucial for immune system development, impacting key processes such as immune cell fate, differentiation, and activation. In addition to their role in development, epigenetic mechanisms are emerging as attractive candidates for the biological embedding of environmental factors on immune function and as mediators between early-life exposures and long-term health. Here, following an overview of the impact of prenatal alcohol exposure on immune function and epigenetic patterns, we discuss the potential role for epigenetic mechanisms in reprogramming of immune function and the consequences for health and development. We highlight a range of both clinical and animal studies to provide insights into the array of immune genes impacted by alcohol-related epigenetic reprogramming. Finally, we discuss potential consequences of alcohol-related reprogramming of immune/neuroimmune functions and their effects on the increased susceptibility to mental health disorders. Overall, the collective findings from animal models and clinical studies highlight a compelling relationship between the immune system and epigenetic pathways. These findings have important implications for our understanding of the biological mechanisms underlying the long-term and multisystem effects of prenatal alcohol exposure, laying the groundwork for possible novel interventions and therapeutic strategies to treat individuals prenatally exposed to alcohol.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States.,Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Tamara S Bodnar
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Loke YJ, Muggli E, Saffery R, Ryan J, Lewis S, Elliott EJ, Halliday J, Craig JM. Sex- and tissue-specific effects of binge-level prenatal alcohol consumption on DNA methylation at birth. Epigenomics 2021; 13:1921-1938. [PMID: 34841896 DOI: 10.2217/epi-2021-0285] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Binge-level prenatal alcohol exposure (PAE) causes developmental abnormalities, which may be mediated in part by epigenetic mechanisms. Despite this, few studies have characterised the association of binge PAE with DNA methylation in offspring. Methods: We investigated the association between binge PAE and genome-wide DNA methylation profiles in a sex-specific manner in neonatal buccal and placental samples. Results: We identified no differentially methylated CpGs or differentially methylated regions (DMRs) at false discovery rate <0.05. However, using a sum-of-ranks approach, we identified a DMR in each tissue of female offspring. The DMR identified in buccal samples is located near regions with previously-reported associations to fetal alcohol spectrum disorder (FASD) and binge PAE. Conclusion: Our findings warrant further replication and highlight a potential epigenetic link between binge PAE and FASD.
Collapse
Affiliation(s)
- Yuk Jing Loke
- Molecular Immunity, Murdoch Children's Research Institute, Victoria, 3052, Australia.,Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia
| | - Evelyne Muggli
- Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia.,Victorian Infant Brain Studies, Murdoch Children's Research Institute, Victoria, 3052, Australia.,Reproductive Epidemiology, Murdoch Children's Research Institute, Victoria, 3052, Australia
| | - Richard Saffery
- Molecular Immunity, Murdoch Children's Research Institute, Victoria, 3052, Australia.,Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia
| | - Joanne Ryan
- Molecular Immunity, Murdoch Children's Research Institute, Victoria, 3052, Australia.,Biological Neuropsychiatry & Dementia Unit, School of Public Health, Monash University, Victoria, 3004, Australia
| | - Sharon Lewis
- Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia.,Reproductive Epidemiology, Murdoch Children's Research Institute, Victoria, 3052, Australia
| | - Elizabeth J Elliott
- Specialty of Child & Adolescent Health, Faculty of Medicine & Health, University of Sydney, NSW, 2050, Australia.,The Australian Paediatric Surveillance Unit, Sydney Children's Hospital Network, NSW, 2045, Australia
| | - Jane Halliday
- Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia.,Reproductive Epidemiology, Murdoch Children's Research Institute, Victoria, 3052, Australia
| | - Jeffrey M Craig
- Molecular Immunity, Murdoch Children's Research Institute, Victoria, 3052, Australia.,Department of Paediatrics, University of Melbourne, Victoria, 3010, Australia.,The Institute of Mental & Physical Health & Clinical Translation, Deakin University, Victoria, 3220, Australia
| |
Collapse
|
17
|
Gard P. Ethanol: Toxicity and Dangers in Women of Child-Bearing Age. EURASIAN CHEMICO-TECHNOLOGICAL JOURNAL 2021. [DOI: 10.18321/ectj1105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The World Health Organisation estimates that alcohol abuse by adults accounts for about 5% of global disease burden. Additionally, prenatal alcohol exposure (PAE) causes ‘fetal alcohol spectrum disorder’ (FASD). Depending on severity, FASD is characterised by low birth weight, small head size at birth and growth retardation. There are also facial features of narrow eyes, flat upper lip and midface and impaired fine motor skills, hearing loss, poor hand-eye coordination and cognitive impairment. World-wide, up to 10% of children may be affected by PAE. It is unclear what dose or pattern of drinking results in these damaging effects, but animal models suggest that high, acute doses of ethanol (‘binge drinking’) in early pregnancy can result in the facial changes of FASD, whilst sustained, lower dose intake in later pregnancy produces anxiety and depression-like symptoms and deficits of learning and memory. The mechanisms underlying the deleterious effects of PAE are also unresolved, but evidence exists of long-lasting damage due to oxidative stress, increases in inflammatory mediators and changes to the brain renin-angiotensin system. There is also evidence of epigenetic changes. There is a need to prevent or limit the potential adverse effects of ethanol on the unborn child. It is highly unlikely, however, that all sexually-active women of child-bearing age not using reliable contraception will abstain from alcohol. There is therefore a need to research methods of reducing ethanol toxicity for the unborn child and / or develop therapeutic strategies to reverse the deleterious effects of ethanol on the unborn child.
Collapse
|
18
|
Legault LM, Doiron K, Breton-Larrivée M, Langford-Avelar A, Lemieux A, Caron M, Jerome-Majewska LA, Sinnett D, McGraw S. Pre-implantation alcohol exposure induces lasting sex-specific DNA methylation programming errors in the developing forebrain. Clin Epigenetics 2021; 13:164. [PMID: 34425890 PMCID: PMC8381495 DOI: 10.1186/s13148-021-01151-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background Prenatal alcohol exposure is recognized for altering DNA methylation profiles of brain cells during development, and to be part of the molecular basis underpinning Fetal Alcohol Spectrum Disorder (FASD) etiology. However, we have negligible information on the effects of alcohol exposure during pre-implantation, the early embryonic window marked with dynamic DNA methylation reprogramming, and on how this may rewire the brain developmental program. Results Using a pre-clinical in vivo mouse model, we show that a binge-like alcohol exposure during pre-implantation at the 8-cell stage leads to surge in morphological brain defects and adverse developmental outcomes during fetal life. Genome-wide DNA methylation analyses of fetal forebrains uncovered sex-specific alterations, including partial loss of DNA methylation maintenance at imprinting control regions, and abnormal de novo DNA methylation profiles in various biological pathways (e.g., neural/brain development). Conclusion These findings support that alcohol-induced DNA methylation programming deviations during pre-implantation could contribute to the manifestation of neurodevelopmental phenotypes associated with FASD. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01151-0.
