1
|
Vieiros M, Almeida-Toledano L, Serra-Delgado M, Navarro-Tapia E, Ramos-Triguero A, Muñoz-Lozano C, Martínez L, Marchei E, Gómez-Roig MD, García-Algar Ó, Andreu-Fernández V. Effects of maternal drinking patterns and epigallocatechin-3-gallate treatment on behavioural and molecular outcomes in a mouse model of fetal alcohol spectrum disorders. Biomed Pharmacother 2025; 187:118138. [PMID: 40349554 DOI: 10.1016/j.biopha.2025.118138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Prenatal alcohol exposure (PAE) impairs fetal development leading to fetal alcohol spectrum disorders (FASD). Antioxidants like epigallocatechin-3-gallate (EGCG) may mitigate alcohol-induced oxidative stress, a major contributor to FASD. This study assessed the effects of PAE on cognition and behaviour under two drinking patterns and the role of postnatal EGCG therapy in a FASD-like mouse model. C57BL/6J mice were divided into five groups: control, moderate drinking (Mod), binge drinking (Bin), Mod+EGCG, and Bin+EGCG. Cognitive and behavioural performance were assessed using Rotarod test, T-Maze, and Morris Water Maze (MWM). Western blot analyses evaluated brain and cerebellum biomarkers related to neuronal plasticity, maturation, differentiation, transport, and proliferation. PAE impaired motor coordination, significantly reducing rotarod walking time in both drinking patterns. Spatial learning and memory were also disrupted, decreasing T-maze success rate. It also decreased time in the platform area and distance travelled in MWM. Both drinking patterns affected neuronal plasticity (BDNF, DYRK1A) and maturation (NeuN), astrocyte differentiation (GFAP, s100β), neuronal transport (MBP) and proliferation (GDNF, Wnt-3) via oxidative stress (Nrf2). Our results show how EGCG treatment significantly improved behavioural tests results and restored most brain and cerebellum biomarkers, reaching levels similar to control. These findings highlight the impact of PAE on cognition and behaviour and how EGCG may counteract its effects by reducing oxidative stress and enhancing brain plasticity. Our findings open the door to future studies on the mechanism of action of this antioxidant in order to use it as a therapeutic tool in this vulnerable population.
Collapse
Affiliation(s)
- Melina Vieiros
- Grup de Recerca Infancia i Entorn, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Spanish network in maternal, neonatal, child, and developmental health research (RICORS-SAMID, RD21/0012/0017, RD24/0013/0019) Instituto de Salud Carlos III, Madrid, Spain; Department de Cirurgia i Especialitats Mèdico-Quirúrgiques, Universitat de Barcelona, Barcelona, Spain
| | - Laura Almeida-Toledano
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat 08950, Spain; BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and and Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Mariona Serra-Delgado
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat 08950, Spain; BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and and Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Elisabet Navarro-Tapia
- Spanish network in maternal, neonatal, child, and developmental health research (RICORS-SAMID, RD21/0012/0017, RD24/0013/0019) Instituto de Salud Carlos III, Madrid, Spain; Faculty of Health Sciences, Valencian International University (VIU), Valencia, Spain; Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, Barcelona, Spain
| | - Anna Ramos-Triguero
- Grup de Recerca Infancia i Entorn, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Department de Cirurgia i Especialitats Mèdico-Quirúrgiques, Universitat de Barcelona, Barcelona, Spain; Institute for Biomedical Research La Paz (IdiPaz), Madrid, Spain
| | - Concha Muñoz-Lozano
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat 08950, Spain; BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and and Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Leopoldo Martínez
- Institute for Biomedical Research La Paz (IdiPaz), Madrid, Spain; Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Emilia Marchei
- National Centre on Addiction and Doping, Istituto Superiore di Sanità, Rome, Italy
| | - María D Gómez-Roig
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat 08950, Spain; BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and and Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Óscar García-Algar
- Grup de Recerca Infancia i Entorn, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Spanish network in maternal, neonatal, child, and developmental health research (RICORS-SAMID, RD21/0012/0017, RD24/0013/0019) Instituto de Salud Carlos III, Madrid, Spain; Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infancia i Entorn, Institut d'investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; Faculty of Health Sciences, Valencian International University (VIU), Valencia, Spain; Biosanitary Research Institute, Valencian International University, Valencia, Spain.
| |
Collapse
|
2
|
Dursun I, Korkmaz ND, Firtina S, Erkoyuncu MS, Akbas F, Elibol B. Exploring epigenetic modification of the stress-related FKBP5 gene in mice exposed to alcohol during early postnatal development. Alcohol 2025; 123:11-17. [PMID: 39245355 DOI: 10.1016/j.alcohol.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Early developmental exposure to alcohol has been implicated in adverse effects on the brain, often associated with the onset of neurodevelopmental disorders. Moreover, maternal alcohol consumption during pregnancy has been linked to the manifestation of mental health disorders, such as depression and anxiety, in subsequent generations. These mood disturbances may be attributed to alterations in protein expressions related to depression and anxiety within the hippocampus. While the precise mechanisms remain elusive, it is likely that pre- and postnatal exposure to alcohol induces changes in hippocampus, potentially through epigenetic modifications. The FKBP5 gene, known to modulate the stress response, is particularly relevant in this context. We postulate that alcohol-induced methylation of the FKBP5 gene disrupts HPA axis function, thereby prompting individuals to anxiety-like and depressive-like behaviors. To investigate this hypothesis, female C57BL/6 pups were subjected to early alcohol exposure via intubation with ethanol mixed in artificial milk from Postnatal Day 3 to Day 20. The intubation control pups were subjected to the same procedures without ethanol or milk, and a non-intubated control group included. Anxiety-like and depressive-like behaviors were assessed using the open field test, plus maze test, forced swim test, and tail suspension test when the pups reached 3 months of age. For epigenetic analysis of the FKBP5 gene, genomic DNA was isolated from hippocampal tissues and subjected to bisulfite conversion to distinguish methylated and unmethylated cytosines. Then, methylation-specific PCR was performed to assess methylation levels. Pups exposed to early postnatal alcohol exhibited increased levels of depression-like behavior and susceptibility to anxiety-like behavior during adolescence, as verified by behavioral assessments. Methylation profiling revealed higher rates of methylation within the stress-associated gene FKBP5 in both the early postnatal alcohol-exposed cohort (13.82%) and the intubation control group (3.93%), in contrast to the control cohort devoid of stress or alcohol exposure. These findings suggest a potential epigenetic mechanism underlying the observed behavioral alterations, implicating FKBP5 methylation as a candidate mediator of the increased vulnerability to mood disorders following early postnatal alcohol exposure.