Collapse
Affiliation(s)
- L M Legault
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - K Doiron
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - M Breton-Larrivée
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - A Langford-Avelar
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - A Lemieux
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - M Caron
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - L A Jerome-Majewska
- McGill University Health Centre Glen Site, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada.,Department of Pediatrics, McGill University, 1001 Boulevard Décarie, Montréal, QC, H4A 3J1, Canada
| | - D Sinnett
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Pediatrics, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada
| | - S McGraw
- CHU Sainte-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada. .,Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada. .,Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
19
|
Alberry B, Laufer BI, Chater-Diehl E, Singh SM. Epigenetic Impacts of Early Life Stress in Fetal Alcohol Spectrum Disorders Shape the Neurodevelopmental Continuum. Front Mol Neurosci 2021; 14:671891. [PMID: 34149355 PMCID: PMC8209299 DOI: 10.3389/fnmol.2021.671891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
Neurodevelopment in humans is a long, elaborate, and highly coordinated process involving three trimesters of prenatal development followed by decades of postnatal development and maturation. Throughout this period, the brain is highly sensitive and responsive to the external environment, which may provide a range of inputs leading to positive or negative outcomes. Fetal alcohol spectrum disorders (FASD) result from prenatal alcohol exposure (PAE). Although the molecular mechanisms of FASD are not fully characterized, they involve alterations to the regulation of gene expression via epigenetic marks. As in the prenatal stages, the postnatal period of neurodevelopment is also sensitive to environmental inputs. Often this sensitivity is reflected in children facing adverse conditions, such as maternal separation. This exposure to early life stress (ELS) is implicated in the manifestation of various behavioral abnormalities. Most FASD research has focused exclusively on the effect of prenatal ethanol exposure in isolation. Here, we review the research into the effect of prenatal ethanol exposure and ELS, with a focus on the continuum of epigenomic and transcriptomic alterations. Interestingly, a select few experiments have assessed the cumulative effect of prenatal alcohol and postnatal maternal separation stress. Regulatory regions of different sets of genes are affected by both treatments independently, and a unique set of genes are affected by the combination of treatments. Notably, epigenetic and gene expression changes converge at the clustered protocadherin locus and oxidative stress pathway. Functional studies using epigenetic editing may elucidate individual contributions of regulatory regions for hub genes and further profiling efforts may lead to the development of non-invasive methods to identify children at risk. Taken together, the results favor the potential to improve neurodevelopmental outcomes by epigenetic management of children born with FASD using favorable postnatal conditions with or without therapeutic interventions.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Biology, Faculty of Science, The University of Western Ontario, London, ON, Canada
| | - Benjamin I Laufer
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States.,Genome Center, University of California, Davis, Davis, CA, United States.,MIND Institute, University of California, Davis, Davis, CA, United States
| | - Eric Chater-Diehl
- Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shiva M Singh
- Department of Biology, Faculty of Science, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
20
|
Jia Z, Wu Q. Clustered Protocadherins Emerge as Novel Susceptibility Loci for Mental Disorders. Front Neurosci 2020; 14:587819. [PMID: 33262685 PMCID: PMC7688460 DOI: 10.3389/fnins.2020.587819] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/26/2020] [Indexed: 12/24/2022] Open
Abstract
The clustered protocadherins (cPcdhs) are a subfamily of type I single-pass transmembrane cell adhesion molecules predominantly expressed in the brain. Their stochastic and combinatorial expression patterns encode highly diverse neural identity codes which are central for neuronal self-avoidance and non-self discrimination in brain circuit formation. In this review, we first briefly outline mechanisms for generating a tremendous diversity of cPcdh cell-surface assemblies. We then summarize the biological functions of cPcdhs in a wide variety of neurodevelopmental processes, such as neuronal migration and survival, dendritic arborization and self-avoidance, axonal tiling and even spacing, and synaptogenesis. We focus on genetic, epigenetic, and 3D genomic dysregulations of cPcdhs that are associated with various neuropsychiatric and neurodevelopmental diseases. A deeper understanding of regulatory mechanisms and physiological functions of cPcdhs should provide significant insights into the pathogenesis of mental disorders and facilitate development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | - Qiang Wu
- Center for Comparative Biomedicine, MOE Key Laboratory of Systems Biomedicine, State Key Laboratory of Oncogenes and Related Genes, School of Life Sciences and Biotechnology, Institute of Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Rossnerova A, Izzotti A, Pulliero A, Bast A, Rattan SIS, Rossner P. The Molecular Mechanisms of Adaptive Response Related to Environmental Stress. Int J Mol Sci 2020; 21:ijms21197053. [PMID: 32992730 PMCID: PMC7582272 DOI: 10.3390/ijms21197053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The exposure of living organisms to environmental stress triggers defensive responses resulting in the activation of protective processes. Whenever the exposure occurs at low doses, defensive effects overwhelm the adverse effects of the exposure; this adaptive situation is referred to as “hormesis”. Environmental, physical, and nutritional hormetins lead to the stimulation and strengthening of the maintenance and repair systems in cells and tissues. Exercise, heat, and irradiation are examples of physical hormetins, which activate heat shock-, DNA repair-, and anti-oxidative-stress responses. The health promoting effect of many bio-actives in fruits and vegetables can be seen as the effect of mildly toxic compounds triggering this adaptive stimulus. Numerous studies indicate that living organisms possess the ability to adapt to adverse environmental conditions, as exemplified by the fact that DNA damage and gene expression profiling in populations living in the environment with high levels of air pollution do not correspond to the concentrations of pollutants. The molecular mechanisms of the hormetic response include modulation of (a) transcription factor Nrf2 activating the synthesis of glutathione and the subsequent protection of the cell; (b) DNA methylation; and (c) microRNA. These findings provide evidence that hormesis is a toxicological event, occurring at low exposure doses to environmental stressors, having the benefit for the maintenance of a healthy status.
Collapse
Affiliation(s)
- Andrea Rossnerova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine, 14220 Prague, Czech Republic;
| | - Alberto Izzotti
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alessandra Pulliero
- Department of Health Science, University of Genoa, 16132 Genoa, Italy
- Correspondence:
| | - Aalt Bast
- Department of Pharmacology and Toxicology, Maastricht University, 6200 MD Maastricht, The Netherlands;
- Campus Venlo, Maastricht University, 5900 AA Venlo, The Netherlands
| | - S. I. S. Rattan
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark;
| | - Pavel Rossner
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine, 14220 Prague, Czech Republic;
| |
Collapse
|
22
|
Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci Biobehav Rev 2020; 118:654-668. [PMID: 32976915 DOI: 10.1016/j.neubiorev.2020.08.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/18/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Ethanol exposure during gestation is an early life stressor that profoundly dysregulates structure and functions of the embryonal nervous system, altering the cognitive and behavioral development. Such dysregulation is also achieved by epigenetic mechanisms, which, altering the chromatin structure, redraw the entire pattern of gene expression. In parallel, an oxidative stress response at the cellular level and a global upregulation of neuroendocrine stress response, regulated by the HPA axis, exist and persist in adulthood. This neurobehavioral framework matches those observed in other psychiatric diseases such as mood diseases, depression, autism; those early life stressing events, although probably triggered by specific and different epigenetic mechanisms, give rise to largely overlapping neurobehavioral phenotypes. An early diagnosis of prenatal alcohol exposure, using reliable markers of ethanol intake, together with a deeper understanding of the pathogenic mechanisms, some of them reversible by their nature, can offer a temporal "window" of intervention. Supplementing a mother's diet with protective and antioxidant substances in addition to supportive psychological therapies can protect newborns from being affected.
Collapse
|
23
|
Noor S, Sanchez JJ, Sun MS, Pervin Z, Sanchez JE, Havard MA, Epler LT, Nysus MV, Norenberg JP, Wagner CR, Davies S, Wagner JL, Savage DD, Jantzie LL, Mellios N, Milligan ED. The LFA-1 antagonist BIRT377 reverses neuropathic pain in prenatal alcohol-exposed female rats via actions on peripheral and central neuroimmune function in discrete pain-relevant tissue regions. Brain Behav Immun 2020; 87:339-358. [PMID: 31918004 PMCID: PMC7316595 DOI: 10.1016/j.bbi.2020.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/20/2019] [Accepted: 01/05/2020] [Indexed: 12/13/2022] Open
Abstract
Previous reports show that moderate prenatal alcohol exposure (PAE) poses a risk factor for developing neuropathic pain following adult-onset peripheral nerve injury in male rats. Recently, evidence suggests that immune-related mechanisms underlying neuropathic pain in females are different compared to males despite the fact that both sexes develop neuropathy of similar magnitude and duration following chronic constriction injury (CCI) of the sciatic nerve. Data suggest that the actions of peripheral T cells play a greater role in mediating neuropathy in females. The goal of the current study is to identify specificity of immune cell and cytokine changes between PAE and non-PAE neuropathic females by utilizing a well-characterized rodent model of sciatic nerve damage, in an effort to unmask unique signatures of immune-related factors underlying the risk of neuropathy from PAE. Cytokines typically associated with myeloid cell actions such as interleukin (IL)-1β, tumor necrosis factor (TNF), IL-6, IL-4 and IL-10 as well as the neutrophil chemoattractant CXCL1, are examined. In addition, transcription factors and cytokines associated with various differentiated T cell subtypes are examined (anti-inflammatory FOXP3, proinflammatory IL-17A, IL-21, ROR-γt, interferon (IFN)-γ and T-bet). Lymphocyte function associated antigen 1 (LFA-1) is an adhesion molecule expressed on peripheral immune cells including T cells, and regulates T cell activation and extravasation into inflamed tissue regions. A potential therapeutic approach was explored with the goal of controlling proinflammatory responses in neuroanatomical regions critical for CCI-induced allodynia by blocking LFA-1 actions using BIRT377. The data show profound development of hindpaw allodynia in adult non-PAE control females following standard CCI, but not following minor CCI, while minor CCI generated allodynia in PAE females. The data also show substantial increases in T cell-associated proinflammatory cytokine mRNA and proteins, along with evidence of augmented myeloid/glial activation (mRNA) and induction of myeloid/glial-related proinflammatory cytokines, CCL2, IL-1β and TNF in discrete regions along the pain pathway (damaged sciatic nerve, dorsal root ganglia; DRG, and spinal cord). Interestingly, the characteristic anti-inflammatory IL-10 protein response to nerve damage is blunted in neuropathic PAE females. Moreover, T cell profiles are predominantly proinflammatory in neuropathic Sac and PAE females, augmented levels of Th17-specific proinflammatory cytokines IL-17A and IL-21, as well as the Th1-specific factor, T-bet, are observed. Similarly, the expression of RORγt, a critical transcription factor for Th17 cells, is detected in the spinal cord of neuropathic females. Blocking peripheral LFA-1 actions with intravenous (i.v.) BIRT377 reverses allodynia in Sac and PAE rats, dampens myeloid (IL-1β, TNF, CXCL1)- and T cell-associated proinflammatory factors (IL-17A and RORγt) and spinal glial activation. Moreover, i.v. BIRT377 treatment reverses the blunted IL-10 response to CCI observed only in neuropathic PAE rats and elevates FOXP3 in pain-reversed Sac rats. Unexpectedly, intrathecal BIRT377 treatment is unable to alter allodynia in either Sac or PAE neuropathic females. Together, these data provide evidence that: 1) fully differentiated proinflammatory Th17 cells recruited at the sciatic nerve, DRGs and lumbar spinal cord may interact with the local environment to shape the immune responses underlying neuropathy in female rats, and, 2) PAE primes peripheral and spinal immune responses in adult females. PAE is a risk factor in females for developing peripheral neuropathy after minor nerve injury.
Collapse
Affiliation(s)
- Shahani Noor
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA.
| | - Joshua J. Sanchez
- Department of Neurosciences, School of Medicine, University of California, San Diego, CA, USA
| | - Melody S. Sun
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| | - Zinia Pervin
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA.
| | - Jacob E. Sanchez
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| | - Mara A. Havard
- Department of Anesthesiology and Critical Care, University of New Mexico, Albuquerque, NM, 8713,USA
| | - Lauren T. Epler
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico, Albuquerque, NM 87131-001, USA
| | - Monique V. Nysus
- Department of Radiopharmaceutical Sciences, College of Pharmacy, New Mexico Center for Isotopes in Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| | - Jeffrey P. Norenberg
- Department of Radiopharmaceutical Sciences, College of Pharmacy, New Mexico Center for Isotopes in Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| | - Carston R. Wagner
- Department of Medicinal Chemistry, University of Minnesota, College of Pharmacy, MN 55455, USA
| | - Suzy Davies
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA.
| | - Jennifer L Wagner
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA.
| | - Daniel D. Savage
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| | - Lauren L. Jantzie
- Department of Pediatrics and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | - Nikolaos Mellios
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA.
| | - Erin. D. Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM, 87131,USA
| |
Collapse
|
24
|
Pentecost M, Meloni M. "It's Never Too Early": Preconception Care and Postgenomic Models of Life. FRONTIERS IN SOCIOLOGY 2020; 5:21. [PMID: 33869430 PMCID: PMC8022598 DOI: 10.3389/fsoc.2020.00021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/11/2020] [Indexed: 05/16/2023]
Abstract
In this article, we are concerned with the expanded public health interest in the "preconception period" as a window of opportunity for intervention to improve long-term population health outcomes. While definitions of the "preconception period" remain vague, new classifications and categories of life are becoming formalized as biomedicine begins to conduct research on, and suggest intervention in, this undefined and potentially unlimited time before conception. In particular, we focus on the burgeoning epidemiological interest in epigenetics and Developmental Origins of Health and Disease (DOHaD) research as simultaneously a theoretical spyglass into postgenomic biology and a catalyst toward a public health focus on preconception care. We historicize the notion that there are long-term implications of parental behaviors before conception, illustrating how, as Han and Das have noted, "newness comes to be embedded in older forms even as it transforms them" (Han and Das, 2015, p. 2). We then consider how DOHaD frameworks justify a number of fragmented claims about preconception by making novel evidentiary assertions. Engaging with the philosophy of Georges Canguilhem, we examine the relationship between reproductive risk and revised understandings of biological permeability, and discuss some of the epistemic and political implications of emerging claims in postgenomics.
Collapse
Affiliation(s)
- Michelle Pentecost
- Department of Global Health and Social Medicine, King's College London, London, United Kingdom
- Department of Anthropology, University of Cape Town, Cape Town, South Africa
| | - Maurizio Meloni
- Alfred Deakin Institute for Citizenship and Globalization, Deakin University, Melbourne, VIC, Australia
| |
Collapse
|
25
|
Prenatal S-Adenosine Methionine (SAMe) Induces Changes in Gene Expression in the Brain of Newborn Mice That Are Prevented by Co-Administration of Valproic Acid (VPA). Int J Mol Sci 2020; 21:ijms21082834. [PMID: 32325788 PMCID: PMC7215397 DOI: 10.3390/ijms21082834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
In previous studies, we produced changes in gene expression in the brain of mice by early postnatal administration of valproic acid (VPA), with distinct differences between genders. The addition of S-adenosine methionine (SAMe) normalized the expression of most genes in both genders, while SAMe alone induced no changes. We treated pregnant dams with a single injection of VPA on day 12.5 of gestation, or with SAMe during gestational days 12–14, or by a combination of VPA and SAMe. In the frontal half of the brain, we studied the expression of 770 genes of the pathways involved in neurophysiology and neuropathology using the NanoString nCounter method. SAMe, but not VPA, induced statistically significant changes in the expression of many genes, with differences between genders. The expression of 112 genes was changed in both sexes, and another 170 genes were changed only in females and 31 only in males. About 30% of the genes were changed by more than 50%. One of the most important pathways changed by SAMe in both sexes was the VEGF (vascular endothelial growth factor) pathway. Pretreatment with VPA prevented almost all the changes in gene expression induced by SAMe. We conclude that large doses of SAMe, if administered prenatally, may induce significant epigenetic changes in the offspring. Hence, SAMe and possibly other methyl donors may be epigenetic teratogens.
Collapse
|
26
|
Prenatal Drinking in the Northern Plains: Differences Between American Indian and Caucasian Mothers. Am J Prev Med 2020; 58:e113-e121. [PMID: 32061456 PMCID: PMC7089814 DOI: 10.1016/j.amepre.2019.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Prenatal alcohol exposure is associated with adverse pregnancy outcomes such as fetal alcohol spectrum disorders. The study characterizes the pattern and risk factors of alcohol use during pregnancy for American Indian and Caucasian mothers in the Northern Plains. METHODS A general population of pregnant women was recruited from 5 sites, including 2,753 Caucasians and 2,124 American Indians (2006-2017). Alcohol consumption was based on self-report using a modified Timeline Followback interview, administered 3-4 times during pregnancy and 1 month postpartum. Risk for prenatal drinking was calculated using logistic regression models after controlling for demographics, reproductive history, prenatal care, mental health, and SES. The analysis was conducted in 2019. RESULTS More Caucasian mothers consumed alcoholic beverages during pregnancy than American Indians (63% vs 52%), whereas more American Indian mothers were binge drinkers than Caucasians (41% vs 28%). American Indian mothers had a lower risk of drinking in the second and third trimesters and postpartum, but a higher risk of binge drinking in the first trimester compared with Caucasians. Frequent relocation increased the risk of prenatal alcohol use among American Indian mothers, whereas age, marriage, income, parity, and fertility treatment affected the risk of prenatal drinking among Caucasian mothers. CONCLUSIONS Alcohol use was more prevalent among Caucasian mothers. Among those who consumed alcohol during pregnancy, American Indian mothers consumed larger quantities. Change of residence was found to be the sole risk factor for prenatal drinking among American Indian mothers, whereas different and multiple risk factors were found for Caucasian mothers.