Collapse
Affiliation(s)
- Ilknur Dursun
- Department of Physiology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| | - Nur Damla Korkmaz
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Sinem Firtina
- Department of Medical Genetics, Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Muhammed Salih Erkoyuncu
- Department of Neuroscience, Graduate School of Health Sciences, Bezmialem Vakif University, Istanbul, Turkey
| | - Fahri Akbas
- Department of Medical Biology, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Turkey
| | - Birsen Elibol
- Department of Medical Biology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey.
| |
Collapse
|
3
|
Hu P, Hao Y, Tang W, Diering GH, Zou F, Kafri T. Analysis of Hepatic Lentiviral Vector Transduction: Implications for Preclinical Studies and Clinical Gene Therapy Protocols. Viruses 2025; 17:276. [PMID: 40007031 PMCID: PMC11861806 DOI: 10.3390/v17020276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Lentiviral vector-transduced T cells were approved by the FDA as gene therapy anti-cancer medications. Little is known about the effects of host genetic variation on the safety and efficacy of the lentiviral vector gene delivery system. To narrow this knowledge gap, we characterized hepatic gene delivery by lentiviral vectors across the Collaborative Cross (CC) mouse genetic reference population. For 24 weeks, we periodically measured hepatic luciferase expression from lentiviral vectors in 41 CC mouse strains. Hepatic and splenic vector copy numbers were determined. We report that the CC mouse strains showed highly diverse outcomes following lentiviral gene delivery. For the first time, a moderate correlation between mouse-strain-specific sleeping patterns and transduction efficiency was observed. We associated two quantitative trait loci (QTLs) with intrastrain variations in transduction phenotypes, which mechanistically relates to the phenomenon of metastable epialleles. An additional QTL was associated with the kinetics of hepatic transgene expression. Genes found in the above QTLs are potential targets for personalized gene therapy protocols. Importantly, we identified two mouse strains that open new directions for characterizing continuous viral vector silencing and HIV latency. Our findings suggest that wide-range patient-specific outcomes of viral vector-based gene therapy should be expected. Thus, novel clinical protocols should be considered for non-fatal diseases.
Collapse
Affiliation(s)
- Peirong Hu
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA (W.T.)
| | - Yajing Hao
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Wei Tang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA (W.T.)
| | - Graham H. Diering
- Department of Cell Biology and Physiology and UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, Carrboro, NC 27510, USA
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA (W.T.)
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Curti L, Rizzi B, Mottarlini F, Bigagli E, Ilari A, Costa A, Sordi V, Ranieri G, Luceri C, Cannella N, Ubaldi M, Masi A, Fumagalli F, Caffino L, Mannaioni G, Gerace E. Prenatal ethanol exposure impairs hippocampal plasticity and cognition in adolescent mice. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111174. [PMID: 39447689 DOI: 10.1016/j.pnpbp.2024.111174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) induces a wide range of neurodevelopmental disabilities that are grouped under the term 'fetal alcohol spectrum disorders' (FASD). The effects of PAE on brain development are dependent on complex neurochemical events, including modification of AMPA receptors (AMPARs). We have recently found that chronic ethanol (EtOH) exposure decreases AMPA-mediated neurotransmission and expression through the overexpression of the specific microRNA (miR)137 and 501-3p, which target GluA1 AMPA subunit, in the developing hippocampus in vitro. Here, we explored how PAE mice may alter AMPAergic synapses in the hippocampus, and its effects on behavior. METHODS To model PAE, we exposed C57Bl/6 pregnant mice to 10 % EtOH during during the first 10 days of gestation (GD 0-10; equivalent to the first trimester of pregnancy in humans). AMPA subunits postsynaptic expression in the hippocampus, electrical properties of CA1 neurons, memory recognition, and locomotor functions were then analyzed in adolescent PAE-exposed offspring. RESULTS PAE adolescent mice showed dysregulation of AMPAergic neurotransmission, and increased miR 501-3p expression, associated with a significant reduction of spontaneous AMPA currents and intrinsic somatic excitability. In addition, PAE reduced the phosphorylation of AMPAR-containing GluA1 subunit, despite an increase in its total levels. Of note, the total levels of GluA2 and GluA3 AMPA receptors were enhanced as well. Consistently, at behavioral level, PAE reduced object recognition without altering locomotor activity. CONCLUSIONS Our study shows that PAE leads to dysfunctional formation of AMPAergic synapses that could be responsible for neurobehavioral impairments, contributing to the understanding of the pathogenesis of FASD.