Collapse
|
27
|
Alberry BL, Singh SM. Hippocampal DNA Methylation in a Mouse Model of Fetal Alcohol Spectrum Disorder That Includes Maternal Separation Stress Only Partially Explains Changes in Gene Expression. Front Genet 2020; 11:70. [PMID: 32174962 PMCID: PMC7056727 DOI: 10.3389/fgene.2020.00070] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is characterized by developmental and behavioral deficits caused by maternal drinking during pregnancy. Children born with FASD often face additional stresses, including maternal separation, that add yet additional deficits. The mechanism associated with this interaction is not known. We have used a mouse model for prenatal ethanol exposure and maternal separation to demonstrate that the combination of the two treatments results in more than additive deficits. Furthermore, the behavioral deficits are associated with changes in hippocampal gene expression that persist into adulthood. What initiates and maintains these changes remains to be established and forms the focus of this report. Specifically, MeDIP-Seq was used to assess if changes in promoter DNA methylation are affected by exposure to prenatal ethanol and maternal separation including its relationship to gene expression. The novel results show that different sets of genes implicated by promoter DNA methylation are affected by both treatments independently, and a relatively unique set of genes are affected by the combination of the two treatments. Prenatal ethanol exposure leads to altered promoter DNA methylation at genes important for transcriptional regulation. Maternal separation leads to changes at genes important for histone methylation and immune response, and the combination of two treatments results in DNA methylation changes at genes important for neuronal migration and immune response. Our dual results from the same hippocampal samples suggest there is minimal complementarity between changes in promoter DNA methylation and gene expression, although genes involved tend to be critical for brain development and function. While remaining to be validated, such results argue that mechanisms beyond promoter DNA methylation must be involved in lasting gene expression alterations leading to behavioral deficits implicated in FASD. They may facilitate early and reliable diagnosis, as well as novel strategies for the amelioration of FASD-related deficits.
Collapse
Affiliation(s)
| | - Shiva M. Singh
- Department of Biology, Western University, London, ON, Canada
| |
Collapse
|
28
|
Sebastiani G, Andreu-Fernández V, Herranz Barbero A, Aldecoa-Bilbao V, Miracle X, Meler Barrabes E, Balada Ibañez A, Astals-Vizcaino M, Ferrero-Martínez S, Gómez-Roig MD, García-Algar O. Eating Disorders During Gestation: Implications for Mother's Health, Fetal Outcomes, and Epigenetic Changes. Front Pediatr 2020; 8:587. [PMID: 33042925 PMCID: PMC7527592 DOI: 10.3389/fped.2020.00587] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction: Eating disorders (EDs) have increased globally in women of childbearing age, related to the concern for body shape promoted in industrialized countries. Pregnancy may exacerbate a previous ED or conversely may be a chance for improving eating patterns due to the mother's concern for the unborn baby. EDs may impact pregnancy evolution and increase the risk of adverse outcomes such as miscarriage, preterm delivery, poor fetal growth, or malformations, but the knowledge on this topic is limited. Methods: We performed a systematic review of studies on humans in order to clarify the mechanisms underpinning the adverse pregnancy outcomes in patients with EDs. Results: Although unfavorable fetal development could be multifactorial, maternal malnutrition, altered hormonal pathways, low pre-pregnancy body mass index, and poor gestational weight gain, combined with maternal psychopathology and stress, may impair the evolution of pregnancy. Environmental factors such as malnutrition or substance of abuse may also induce epigenetic changes in the fetal epigenome, which mark lifelong health concerns in offspring. Conclusions: The precocious detection of dysfunctional eating behaviors in the pre-pregnancy period and an early multidisciplinary approach comprised of nutritional support, psychotherapeutic techniques, and the use of psychotropics if necessary, would prevent lifelong morbidity for both mother and fetus. Further prospective studies with large sample sizes are needed in order to design a structured intervention during every stage of pregnancy and in the postpartum period.
Collapse
Affiliation(s)
- Giorgia Sebastiani
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Valencian International University (VIU), Valencia, Spain
| | - Ana Herranz Barbero
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Victoria Aldecoa-Bilbao
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Xavier Miracle
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Eva Meler Barrabes
- Fetal i+D Fetal Medicine Research Center, BCNatal-Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Arantxa Balada Ibañez
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Marta Astals-Vizcaino
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Silvia Ferrero-Martínez
- Hospital Sant Joan de Déu, Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - María Dolores Gómez-Roig
- Hospital Sant Joan de Déu, Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain
| | - Oscar García-Algar
- Neonatal Unit, Hospital Clinic-Maternitat, Institut Clinic de Ginecologia, Obstetricia i Neonatologia (ICGON), Barcelona Center for Maternal Fetal and Neonatal Medicine (BCNatal), Barcelona, Spain.,Grup de Recerca Infancia i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
29
|
Abstract
Emerging evidence suggests that epigenetic regulation is dependent on metabolic state, and implicates specific metabolic factors in neural functions that drive behaviour1. In neurons, acetylation of histones relies on the metabolite acetyl-CoA, which is produced from acetate by chromatin-bound acetyl-CoA synthetase 2 (ACSS2)2. Notably, the breakdown of alcohol in the liver leads to a rapid increase in levels of blood acetate3, and alcohol is therefore a major source of acetate in the body. Histone acetylation in neurons may thus be under the influence of acetate that is derived from alcohol4, with potential effects on alcohol-induced gene expression in the brain, and on behaviour5. Here, using in vivo stable-isotope labelling in mice, we show that the metabolism of alcohol contributes to rapid acetylation of histones in the brain, and that this occurs in part through the direct deposition of acetyl groups that are derived from alcohol onto histones in an ACSS2-dependent manner. A similar direct deposition was observed when mice were injected with heavy-labelled acetate in vivo. In a pregnant mouse, exposure to labelled alcohol resulted in the incorporation of labelled acetyl groups into gestating fetal brains. In isolated primary hippocampal neurons ex vivo, extracellular acetate induced transcriptional programs related to learning and memory, which were sensitive to ACSS2 inhibition. We show that alcohol-related associative learning requires ACSS2 in vivo. These findings suggest that there is a direct link between alcohol metabolism and gene regulation, through the ACSS2-dependent acetylation of histones in the brain.
Collapse
|
30
|
Xu K, Montalvo‐Ortiz JL, Zhang X, Southwick SM, Krystal JH, Pietrzak RH, Gelernter J. Epigenome‐Wide
DNA
Methylation Association Analysis Identified Novel Loci in Peripheral Cells for Alcohol Consumption Among European American Male Veterans. Alcohol Clin Exp Res 2019; 43:2111-2121. [PMID: 31386212 DOI: 10.1111/acer.14168] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/25/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Ke Xu
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
| | - Janitza L. Montalvo‐Ortiz
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
| | - Xinyu Zhang
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
| | - Steven M. Southwick
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
- Clinical Neurosciences Division U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder West Haven Connecticut
| | - John H. Krystal
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
- Clinical Neurosciences Division U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder West Haven Connecticut
| | - Robert H. Pietrzak
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
- Clinical Neurosciences Division U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder West Haven Connecticut
| | - Joel Gelernter
- Department of Psychiatry Yale School of Medicine New Haven Connecticut
- VA Connecticut Healthcare System West Haven Connecticut
- Clinical Neurosciences Division U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder West Haven Connecticut
| |
Collapse
|
31
|
Jarmasz JS, Stirton H, Basalah D, Davie JR, Clarren SK, Astley SJ, Del Bigio MR. Global DNA Methylation and Histone Posttranslational Modifications in Human and Nonhuman Primate Brain in Association with Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2019; 43:1145-1162. [PMID: 31074890 PMCID: PMC6593679 DOI: 10.1111/acer.14052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/26/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022]
Abstract
Background Based upon experimental animal studies, the neurodevelopmental abnormalities associated with prenatal alcohol exposure (PNAE)/fetal alcohol spectrum disorder (FASD) have been attributed, at least in part, to epigenetic modifications. However, there are no direct analyses of human brain tissue. Methods Immunohistochemical detection of global epigenetic markers was performed on temporal lobe samples of autopsied fetuses and infants with documented PNAE. They were compared to age‐, sex‐, and postmortem delay‐matched control cases (18 pairs; 20 to 70.5 weeks postconception). Temporal lobe tissue from a macaque monkey model of PNAE was also studied (5.7 to 6 months of age). We used antibodies targeting 4 DNA cytosine, 4 histone methylation, and 6 histone acetylation modifications and assigned scores based upon the semiquantitatively graded intensity and proportion of positively labeled nuclei in the ventricular and subventricular zones, ependyma, temporal cortex, temporal white matter, dentate gyrus (DG), and CA1 pyramidal layer. Results Temporal changes were identified for almost all marks according to the state of maturation in the human brain. In the DG (and 3 other brain regions), a statistically significant increase in H3K9ac was associated with PNAE. Statistically significant decreases were seen among 5mC, H3K4me3, H3K9ac, H3K27ac, H4K12ac, and H4K16ac in select regions. In the macaques, H3K36me3 decreased in the DG, and the ependyma showed decreases in 5fC and H3K36me3. Conclusions In human brain, global intranuclear epigenetic modifications are brain region and maturation state‐specific. These exploratory results support the general hypothesis that PNAE is associated with a global decrease in DNA methylation, a global decrease in histone methylation, and a global increase in histone acetylation. Although the human and monkey subjects are not directly comparable in terms of brain maturation, considering the rapid temporal changes in global epigenetic modifications during brain development, interspecies comparisons may be extremely difficult.
Collapse
Affiliation(s)
- Jessica S Jarmasz
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hannah Stirton
- Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Duaa Basalah
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James R Davie
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sterling K Clarren
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Pediatrics, University of British Columbia Faculty of Medicine, Vancouver, British Columbia
| | - Susan J Astley
- Departments of Epidemiology/Pediatrics, University of Washington, Seattle, Washington
| | - Marc R Del Bigio
- Department of Pathology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
32
|
Cobben JM, Krzyzewska IM, Venema A, Mul AN, Polstra A, Postma AV, Smigiel R, Pesz K, Niklinski J, Chomczyk MA, Henneman P, Mannens MMAM. DNA methylation abundantly associates with fetal alcohol spectrum disorder and its subphenotypes. Epigenomics 2019; 11:767-785. [DOI: 10.2217/epi-2018-0221] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Fetal alcohol spectrum disorder (FASD) involves prenatal growth delay, impaired facial and CNS development and causes severe clinical, social-economic burdens. Here, we aim to detect DNA-methylation aberrations associated with FASD and potential FASD diagnostic and prognostic biomarkers. Patients & methods: The FASD diagnosis was established according to golden-standard protocols in a discovery and independent replication cohort. Genome-wide differential methylation association and replication analyses were performed. Results: We identified several loci that were robustly associated with FASD or one of its sub phenotypes. Our findings were evaluated using previously reported genome-wide surveys. Conclusion: We have detected robust FASD associated differentially methylated positions and differentially methylated regions for FASD in general and for FASD subphenotypes, in other words on growth delay, impaired facial and CNS development.
Collapse
Affiliation(s)
- Jan Maarten Cobben
- Department of Pediatrics, Amsterdam University Medical Centers, Location AMC, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Izabela M Krzyzewska
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Andrea Venema
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Adri N Mul
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Abeltje Polstra
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Alex V Postma
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Robert Smigiel
- Department of Pediatrics & Rare Disorders, Medical University of Wroclaw, Poland
| | - Karolina Pesz
- Department of Genetics, Medical University of Wroclaw, Poland
| | - Jacek Niklinski
- Department of Molecular Biology, Medical University of Bialystok, Poland
| | - Monika A Chomczyk
- Department of Molecular Biology, Medical University of Bialystok, Poland
| | - Peter Henneman
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Marcel MAM Mannens
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Bedi Y, Golding MC. Context is King — Questioning the causal role of DNA methylation in environmentally induced changes in gene expression. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Ma B, Allard C, Bouchard L, Perron P, Mittleman MA, Hivert MF, Liang L. Locus-specific DNA methylation prediction in cord blood and placenta. Epigenetics 2019; 14:405-420. [PMID: 30885044 DOI: 10.1080/15592294.2019.1588685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
DNA methylation is known to be responsive to prenatal exposures, which may be a part of the mechanism linking early developmental exposures to future chronic diseases. Many studies use blood to measure DNA methylation, yet we know that DNA methylation is tissue specific. Placenta is central to fetal growth and development, but it is rarely feasible to collect this tissue in large epidemiological studies; on the other hand, cord blood samples are more accessible. In this study, based on paired samples of both placenta and cord blood tissues from 169 individuals, we investigated the methylation concordance between placenta and cord blood. We then employed a machine-learning-based model to predict locus-specific DNA methylation levels in placenta using DNA methylation levels in cord blood. We found that methylation correlation between placenta and cord blood is lower than other tissue pairs, consistent with existing observations that placenta methylation has a distinct pattern. Nonetheless, there are still a number of CpG sites showing robust association between the two tissues. We built prediction models for placenta methylation based on cord blood data and documented a subset of 1,012 CpG sites with high correlation between measured and predicted placenta methylation levels. The resulting list of CpG sites and prediction models could help to reveal the loci where internal or external influences may affect DNA methylation in both placenta and cord blood, and provide a reference data to predict the effects on placenta in future study even when the tissue is not available in an epidemiological study.
Collapse
Affiliation(s)
- Baoshan Ma
- a College of Information Science and Technology , Dalian Maritime University , Dalian , Liaoning Province , China
| | - Catherine Allard
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke , Quebec , Canada
| | - Luigi Bouchard
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke , Quebec , Canada.,c Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , Quebec , Canada.,d ECOGENE-21 Biocluster , CSSS de Chicoutimi , Chicoutimi , Quebec , Canada
| | - Patrice Perron
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke , Quebec , Canada.,e Department of Medicine, Faculty of Medicine and Life Sciences , Université de Sherbrooke , Sherbrooke , Quebec , Canada
| | - Murray A Mittleman
- f Department of Epidemiology , Harvard T.H. Chan School of Public Health , Boston , MA , USA.,g Cardiovascular Epidemiology Research Unit , Beth Israel Deaconess Medical Center , Boston , MA , USA
| | - Marie-France Hivert
- b Centre de Recherche du Center Hospitalier Universitaire de Sherbrooke , Sherbrooke , Quebec , Canada.,e Department of Medicine, Faculty of Medicine and Life Sciences , Université de Sherbrooke , Sherbrooke , Quebec , Canada.,h Department of Population Medicine , Harvard Pilgrim Health Care Institute, Harvard Medical School , Boston , MA , USA.,i Diabetes Unit , Massachusetts General Hospital , Boston , MA , USA
| | - Liming Liang
- f Department of Epidemiology , Harvard T.H. Chan School of Public Health , Boston , MA , USA.,j Department of Biostatistics , Harvard T.H. Chan School of Public Health , Boston , MA , USA
| |
Collapse
|
35
|
Lussier AA, Bodnar TS, Mingay M, Morin AM, Hirst M, Kobor MS, Weinberg J. Prenatal Alcohol Exposure: Profiling Developmental DNA Methylation Patterns in Central and Peripheral Tissues. Front Genet 2018; 9:610. [PMID: 30568673 PMCID: PMC6290329 DOI: 10.3389/fgene.2018.00610] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022] Open
Abstract
Background: Prenatal alcohol exposure (PAE) can alter the development of neurobiological systems, leading to lasting neuroendocrine, neuroimmune, and neurobehavioral deficits. Although the etiology of this reprogramming remains unknown, emerging evidence suggests DNA methylation as a potential mediator and biomarker for the effects of PAE due to its responsiveness to environmental cues and relative stability over time. Here, we utilized a rat model of PAE to examine the DNA methylation profiles of rat hypothalami and leukocytes at four time points during early development to assess the genome-wide impact of PAE on the epigenome and identify potential biomarkers of PAE. Our model of PAE resulted in blood alcohol levels of ~80-150 mg/dl throughout the equivalent of the first two trimesters of human pregnancy. Hypothalami were analyzed on postnatal days (P) 1, 8, 15, 22 and leukocytes at P22 to compare central and peripheral markers. Genome-wide DNA methylation analysis was performed by methylated DNA immunoprecipitation followed by next-generation sequencing. Results: PAE resulted in lasting changes to DNA methylation profiles across all four ages, with 118 differentially methylated regions (DMRs) displaying persistent alterations across the developmental period at a false-discovery rate (FDR) < 0.05. In addition, 299 DMRs showed the same direction of change in the hypothalamus and leukocytes of P22 pups at an FDR < 0.05, with some genes overlapping with the developmental profile findings. The majority of these DMRs were located in intergenic regions, which contained several computationally-predicted transcription factor binding sites. Differentially methylated genes were generally involved in immune function, epigenetic remodeling, metabolism, and hormonal signaling, as determined by gene ontology analyses. Conclusions: Persistent DNA methylation changes in the hypothalamus may be associated with the long-term physiological and neurobehavioral alterations in observed in PAE. Furthermore, correlations between epigenetic alterations in peripheral tissues and those in the brain will provide a foundation for the development of biomarkers of fetal alcohol spectrum disorder (FASD). Finally, findings from studies of PAE provide important insight into the etiology of neurodevelopmental and mental health disorders, as they share numerous phenotypes and comorbidities.