Collapse
Affiliation(s)
- Lorenzo Curti
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Beatrice Rizzi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy, University of Florence, Florence, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy, University of Florence, Florence, Italy
| | - Elisabetta Bigagli
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Alice Ilari
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Alessia Costa
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Virginia Sordi
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; School of Pharmacy, Pharmacology Unit, Centre for Neuroscience, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Giuseppe Ranieri
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Cristina Luceri
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, Centre for Neuroscience, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Massimo Ubaldi
- School of Pharmacy, Pharmacology Unit, Centre for Neuroscience, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy, University of Florence, Florence, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy, University of Florence, Florence, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Elisabetta Gerace
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; Department of Health Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
5
|
Liu H, Ren Q, Gong M, Zuo F, Li Q, Huo D, Yuan Y, Zhang Y, Kong Y, Liu X, Lu C, Wu X. Enforced activation of the CREB/KDM2B axis prevents alcohol-induced embryonic developmental delay. Cell Rep 2024; 43:115075. [PMID: 39661511 DOI: 10.1016/j.celrep.2024.115075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Unintentional, early pregnancy alcohol consumption affects embryonic development. During the peri-implantation stage, coinciding with the transition from naive to primed pluripotency, the long isoform of KDM2B (KDM2BLF) underlies the de novo establishment of polycomb repressive complex (PRC) functions at promoters after fertilization. However, it remains unclear whether and how ethanol exposure affects this spatiotemporal chromatin setting. Here, we show that exposing peri-implantation mouse embryos to ethanol leads to impaired post-implantation development, mirrored by the delayed exit of naive pluripotency in acetaldehyde-treated embryonic stem cells. Remarkably, these abnormalities are linked to inadequate KDM2BLF expression and compromised deposition of PRC marks, which arise from cAMP response element-binding protein (CREB) inactivation. Accordingly, pharmacological activation of CREB effectively restores pluripotency transition partly dependent on KDM2BLF in vitro and ameliorates post-implantation embryonic defects in vivo. Therefore, our study highlights the pivotal role of the CREB/KDM2B axis in chromatin configuration and developmental programming, proposing potential preventive strategies against ethanol exposure-induced detrimental effects.
Collapse
Affiliation(s)
- Hang Liu
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Qiyu Ren
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100081, China
| | - Meihan Gong
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Feifei Zuo
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Qian Li
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Dawei Huo
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China; Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Laboratory, Institute of Hematology, Zhejiang University, Hangzhou 311113, China
| | - Ye Yuan
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Yutong Zhang
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100081, China
| | - Yu Kong
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin 300450, China
| | - Cailing Lu
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100081, China.
| | - Xudong Wu
- State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China; Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin 300450, China.
| |
Collapse
|
6
|
Gillespie CA, Chowdhury A, Quinn KA, Jenkins MW, Rollins AM, Watanabe M, Ford SM. Fundamentals of DNA methylation in development. Pediatr Res 2024:10.1038/s41390-024-03674-7. [PMID: 39658604 DOI: 10.1038/s41390-024-03674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/12/2024] [Accepted: 09/30/2024] [Indexed: 12/12/2024]
Abstract
DNA methyation is critical to regulation of gene expression especially during developmentally dynamic changes. A large proportion occurs at CpG (a cytosine followed by a guanine nucleotide) sites and impacts gene expression based on location, timing and level of DNA methylation. The spectrum of effects produced by DNA methylation ranges from inhibition to enhancement of gene expression. Here basic terms and concepts in the study of DNA methylation are introduced. In addition, some of the commonly used techniques to assay DNA methylation are explained. New methods that allow the precise addition and removal of DNA methylation at specific sites will likely enhance our understanding of DNA methylation in development and may even lead to long-lasting therapeutic strategies to cure diseases. IMPACT: Fundamentals of DNA methylation including its significance are made accessible to a broad audience. Common assays for detecting DNA methylation are explained succinctly. Developmental patterns of DNA methylation detected in commonly used animal models are discussed and explained. Novel methodologies to investigate consequences of DNA methylation and demethylation are introduced.
Collapse
Affiliation(s)
- Caitlyn A Gillespie
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Amrin Chowdhury
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Katie A Quinn
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michiko Watanabe
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Stephanie M Ford
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Divisions of Neonatology and Pediatric Cardiology, UH Rainbow Babies and Children's Hospital, Cleveland, OH, 44106, USA.
| |
Collapse
|
7
|
Bestry M, Larcombe AN, Kresoje N, Chivers EK, Bakker C, Fitzpatrick JP, Elliott EJ, Craig JM, Muggli E, Halliday J, Hutchinson D, Buckberry S, Lister R, Symons M, Martino D. Early moderate prenatal alcohol exposure and maternal diet impact offspring DNA methylation across species. eLife 2024; 12:RP92135. [PMID: 39239947 PMCID: PMC11379454 DOI: 10.7554/elife.92135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Alcohol consumption in pregnancy can affect genome regulation in the developing offspring but results have been contradictory. We employed a physiologically relevant murine model of short-term moderate prenatal alcohol exposure (PAE) resembling common patterns of alcohol consumption in pregnancy in humans. Early moderate PAE was sufficient to affect site-specific DNA methylation in newborn pups without altering behavioural outcomes in adult littermates. Whole-genome bisulfite sequencing of neonatal brain and liver revealed stochastic influence on DNA methylation that was mostly tissue-specific, with some perturbations likely originating as early as gastrulation. DNA methylation differences were enriched in non-coding genomic regions with regulatory potential indicative of broad effects of alcohol on genome regulation. Replication studies in human cohorts with fetal alcohol spectrum disorder suggested some effects were metastable at genes linked to disease-relevant traits including facial morphology, intelligence, educational attainment, autism, and schizophrenia. In our murine model, a maternal diet high in folate and choline protected against some of the damaging effects of early moderate PAE on DNA methylation. Our studies demonstrate that early moderate exposure is sufficient to affect fetal genome regulation even in the absence of overt phenotypic changes and highlight a role for preventative maternal dietary interventions.