Collapse
Affiliation(s)
- Alexandre A Lussier
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.,Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Tamara S Bodnar
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Matthew Mingay
- Department of Microbiology and Immunology, Michael Smith Laboratories Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Alexandre M Morin
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology, Michael Smith Laboratories Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada.,Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency Research Centre, BC Cancer Agency, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Human Early Learning Partnership, University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Viljoen D, Louw JG, Lombard C, Olivier L. Comparing diagnostic outcomes of children with fetal alcohol syndrome in South Africa with diagnostic outcomes when using the updated Institute of Medicine diagnostic guidelines. Birth Defects Res 2018; 110:1335-1342. [PMID: 30347134 DOI: 10.1002/bdr2.1399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/16/2018] [Accepted: 08/22/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION During fetal alcohol spectrum disorder (FASD) prevalence studies in South Africa, cases of fetal alcohol syndrome (FAS) were identified that presented differently from the 2016 Hoyme et al. modified Institute of Medicine (IOM) criteria. We compared diagnostic outcomes of children diagnosed with FAS using a combination of the 2005 Hoyme et al. criteria and the "gestalt method" in South Africa to the diagnosis they would have received using the latest Hoyme et al. criteria. The frequency with which dysmorphic features presented was compared to the frequency with which they were reported in the revised criteria which drew on a larger sample. METHODS Data were gathered from four South African FASD prevalence studies. Dysmorphology data, anthropometric data, and final diagnosis for participants (N = 917) were extracted. RESULTS Of the 390 participants with diagnoses of "full FAS," 175 would not have received a "full FAS" diagnosis using the 2016 criteria. Of these, 21 would have received a pFAS diagnosis, and 154 would have received a diagnosis of ARND or a "no-FASD" diagnosis. The frequency of all but five dysmorphic features differ significantly between this sample and the sample examined for the 2016 criteria. There is more variability in the features present in the current sample. DISCUSSION Differences regarding diagnostic outcomes and prevalence of dysmorphic features suggest that strict application of the diagnostic criteria may miss children who present with FAS. We recommend including gestalt-based screening in a research setting where the clinical experience is available to inform future guidelines.
Collapse
Affiliation(s)
- Denis Viljoen
- Foundation for Alcohol-Related Research (FARR), Cape Town, South Africa.,Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Jacobus G Louw
- Foundation for Alcohol-Related Research (FARR), Cape Town, South Africa.,Department of Psychology, Stellenbosch University, Stellenbosch, South Africa
| | - Chanelle Lombard
- Foundation for Alcohol-Related Research (FARR), Cape Town, South Africa
| | - Leana Olivier
- Foundation for Alcohol-Related Research (FARR), Cape Town, South Africa.,Division of Molecular Biology and Human Genetics, Faculty of Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| |
Collapse
|
37
|
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: 57] [Impact Index Per Article: 9.5] [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
|
38
|
Loke YJ, Muggli E, Nguyen L, Ryan J, Saffery R, Elliott EJ, Halliday J, Craig JM. Time- and sex-dependent associations between prenatal alcohol exposure and placental global DNA methylation. Epigenomics 2018; 10:981-991. [PMID: 29956547 DOI: 10.2217/epi-2017-0147] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
AIM Epigenetic changes, in particular in the placenta, may mediate the effects of prenatal alcohol exposure (PAE) on children's health. We examined the relationship between PAE patterns, based on dose and timing, and placental global DNA methylation. METHODS Using linear regression analysis, we examined the association between different PAE categories and placental global DNA methylation (n = 187), using the proxy measure of Alu-interspersed repeats. RESULTS Following adjustment for important covariates, we found no evidence of an association between PAE and placental global DNA methylation overall. However, when stratifying by newborn sex, PAE throughout pregnancy was associated with higher placental global DNA methylation (1.5%; p = 0.01) of male newborns. CONCLUSION PAE may have sex-specific effects on placental global DNA methylation if alcohol is consumed throughout pregnancy.
Collapse
Affiliation(s)
- Yuk Jing Loke
- Environmental & Genetic Epidemiology Research, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Evelyne Muggli
- Public Health Genetics, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Linh Nguyen
- Environmental & Genetic Epidemiology Research, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Joanne Ryan
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia.,Department of Epidemiology & Preventive Medicine, Monash University, Melbourne, Victoria 3004, Australia.,Cancer & Disease Epigenetics, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Richard Saffery
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia.,Cancer & Disease Epigenetics, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | - Elizabeth J Elliott
- Discipline of Child & Adolescent Health, School of Medicine and Health, University of Sydney, Sydney 2006, New South Wales, Australia.,Australian Paediatric Surveillance Unit, Sydney Childrens Hospitals Network, Westmead, Sydney, New South Wales, Australia
| | - Jane Halliday
- Public Health Genetics, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jeffrey M Craig
- Environmental & Genetic Epidemiology Research, Murdoch Children's Research Institute, the Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia.,Centre for Molecular and Medical Research, Deakin University, Geelong Waurn Ponds Campus, Locked Bag 20000, Geelong, Victoria 3220, Australia
| |
Collapse
|
39
|
Frey S, Eichler A, Stonawski V, Kriebel J, Wahl S, Gallati S, Goecke TW, Fasching PA, Beckmann MW, Kratz O, Moll GH, Heinrich H, Kornhuber J, Golub Y. Prenatal Alcohol Exposure Is Associated With Adverse Cognitive Effects and Distinct Whole-Genome DNA Methylation Patterns in Primary School Children. Front Behav Neurosci 2018; 12:125. [PMID: 29997484 PMCID: PMC6028559 DOI: 10.3389/fnbeh.2018.00125] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Prenatal alcohol exposure (PAE) is known to elicit a broad range of systemic effects, including neurophysiological alterations that result in adverse behavioral and cognitive outcomes. However, molecular pathways underlying these long-term intrauterine effects remain to be investigated. Here, we tested a hypothesis that PAE may lead to epigenetic alterations to the DNA resulting in attentional and cognitive alterations of the children. We report the results of the study that included 156 primary school children of the Franconian Cognition and Emotion Studies (FRANCES) cohort which were tested for an objective marker of PAE, ethyl glucuronide (EtG) in meconium at birth. Thirty-two newborns were found to be exposed to alcohol with EtG values above 30 ng/g (EtG+). Previously we described PAE being associated with lower IQ and smaller amplitude of the event-related potential component P3 in go trials (Go-P3), which indicates a reduced capacity of attentional resources. Whole-genome methylation analysis of the buccal cell DNA revealed 193 differentially methylated genes in children with positive meconium EtG, that were clustered into groups involved in epigenetic modifications, neurodegeneration, neurodevelopment, axon guidance and neuronal excitability. Furthermore, we detected mediation effects of the methylation changes in DPP10 and SLC16A9 genes on the EtG related cognitive and attention-related deficits. Our results suggest that system-wide epigenetic changes are involved in long-term effects of PAE. In particular, we show an epigenetic mediation of PAE effects on cognition and attention-related processes.
Collapse
Affiliation(s)
- Stefan Frey
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Eichler
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Valeska Stonawski
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, German Research Center for Environmental Health - Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, German Research Center for Environmental Health - Institute of Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Sabina Gallati
- Division of Human Genetics, Department of Paediatrics, Inselspital University of Bern, Bern, Switzerland
| | - Tamme W Goecke
- Department of Obstetrics and Gynecology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Peter A Fasching
- Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Matthias W Beckmann
- Department of Obstetrics and Gynecology, RWTH Aachen University, Aachen, Germany
| | - Oliver Kratz
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Gunther H Moll
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Hartmut Heinrich
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,kbo-Heckscher-Klinikum, Munich, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Yulia Golub
- Department of Child and Adolescent Mental Health, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
| |
Collapse
|
40
|
Noor S, Milligan ED. Lifelong Impacts of Moderate Prenatal Alcohol Exposure on Neuroimmune Function. Front Immunol 2018; 9:1107. [PMID: 29910801 PMCID: PMC5992426 DOI: 10.3389/fimmu.2018.01107] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/02/2018] [Indexed: 12/26/2022] Open
Abstract
In utero alcohol exposure is emerging as a major risk factor for lifelong aberrant neuroimmune function. Fetal alcohol spectrum disorder encompasses a range of behavioral and physiological sequelae that may occur throughout life and includes cognitive developmental disabilities as well as disease susceptibility related to aberrant immune and neuroimmune actions. Emerging data from clinical studies and findings from animal models support that very low to moderate levels of fetal alcohol exposure may reprogram the developing central nervous system leading to altered neuroimmune and neuroglial signaling during adulthood. In this review, we will focus on the consequences of low to moderate prenatal alcohol exposure (PAE) on neuroimmune interactions during early life and at different stages of adulthood. Data discussed here will include recent studies suggesting that while abnormal immune function is generally minimal under basal conditions, following pathogenic stimuli or trauma, significant alterations in the neuroimmune axis occur. Evidence from published reports will be discussed with a focus on observations that PAE may bias later-life peripheral immune responses toward a proinflammatory phenotype. The propensity for proinflammatory responses to challenges in adulthood may ultimately shape neuron–glial-immune processes suspected to underlie various neuropathological outcomes including chronic pain and cognitive impairment.