Collapse
Affiliation(s)
- Mitchell Bestry
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Alexander N Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
- Occupation, Environment and Safety, School of Population Health, Curtin University, Perth, Australia
| | | | - Emily K Chivers
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - Chloe Bakker
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| | - James P Fitzpatrick
- School of Psychological Sciences, University of Western Australia, Perth, Australia
| | - Elizabeth J Elliott
- University of Sydney, Faculty of Medicine and Health, Specialty of Child and Adolescent Health, Melbourne, Australia
- Sydney Children's Hospitals Network (Westmead) and Kids Research, Geelong, Australia
| | - Jeffrey M Craig
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Evelyne Muggli
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Jane Halliday
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Australia
- Reproductive Epidemiology, Murdoch Children's Research Institute, Parkville, Australia
| | - Delyse Hutchinson
- Reproductive Epidemiology, Murdoch Children's Research Institute, Parkville, Australia
- Deakin University, School of Psychology, Faculty of Health, Geelong, Australia
- Murdoch Children's Research Institute, Centre for Adolescent Health, Royal Children's Hospital, Melbourne, Australia
- University New South Wales, National Drug and Alcohol Research Centre, Sydney, Australia
| | - Sam Buckberry
- Telethon Kids Institute, Nedlands, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Australia
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Ryan Lister
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Australia
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Martyn Symons
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
- National Drug Research Institute, enAble Institute, Curtin University, Perth, Australia
| | - David Martino
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Australia
| |
Collapse
|
8
|
Hu P, Hao Y, Tang W, Diering GH, Zou F, Kafri T. Analysis of hepatic lentiviral vector transduction; implications for preclinical studies and clinical gene therapy protocols. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608805. [PMID: 39229157 PMCID: PMC11370356 DOI: 10.1101/2024.08.20.608805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Lentiviral vector-transduced T-cells were approved by the FDA as gene therapy anti-cancer medications. Little is known about the host genetic variation effects on the safety and efficacy of the lentiviral vector gene delivery system. To narrow this knowledge-gap, we characterized hepatic gene delivery by lentiviral vectors across the Collaborative Cross (CC) mouse genetic reference population. For 24 weeks, we periodically measured hepatic luciferase expression from lentiviral vectors in 41 CC mouse strains. Hepatic and splenic vector copy numbers were determined. We report that CC mouse strains showed highly diverse outcomes following lentiviral gene delivery. For the first time, moderate correlation between mouse strain-specific sleeping patterns and transduction efficiency was observed. We associated two quantitative trait loci (QTLs) with intra-strain variations in transduction phenotypes, which mechanistically relates to the phenomenon of metastable epialleles. An additional QTL was associated with the kinetics of hepatic transgene expression. Genes comprised in the above QTLs are potential targets to personalize gene therapy protocols. Importantly, we identified two mouse strains that open new directions in characterizing continuous viral vector silencing and HIV latency. Our findings suggest that wide-range patient-specific outcomes of viral vector-based gene therapy should be expected. Thus, novel escalating dose-based clinical protocols should be considered.
Collapse
Affiliation(s)
- Peirong Hu
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- These authors contributed equally
| | - Yajing Hao
- Department of Biostatistics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- These authors contributed equally
| | - Wei Tang
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
| | - Graham H. Diering
- Department of Cell Biology and Physiology and UNC Neuroscience Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Carolina Institute for developmental disabilities, 27510 Carrboro, North Carolina
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
| | - Tal Kafri
- Gene Therapy Center, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, 27599 Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, 27599 Chapel Hill, North Carolina
| |
Collapse
|
9
|
Higgins SL, Bhadsavle SS, Gaytan MN, Thomas KN, Golding MC. Chronic paternal alcohol exposures induce dose-dependent changes in offspring craniofacial shape and symmetry. Front Cell Dev Biol 2024; 12:1415653. [PMID: 39011393 PMCID: PMC11246915 DOI: 10.3389/fcell.2024.1415653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
Although dose-response analyses are a fundamental tool in developmental toxicology, few studies have examined the impacts of toxicant dose on the non-genetic paternal inheritance of offspring disease and dysgenesis. In this study, we used geometric morphometric analyses to examine the impacts of different levels of preconception paternal alcohol exposure on offspring craniofacial shape and symmetry in a mouse model. Procrustes ANOVA followed by canonical variant analysis of geometric facial relationships revealed that Low-, Medium-, and High-dose treatments each induced distinct changes in craniofacial shape and symmetry. Our analyses identified a dose threshold between 1.543 and 2.321 g/kg/day. Below this threshold, preconception paternal alcohol exposure induced changes in facial shape, including a right shift in facial features. In contrast, above this threshold, paternal exposures caused shifts in both shape and center, disrupting facial symmetry. Consistent with previous clinical studies, changes in craniofacial shape predominantly mapped to regions in the lower portion of the face, including the mandible (lower jaw) and maxilla (upper jaw). Notably, high-dose exposures also impacted the positioning of the right eye. Our studies reveal that paternal alcohol use may be an unrecognized factor contributing to the incidence and severity of alcohol-related craniofacial defects, complicating diagnostics of fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Samantha L Higgins
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Sanat S Bhadsavle
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Matthew N Gaytan
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Kara N Thomas
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Michael C Golding
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
10
|
Vieiros M, Navarro-Tapia E, Ramos-Triguero A, García-Meseguer À, Martínez L, García-Algar Ó, Andreu-Fernández V. Analysis of alcohol-metabolizing enzymes genetic variants and RAR/RXR expression in patients diagnosed with fetal alcohol syndrome: a case-control study. BMC Genomics 2024; 25:610. [PMID: 38886650 PMCID: PMC11184718 DOI: 10.1186/s12864-024-10516-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Understanding the mechanisms underlying alcohol metabolism and its regulation, including the effect of polymorphisms in alcohol-metabolizing enzymes, is crucial for research on Fetal Alcohol Spectrum Disorders. The aim of this study was to identify specific single nucleotide polymorphisms in key alcohol-metabolizing enzymes in a cohort of 71 children, including children with fetal alcohol syndrome, children prenatally exposed to ethanol but without fetal alcohol spectrum disorder, and controls. We hypothesized that certain genetic variants related to alcohol metabolism may be fixed in these populations, giving them a particular alcohol metabolism profile. In addition, the difference in certain isoforms of these enzymes determines their affinity for alcohol, which also affects the metabolism of retinoic acid, which is key to the proper development of the central nervous system. Our results showed that children prenatally exposed to ethanol without fetal alcohol spectrum disorder traits had a higher frequency of the ADH1B*3 and ADH1C*1 alleles, which are associated with increased alcohol metabolism and therefore a protective factor against circulating alcohol in the fetus after maternal drinking, compared to FAS children who had an allele with a lower affinity for alcohol. This study also revealed the presence of an ADH4 variant in the FAS population that binds weakly to the teratogen, allowing increased circulation of the toxic agent and direct induction of developmental abnormalities in the fetus. However, both groups showed dysregulation in the expression of genes related to the retinoic acid pathway, such as retinoic acid receptor and retinoid X receptor, which are involved in the development, regeneration, and maintenance of the nervous system. These findings highlight the importance of understanding the interplay between alcohol metabolism, the retinoic acid pathway and genetic factors in the development of fetal alcohol syndrome.
Collapse
Affiliation(s)
- Melina Vieiros
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Department de Cirurgia i Especialitats Mèdico-Quirúrgiques, Universitat de Barcelona, Barcelona, Spain
| | - Elisabet Navarro-Tapia
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain.
- Faculty of Health Sciences, Valencian International University, Valencia, Spain.
| | - Anna Ramos-Triguero
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Àgueda García-Meseguer
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Leopoldo Martínez
- IdiPAZ - Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
- Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Óscar García-Algar
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, BCNatal, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Grup de Recerca Infància i Entorn (GRIE), Institut d'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Biosanitary Research Institute, Valencian International University, Valencia, Spain.
| |
Collapse
|
11
|
McKay L, Petrelli B, Pind M, Reynolds JN, Wintle RF, Chudley AE, Drögemöller B, Fainsod A, Scherer SW, Hanlon-Dearman A, Hicks GG. Risk and Resilience Variants in the Retinoic Acid Metabolic and Developmental Pathways Associated with Risk of FASD Outcomes. Biomolecules 2024; 14:569. [PMID: 38785976 PMCID: PMC11117505 DOI: 10.3390/biom14050569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a common neurodevelopmental disorder that affects an estimated 2-5% of North Americans. FASD is induced by prenatal alcohol exposure (PAE) during pregnancy and while there is a clear genetic contribution, few genetic factors are currently identified or understood. In this study, using a candidate gene approach, we performed a genetic variant analysis of retinoic acid (RA) metabolic and developmental signaling pathway genes on whole exome sequencing data of 23 FASD-diagnosed individuals. We found risk and resilience alleles in ADH and ALDH genes known to normally be involved in alcohol detoxification at the expense of RA production, causing RA deficiency, following PAE. Risk and resilience variants were also identified in RA-regulated developmental pathway genes, especially in SHH and WNT pathways. Notably, we also identified significant variants in the causative genes of rare neurodevelopmental disorders sharing comorbidities with FASD, including STRA6 (Matthew-Wood), SOX9 (Campomelic Dysplasia), FDG1 (Aarskog), and 22q11.2 deletion syndrome (TBX1). Although this is a small exploratory study, the findings support PAE-induced RA deficiency as a major etiology underlying FASD and suggest risk and resilience variants may be suitable biomarkers to determine the risk of FASD outcomes following PAE.
Collapse
Affiliation(s)
- Leo McKay
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Berardino Petrelli
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Molly Pind
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - James N. Reynolds
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 2V7, Canada
| | - Richard F. Wintle
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Albert E. Chudley
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Britt Drögemöller
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Centre on Aging, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, P.O. Box 12271, Jerusalem 9112102, Israel
| | - Stephen W. Scherer
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, ON M5G 1L7, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Ana Hanlon-Dearman
- Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1S1, Canada
| | - Geoffrey G. Hicks
- Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
12
|
Nazzari S, Grumi S, Mambretti F, Villa M, Giorda R, Bordoni M, Pansarasa O, Borgatti R, Provenzi L. Sex-dimorphic pathways in the associations between maternal trait anxiety, infant BDNF methylation, and negative emotionality. Dev Psychopathol 2024; 36:908-918. [PMID: 36855816 DOI: 10.1017/s0954579423000172] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Maternal antenatal anxiety is an emerging risk factor for child emotional development. Both sex and epigenetic mechanisms, such as DNA methylation, may contribute to the embedding of maternal distress into emotional outcomes. Here, we investigated sex-dependent patterns in the association between antenatal maternal trait anxiety, methylation of the brain-derived neurotrophic factor gene (BDNF DNAm), and infant negative emotionality (NE). Mother-infant dyads (N = 276) were recruited at delivery. Maternal trait anxiety, as a marker of antenatal chronic stress exposure, was assessed soon after delivery using the Stait-Trait Anxiety Inventory (STAI-Y). Infants' BDNF DNAm at birth was assessed in 11 CpG sites in buccal cells whereas infants' NE was assessed at 3 (N = 225) and 6 months (N = 189) using the Infant Behavior Questionnaire-Revised (IBQ-R). Hierarchical linear analyses showed that higher maternal antenatal anxiety was associated with greater 6-month-olds' NE. Furthermore, maternal antenatal anxiety predicted greater infants' BDNF DNAm in five CpG sites in males but not in females. Higher methylation at these sites was associated with greater 3-to-6-month NE increase, independently of infants' sex. Maternal antenatal anxiety emerged as a risk factor for infant's NE. BDNF DNAm might mediate this effect in males. These results may inform the development of strategies to promote mothers and infants' emotional well-being.