Collapse
Affiliation(s)
- Shahani Noor
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Erin D Milligan
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
41
|
Nonprotein-coding RNAs in Fetal Alcohol Spectrum Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:299-342. [PMID: 29933954 DOI: 10.1016/bs.pmbts.2017.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Early developmental exposure to ethanol, a known teratogen, can result in a range of neurodevelopmental disorders, collectively referred to as Fetal Alcohol Spectrum Disorders (FASDs). Changes in the environment, including exposure to teratogens, can result in long term alterations to the epigenetic landscape of a cell, thereby altering gene expression. Noncoding RNAs (ncRNAs) can affect transcription and translation of networks of genes. ncRNAs are dynamically expressed during development and have been identified as a target of alcohol. ncRNAs therefore make for attractive targets for novel therapeutics to address the developmental deficits associated with FASDs.
Collapse
|
42
|
Liu C, Jiao C, Wang K, Yuan N. DNA Methylation and Psychiatric Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 157:175-232. [PMID: 29933950 DOI: 10.1016/bs.pmbts.2018.01.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
DNA methylation has been an important area of research in the study of molecular mechanism to psychiatric disorders. Recent evidence has suggested that abnormalities in global methylation, methylation of genes, and pathways could play a role in the etiology of many forms of mental illness. In this article, we review the mechanisms of DNA methylation, including the genetic and environmental factors affecting methylation changes. We report and discuss major findings regarding DNA methylation in psychiatric patients, both within the context of global methylation studies and gene-specific methylation studies. Finally, we discuss issues surrounding data quality improvement, the limitations of current methylation analysis methods, and the possibility of using DNA methylation-based treatment for psychiatric disorders in the future.
Collapse
Affiliation(s)
- Chunyu Liu
- University of Illinois, Chicago, IL, United States; School of Life Science, Central South University, Changsha, China.
| | - Chuan Jiao
- School of Life Science, Central South University, Changsha, China
| | - Kangli Wang
- School of Life Science, Central South University, Changsha, China
| | - Ning Yuan
- Hunan Brain Hospital, Changsha, China
| |
Collapse
|
43
|
Legault LM, Bertrand-Lehouillier V, McGraw S. Pre-implantation alcohol exposure and developmental programming of FASD: an epigenetic perspective. Biochem Cell Biol 2018; 96:117-130. [DOI: 10.1139/bcb-2017-0141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Exposure to alcohol during in-utero development can permanently change the developmental programming of physiological responses, thereby increasing the risk of neurological illnesses during childhood and later adverse health outcomes associated with fetal alcohol spectrum disorder (FASD). There is an increasing body of evidence indicating that exposure to alcohol during gestation triggers lasting epigenetic alterations in offspring, long after the initial insult; together, these studies support the role of epigenetics in FASD etiology. However, we still have little information about how ethanol interferes with the fundamental epigenetic reprogramming wave (e.g., erasure and re-establishment of DNA methylation marks) that characterizes pre-implantation embryo development. This review examines key epigenetic processes that occur during pre-implantation development and especially focus on the current knowledge regarding how prenatal exposure to alcohol during this period could affect the developmental programming of the early stage pre-implantation embryo. We will also outline the current limitations of studies examining the in-vivo and in-vitro effects of alcohol exposure on embryos and underline the next critical steps to be taken if we want to better understand the implicated mechanisms to strengthen the translational potential for epigenetic markers for non-invasive early detection, and the treatment of newborns that have higher risk of developing FASD.
Collapse
Affiliation(s)
- Lisa-Marie Legault
- Department of Biochemistry and Molecular Medicine, Université de Montreal, Research Center of the CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC H3T 1C5, Canada
| | - Virginie Bertrand-Lehouillier
- Department of Biochemistry and Molecular Medicine, Université de Montreal, Research Center of the CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC H3T 1C5, Canada
| | - Serge McGraw
- Department of Biochemistry and Molecular Medicine, Université de Montreal, Research Center of the CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC H3T 1C5, Canada
- Obstetrics and Gynecology, Université de Montreal, Research Center of the CHU Sainte-Justine, Montreal, Canada
| |
Collapse
|
44
|
Knopik VS, Marceau K, Bidwell LC, Rolan E. Prenatal substance exposure and offspring development: Does DNA methylation play a role? Neurotoxicol Teratol 2018; 71:50-63. [PMID: 29408446 DOI: 10.1016/j.ntt.2018.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/12/2018] [Accepted: 01/24/2018] [Indexed: 12/17/2022]
Abstract
The period of in utero development is one of the most critical windows during which adverse conditions and exposures may influence the growth and development of the fetus as well as its future postnatal health and behavior. Maternal substance use during pregnancy remains a relatively common but nonetheless hazardous in utero exposure. For example, previous epidemiological studies have associated prenatal substance exposure with reduced birth weight, poor developmental and psychological outcomes, and increased risk for diseases and behavioral disorders (e.g., externalizing behaviors like ADHD, conduct disorder, and substance use) later in life. Researchers are now learning that many of the mechanisms whereby adverse in utero exposures may affect key pathways crucial for proper fetal growth and development are epigenetic in nature, with the majority of work in humans considering DNA methylation specifically. This review will explore the research to date on epigenetic alterations tied to maternal substance use during pregnancy and will also discuss the possible role of DNA methylation in the robust relationship between maternal substance use and later behavioral and developmental sequelae in offspring.
Collapse
Affiliation(s)
- Valerie S Knopik
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA.
| | - Kristine Marceau
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| | - L Cinnamon Bidwell
- Institute of Cognitive Science, University of Colorado, Boulder, CO, USA
| | - Emily Rolan
- Department of Human Development and Family Studies, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
45
|
Saffari A, Silver MJ, Zavattari P, Moi L, Columbano A, Meaburn EL, Dudbridge F. Estimation of a significance threshold for epigenome-wide association studies. Genet Epidemiol 2018; 42:20-33. [PMID: 29034560 PMCID: PMC5813244 DOI: 10.1002/gepi.22086] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/30/2017] [Accepted: 07/24/2017] [Indexed: 12/17/2022]
Abstract
Epigenome-wide association studies (EWAS) are designed to characterise population-level epigenetic differences across the genome and link them to disease. Most commonly, they assess DNA-methylation status at cytosine-guanine dinucleotide (CpG) sites, using platforms such as the Illumina 450k array that profile a subset of CpGs genome wide. An important challenge in the context of EWAS is determining a significance threshold for declaring a CpG site as differentially methylated, taking multiple testing into account. We used a permutation method to estimate a significance threshold specifically for the 450k array and a simulation extrapolation approach to estimate a genome-wide threshold. These methods were applied to five different EWAS datasets derived from a variety of populations and tissue types. We obtained an estimate of α=2.4×10-7 for the 450k array, and a genome-wide estimate of α=3.6×10-8. We further demonstrate the importance of these results by showing that previously recommended sample sizes for EWAS should be adjusted upwards, requiring samples between ∼10% and ∼20% larger in order to maintain type-1 errors at the desired level.