Collapse
Affiliation(s)
- Sarah Nazzari
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Serena Grumi
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Fabiana Mambretti
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Marco Villa
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Roberto Giorda
- Molecular Biology Lab, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Matteo Bordoni
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Cellular Models and Neuroepigenetics Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Renato Borgatti
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| | - Livio Provenzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Developmental Psychobiology Lab, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
13
|
Derakhshan M, Kessler NJ, Hellenthal G, Silver MJ. Metastable epialleles in humans. Trends Genet 2024; 40:52-68. [PMID: 38000919 DOI: 10.1016/j.tig.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 11/26/2023]
Abstract
First identified in isogenic mice, metastable epialleles (MEs) are loci where the extent of DNA methylation (DNAm) is variable between individuals but correlates across tissues derived from different germ layers within a given individual. This property, termed systemic interindividual variation (SIV), is attributed to stochastic methylation establishment before germ layer differentiation. Evidence suggests that some putative human MEs are sensitive to environmental exposures in early development. In this review we introduce key concepts pertaining to human MEs, describe methods used to identify MEs in humans, and review their genomic features. We also highlight studies linking DNAm at putative human MEs to early environmental exposures and postnatal (including disease) phenotypes.
Collapse
Affiliation(s)
- Maria Derakhshan
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Noah J Kessler
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | - Matt J Silver
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul, The Gambia.
| |
Collapse
|
14
|
Jones JD, Martinez S, Gonzalez I, Odom GJ, Comer SD. No evidence of accelerated epigenetic aging among black heroin users: A case vs control analysis. ADDICTION NEUROSCIENCE 2023; 7:100096. [PMID: 37388854 PMCID: PMC10305791 DOI: 10.1016/j.addicn.2023.100096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
This study sought to assess the association between illicit opioid use and accelerated epigenetic aging (A.K.A. DNAm Age) among people of African ancestry who use heroin. DNA was obtained from participants with opioid use disorder (OUD) who confirmed heroin as their primary drug of choice. Clinical inventories of drug use included: the Addiction Severity Index (ASI) Drug-Composite Score (range: 0-1), and Drug Abuse Screening Test (DAST-10; range: 0-10). A control group of participants of African ancestry who did not use heroin was recruited and matched to heroin users on sex, age, socioeconomic level, and smoking status. Methylation data were assessed in an epigenetic clock to determined and compare Epigenetic Age to Chronological Age (i.e., age acceleration or deceleration). Data were obtained from 32 controls [mean age 36.3 (±7.5) years] and 64 heroin users [mean age 48.1 (±6.6) years]. The experimental group used heroin for an average of 18.1 (±10.6) years, reported use of 6.4 (±6.1) bags of heroin/day, with a mean DAST-10 score of 7.0 (±2.6) and ASI Score of 0.33 (±0.19). Mean age acceleration for heroin users [+0.56 (± 9.5) years] was significantly (p< 0.05) lower than controls [+5.19 (± 9.1) years]. This study did not find evidence that heroin use causes epigenetic age acceleration.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Department of Psychiatry, Division on Substance Use Disorders, New York State Psychiatric Institute, and Columbia University Irving Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - Suky Martinez
- Department of Psychiatry, Division on Substance Use Disorders, New York State Psychiatric Institute, and Columbia University Irving Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - Ingrid Gonzalez
- Department of Biostatistics, Robert Stempel College of Public Health, Florida International University, 1200 SW 8th St, Miami, FL 33174, USA
| | - Gabriel J. Odom
- Department of Biostatistics, Robert Stempel College of Public Health, Florida International University, 1200 SW 8th St, Miami, FL 33174, USA
| | - Sandra D. Comer
- Department of Psychiatry, Division on Substance Use Disorders, New York State Psychiatric Institute, and Columbia University Irving Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
15
|
Chaudoin TR, Bonasera SJ, Dunaevsky A, Padmashri R. Exploring behavioral phenotypes in a mouse model of fetal alcohol spectrum disorders. Dev Neurobiol 2023; 83:184-204. [PMID: 37433012 PMCID: PMC10546278 DOI: 10.1002/dneu.22922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/18/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Fetal alcohol spectrum disorders are one of the leading causes of developmental abnormalities worldwide. Maternal consumption of alcohol during pregnancy leads to a diverse range of cognitive and neurobehavioral deficits. Although moderate-to-heavy levels of prenatal alcohol exposure (PAE) have been associated with adverse offspring outcomes, there is limited data on the consequences of chronic low-level PAE. Here, we use a model of maternal voluntary alcohol consumption throughout gestation in a mouse model to investigate the effects of PAE on behavioral phenotypes during late adolescence and early adulthood in male and female offspring. Body composition was measured by dual-energy X-ray absorptiometry. Baseline behaviors, including feeding, drinking, and movement, were examined by performing home cage monitoring studies. The impact of PAE on motor function, motor skill learning, hyperactivity, acoustic reactivity, and sensorimotor gating was investigated by performing a battery of behavioral tests. PAE was found to be associated with altered body composition. No differences in overall movement, food, or water consumption were observed between control and PAE mice. Although PAE offspring of both sexes exhibited deficits in motor skill learning, no differences were observed in basic motor skills such as grip strength and motor coordination. PAE females exhibited a hyperactive phenotype in a novel environment. PAE mice exhibited increased reactivity to acoustic stimuli, and PAE females showed disrupted short-term habituation. Sensorimotor gating was not altered in PAE mice. Collectively, our data show that chronic low-level exposure to alcohol in utero results in behavioral impairments.
Collapse
Affiliation(s)
- Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
16
|
Petrelli B, Oztürk A, Pind M, Ayele H, Fainsod A, Hicks GG. Genetically programmed retinoic acid deficiency during gastrulation phenocopies most known developmental defects due to acute prenatal alcohol exposure in FASD. Front Cell Dev Biol 2023; 11:1208279. [PMID: 37397253 PMCID: PMC10311642 DOI: 10.3389/fcell.2023.1208279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) arises from maternal consumption of alcohol during pregnancy affecting 2%-5% of the Western population. In Xenopus laevis studies, we showed that alcohol exposure during early gastrulation reduces retinoic acid (RA) levels at this critical embryonic stage inducing craniofacial malformations associated with Fetal Alcohol Syndrome. A genetic mouse model that induces a transient RA deficiency in the node during gastrulation is described. These mice recapitulate the phenotypes characteristic of prenatal alcohol exposure (PAE) suggesting a molecular etiology for the craniofacial malformations seen in children with FASD. Gsc +/Cyp26A1 mouse embryos have a reduced RA domain and expression in the developing frontonasal prominence region and delayed HoxA1 and HoxB1 expression at E8.5. These embryos also show aberrant neurofilament expression during cranial nerve formation at E10.5 and have significant FASD sentinel-like craniofacial phenotypes at E18.5. Gsc +/Cyp26A1 mice develop severe maxillary malocclusions in adulthood. Phenocopying the PAE-induced developmental malformations with a genetic model inducing RA deficiency during early gastrulation strongly supports the alcohol/vitamin A competition model as a major molecular etiology for the neurodevelopmental defects and craniofacial malformations seen in children with FASD.
Collapse
Affiliation(s)
- B. Petrelli
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - A. Oztürk
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - M. Pind
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - H. Ayele
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - A. Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - G. G. Hicks
- Department of Biochemistry and Medical Genetics, Regenerative Medicine Program, Faculty of Medicine, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
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: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [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
|
18
|
Breton‐Larrivée M, Elder E, Legault L, 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 PMCID: PMC11977608 DOI: 10.1096/fj.202201564r] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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 CenterMontrealCanada
- Department of Biochemistry and Molecular MedicineUniversité de MontréalMontrealCanada
| | - Elizabeth Elder
- Centre Hospitalier Universitaire Sainte‐Justine Research CenterMontrealCanada
- Department of Biochemistry and Molecular MedicineUniversité de MontréalMontrealCanada
| | - Lisa‐Marie Legault
- Centre Hospitalier Universitaire Sainte‐Justine Research CenterMontrealCanada
- Department of Biochemistry and Molecular MedicineUniversité de MontréalMontrealCanada
| | - Alexandra Langford‐Avelar
- Centre Hospitalier Universitaire Sainte‐Justine Research CenterMontrealCanada
- Department of Biochemistry and Molecular MedicineUniversité de MontréalMontrealCanada
| | - Amanda J. MacFarlane
- Agriculture, Food, and Nutrition Evidence CenterTexas A&M UniversityTexasFort WorthUSA
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Serge McGraw
- Centre Hospitalier Universitaire Sainte‐Justine Research CenterMontrealCanada
- Department of Biochemistry and Molecular MedicineUniversité de MontréalMontrealCanada
- Department of Obstetrics and GynecologyUniversité de MontréalMontrealCanada
| |
Collapse
|
19
|
Santilli F, Boskovic A. Mechanisms of transgenerational epigenetic inheritance: lessons from animal model organisms. Curr Opin Genet Dev 2023; 79:102024. [PMID: 36893483 DOI: 10.1016/j.gde.2023.102024] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/24/2022] [Accepted: 01/26/2023] [Indexed: 03/09/2023]
Abstract
Epigenetic inheritance is a phenomenon whereby stochastic or signal-induced changes to parental germline epigenome modulate phenotypic output in one or more subsequent generations, independently of mutations in the genomic DNA. While the number of reported epigenetic inheritance phenomena across phyla is exponentially growing, much remains to be elucidated about their mechanistic underpinnings, and their significance for organismal homeostasis and adaptation. Here, we review the most recent epigenetic inheritance examples in animal models, outlining molecular details behind environmental sensing by the germline, and the functional relationships connecting epigenetic mechanisms and phenotypic traits after fertilization. We touch upon the experimental challenges associated with studying the scope of environmental input on phenotypic outcomes between generations. Finally, we discuss the implications of mechanistic findings from model organisms for the emergent examples of parental effects in human populations.
Collapse
Affiliation(s)
- Flavio Santilli
- European Molecular Biology Laboratory Epigenetics & Neurobiology Unit, Monterotondo, RM, Italy. https://twitter.com/@santilli_flavio
| | - Ana Boskovic
- European Molecular Biology Laboratory Epigenetics & Neurobiology Unit, Monterotondo, RM, Italy.
| |
Collapse
|
20
|
Colwell ML, Townsel C, Petroff RL, Goodrich JM, Dolinoy DC. Epigenetics and the Exposome: DNA Methylation as a Proxy for Health Impacts of Prenatal Environmental Exposures. EXPOSOME 2023; 3:osad001. [PMID: 37333730 PMCID: PMC10275510 DOI: 10.1093/exposome/osad001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The accumulation of every day exposures can impact health across the life course, but our understanding of such exposures is impeded by our ability to delineate the relationship between an individual's early life exposome and later life health effects. Measuring the exposome is challenging. Exposure assessed at a given time point captures a snapshot of the exposome but does not represent the full spectrum of exposures across the life course. In addition, the assessment of early life exposures and their effects is often further challenged by lack of relevant samples and the time gap between exposures and related health outcomes in later life. Epigenetics, specifically DNA methylation, has the potential to overcome these barriers as environmental epigenetic perturbances can be retained through time. In this review, we describe how DNA methylation can be framed in the world of the exposome. We offer three compelling examples of common environmental exposures, including cigarette smoke, the endocrine active compound bisphenol A (BPA), and the metal lead (Pb), to illustrate the application of DNA methylation as a proxy to measure the exposome. We discuss areas for future explorations and current limitations of this approach. Epigenetic profiling is a promising and rapidly developing tool and field of study, offering us a unique and powerful way to assess the early life exposome and its effects across different life stages.