Collapse
Affiliation(s)
- Ayden Saffari
- Department of Non‐Communicable Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondonUnited Kingdom
- MRC Unit, The Gambia and MRC International Nutrition GroupLondon School of Hygiene and Tropical MedicineLondonUnited Kingdom
- Department of Psychological Sciences, BirkbeckUniversity of LondonLondonUnited Kingdom
| | - Matt J. Silver
- MRC Unit, The Gambia and MRC International Nutrition GroupLondon School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Patrizia Zavattari
- Department of Biomedical SciencesUniversity of CagliariCagliariSardiniaItaly
| | - Loredana Moi
- Department of Biomedical SciencesUniversity of CagliariCagliariSardiniaItaly
| | - Amedeo Columbano
- Department of Biomedical SciencesUniversity of CagliariCagliariSardiniaItaly
| | - Emma L. Meaburn
- Department of Psychological Sciences, BirkbeckUniversity of LondonLondonUnited Kingdom
| | - Frank Dudbridge
- Department of Non‐Communicable Disease EpidemiologyLondon School of Hygiene and Tropical MedicineLondonUnited Kingdom
- Department of Health SciencesUniversity of LeicesterLeicesterUnited Kingdom
| |
Collapse
|
46
|
Lussier AA, Morin AM, MacIsaac JL, Salmon J, Weinberg J, Reynolds JN, Pavlidis P, Chudley AE, Kobor MS. DNA methylation as a predictor of fetal alcohol spectrum disorder. Clin Epigenetics 2018; 10:5. [PMID: 29344313 PMCID: PMC5767049 DOI: 10.1186/s13148-018-0439-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/04/2018] [Indexed: 12/22/2022] Open
Abstract
Background Fetal alcohol spectrum disorder (FASD) is a developmental disorder that manifests through a range of cognitive, adaptive, physiological, and neurobiological deficits resulting from prenatal alcohol exposure. Although the North American prevalence is currently estimated at 2-5%, FASD has proven difficult to identify in the absence of the overt physical features characteristic of fetal alcohol syndrome. As interventions may have the greatest impact at an early age, accurate biomarkers are needed to identify children at risk for FASD. Building on our previous work identifying distinct DNA methylation patterns in children and adolescents with FASD, we have attempted to validate these associations in a different clinical cohort and to use our DNA methylation signature to develop a possible epigenetic predictor of FASD. Methods Genome-wide DNA methylation patterns were analyzed using the Illumina HumanMethylation450 array in the buccal epithelial cells of a cohort of 48 individuals aged 3.5-18 (24 FASD cases, 24 controls). The DNA methylation predictor of FASD was built using a stochastic gradient boosting model on our previously published dataset FASD cases and controls (GSE80261). The predictor was tested on the current dataset and an independent dataset of 48 autism spectrum disorder cases and 48 controls (GSE50759). Results We validated findings from our previous study that identified a DNA methylation signature of FASD, replicating the altered DNA methylation levels of 161/648 CpGs in this independent cohort, which may represent a robust signature of FASD in the epigenome. We also generated a predictive model of FASD using machine learning in a subset of our previously published cohort of 179 samples (83 FASD cases, 96 controls), which was tested in this novel cohort of 48 samples and resulted in a moderately accurate predictor of FASD status. Upon testing the algorithm in an independent cohort of individuals with autism spectrum disorder, we did not detect any bias towards autism, sex, age, or ethnicity. Conclusion These findings further support the association of FASD with distinct DNA methylation patterns, while providing a possible entry point towards the development of epigenetic biomarkers of FASD.
Collapse
Affiliation(s)
- Alexandre A. Lussier
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia Canada
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia Canada
| | - Alexander M. Morin
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia Canada
| | - Julia L. MacIsaac
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia Canada
| | - Jenny Salmon
- Department of Pediatrics and Child Health, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia Canada
| | - James N. Reynolds
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience Studies, Queen’s University, Kingston, Ontario Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columnbia Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia Canada
| | - Albert E. Chudley
- Department of Pediatrics and Child Health, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba Canada
- Department of Biochemistry and Medical Genetics, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba Canada
| | - Michael S. Kobor
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, British Columbia Canada
- Human Early Learning Partnership, University of British Columbia, Vancouver, British Columbia Canada
| |
Collapse
|
47
|
Boschen KE, Keller SM, Roth TL, Klintsova AY. Epigenetic mechanisms in alcohol- and adversity-induced developmental origins of neurobehavioral functioning. Neurotoxicol Teratol 2018; 66:63-79. [PMID: 29305195 DOI: 10.1016/j.ntt.2017.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022]
Abstract
The long-term effects of developmental alcohol and stress exposure are well documented in both humans and non-human animal models. Damage to the brain and attendant life-long impairments in cognition and increased risk for psychiatric disorders are debilitating consequences of developmental exposure to alcohol and/or psychological stress. Here we discuss evidence for a role of epigenetic mechanisms in mediating these consequences. While we highlight some of the common ways in which stress or alcohol impact the epigenome, we point out that little is understood of the epigenome's response to experiencing both stress and alcohol exposure, though stress is a contributing factor as to why women drink during pregnancy. Advancing our understanding of this relationship is of critical concern not just for the health and well-being of individuals directly exposed to these teratogens, but for generations to come.
Collapse
Affiliation(s)
- K E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599, United States
| | - S M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - T L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - A Y Klintsova
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
48
|
Comasco E, Rangmar J, Eriksson UJ, Oreland L. Neurological and neuropsychological effects of low and moderate prenatal alcohol exposure. Acta Physiol (Oxf) 2018; 222. [PMID: 28470828 DOI: 10.1111/apha.12892] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/17/2017] [Accepted: 04/27/2017] [Indexed: 01/18/2023]
Abstract
Several explanations for the diverse results in research on foetal alcohol spectrum disorders or alcohol-related neurodevelopmental disorder might be at hand: timing, amount and patterns of alcohol exposure, as well as complex epigenetic responses. The genetic background of the offspring and its interaction with other prenatal and post-natal environmental cues are likely also of importance. In the present report, key findings about the possible effects of low and moderate doses of maternal alcohol intake on the neuropsychological development of the offspring are reviewed and plausible mechanisms discussed. Special focus is put on the serotonergic system within developmental and gene-environment frameworks. The review also suggests guidelines for future studies and also summarizes some of to-be-answered questions of relevance to clinical practice. Contradictory findings and paucity of studies on the effects of exposure to low alcohol levels during foetal life for the offspring's neuropsychological development call for large prospective studies, as well as for studies including neuroimaging and multi-omics analyses to dissect the neurobiological underpinnings of alcohol exposure-related phenotypes and to identify biomarkers. Finally, it remains to be investigated whether any safe threshold of alcohol drinking during pregnancy can be identified.
Collapse
Affiliation(s)
- E. Comasco
- Department of Neuroscience; Uppsala University; Uppsala Sweden
| | - J. Rangmar
- Department of Psychology; University of Gothenburg; Gothenburg Sweden
| | - U. J. Eriksson
- Department of Medical Cell Biology; Uppsala University; Uppsala Sweden
| | - L. Oreland
- Department of Neuroscience; Uppsala University; Uppsala Sweden
| |
Collapse
|
49
|
Miozzo F, Arnould H, de Thonel A, Schang AL, Sabéran-Djoneidi D, Baudry A, Schneider B, Mezger V. Alcohol exposure promotes DNA methyltransferase DNMT3A upregulation through reactive oxygen species-dependent mechanisms. Cell Stress Chaperones 2018; 23:115-126. [PMID: 28712054 PMCID: PMC5741586 DOI: 10.1007/s12192-017-0829-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022] Open
Abstract
Abundant evidence has accumulated showing that fetal alcohol exposure broadly modifies DNA methylation profiles in the brain. DNA methyltransferases (DNMTs), the enzymes responsible for DNA methylation, are likely implicated in this process. However, their regulation by ethanol exposure has been poorly addressed. Here, we show that alcohol exposure modulates DNMT protein levels through multiple mechanisms. Using a neural precursor cell line and primary mouse embryonic fibroblasts (MEFs), we found that ethanol exposure augments the levels of Dnmt3a, Dnmt3b, and Dnmt3l transcripts. We also unveil similar elevation of mRNA levels for other epigenetic actors upon ethanol exposure, among which the induction of lysine demethylase Kdm6a shows heat shock factor dependency. Furthermore, we show that ethanol exposure leads to specific increase in DNMT3A protein levels. This elevation not only relies on the upregulation of Dnmt3a mRNA but also depends on posttranscriptional mechanisms that are mediated by NADPH oxidase-dependent production of reactive oxygen species (ROS). Altogether, our work underlines complex regulation of epigenetic actors in response to alcohol exposure at both transcriptional and posttranscriptional levels. Notably, the upregulation of DNMT3A emerges as a prominent molecular event triggered by ethanol, driven by the generation of ROS.
Collapse
Affiliation(s)
- Federico Miozzo
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
- Department of Genetics and Evolution, Sciences III, University of Geneva, Geneva, Switzerland
| | - Hélène Arnould
- INSERM UMR-S1124, Paris Cedex 6, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France
| | - Aurélie de Thonel
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
| | - Anne-Laure Schang
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
- UMR CNRS 8638-Chimie Toxicologie Analytique et Cellulaire, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France
- Département Hospitalo-Universitaire PROTECT, Paris, France
| | - Anne Baudry
- INSERM UMR-S1124, Paris Cedex 6, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France
| | - Benoît Schneider
- INSERM UMR-S1124, Paris Cedex 6, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris Cedex 6, France.
| | - Valérie Mezger
- CNRS, UMR7216 Épigénétique et Destin Cellulaire, F-75205, Paris Cedex 13, France.
- Univ Paris Diderot, Sorbonne Paris Cité, F-75205, Paris Cedex 13, France.
- Département Hospitalo-Universitaire PROTECT, Paris, France.
| |
Collapse
|
50
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|