Collapse
Affiliation(s)
- Mathia L. Colwell
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Courtney Townsel
- Department of Obstetrics and Gynecology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Rebekah L. Petroff
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Cheng X, Li X, Liu Y, Ma Y, Zhang R, Zhang Y, Fan C, Qu L, Ning Z. DNA methylome and transcriptome identified Key genes and pathways involved in Speckled Eggshell formation in aged laying hens. BMC Genomics 2023; 24:31. [PMID: 36658492 PMCID: PMC9854222 DOI: 10.1186/s12864-022-09100-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 12/26/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The quality of poultry eggshells is closely related to the profitability of egg production. Eggshell speckles reflect an important quality trait that influences egg appearance and customer preference. However, the mechanism of speckle formation remains poorly understood. In this study, we systematically compared serum immune and antioxidant indices of hens laying speckled and normal eggs. Transcriptome and methylome analyses were used to elucidate the mechanism of eggshell speckle formation. RESULTS The results showed that seven differentially expressed genes (DEGs) were identified between the normal and speckle groups. Gene set enrichment analysis (GSEA) revealed that the expressed genes were mainly enriched in the calcium signaling pathway, focal adhesion, and MAPK signaling pathway. Additionally, 282 differentially methylated genes (DMGs) were detected, of which 15 genes were associated with aging, including ARNTL, CAV1, and GCLC. Pathway analysis showed that the DMGs were associated with T cell-mediated immunity, response to oxidative stress, and cellular response to DNA damage stimulus. Integrative analysis of transcriptome and DNA methylation data identified BFSP2 as the only overlapping gene, which was expressed at low levels and hypomethylated in the speckle group. CONCLUSIONS Overall, these results indicate that aging- and immune-related genes and pathways play a crucial role in the formation of speckled eggshells, providing useful information for improving eggshell quality.
Collapse
Affiliation(s)
- Xue Cheng
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Xinghua Li
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Yuchen Liu
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Ying Ma
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Ruiqi Zhang
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Yalan Zhang
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Cuidie Fan
- Rongde Breeding Company Limited, Hebei, 053000 China
| | - Lujiang Qu
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| | - Zhonghua Ning
- grid.22935.3f0000 0004 0530 8290National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193 China
| |
Collapse
|
22
|
Epigenetics in fetal alcohol spectrum disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:211-239. [PMID: 37019593 DOI: 10.1016/bs.pmbts.2023.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During pregnancy, alcohol abuse and its detrimental effects on developing offspring are major public health, economic and social challenges. The prominent characteristic attributes of alcohol (ethanol) abuse during pregnancy in humans are neurobehavioral impairments in offspring due to damage to the central nervous system (CNS), causing structural and behavioral impairments that are together named fetal alcohol spectrum disorder (FASD). Development-specific alcohol exposure paradigms were established to recapitulate the human FASD phenotypes and establish the underlying mechanisms. These animal studies have offered some critical molecular and cellular underpinnings likely to account for the neurobehavioral impairments associated with prenatal ethanol exposure. Although the pathogenesis of FASD remains unclear, emerging literature proposes that the various genomic and epigenetic components that cause the imbalance in gene expression can significantly contribute to the development of this disease. These studies acknowledged numerous immediate and enduring epigenetic modifications, such as methylation of DNA, post-translational modifications (PTMs) of histone proteins, and regulatory networks related to RNA, using many molecular approaches. Methylated DNA profiles, PTMs of histone proteins, and RNA-regulated expression of genes are essential for synaptic and cognitive behavior. Thus, offering a solution to many neuronal and behavioral impairments reported in FASD. In the current chapter, we review the recent advances in different epigenetic modifications that cause the pathogenesis of FASD. The information discussed can help better explain the pathogenesis of FASD and thereby might provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
|
23
|
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: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
24
|
Sestario CS, de Fátima Mestre V, Nantes Martins CC, Campos Zeffa A, Frítola M, Sparça Salles MJ. Congenital anomalies and spontaneous abortion in mice resulting from the use of escitalopram. Reprod Fertil Dev 2022; 34:1099-1106. [PMID: 36219877 DOI: 10.1071/rd22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/12/2022] [Indexed: 06/16/2023] Open
Abstract
CONTEXT Escitalopram (ESC) use during pregnancy has not been associated with teratogenic effects in fetuses. AIMS To investigate whether ESC administered during pregnancy in mice induces maternal toxicity and teratogenicity in offspring. METHODS Treated mice groups G1 and control G0 (n =15 per group). Administration of ESC (G1) and saline solution (G0) during pregnancy and euthanasia on the 18thday. Pregnant female mice were treated with ESC (20mg/kg, via gavage) or saline solution (control group) from the 5th to the 17thday of gestation, when implantation was consolidated. During intraembryonic development until the day before delivery, the drug had an influence on the development of alterations from its maintenance in the uterine environment and its development to the disturbance causing skeletal or visceral malformations. KEY RESULTS The intrauterine development parameters that were altered by ESC treatment were: number of resorptions (G0: [0.93±0.24]); G1: [3.33±0.51]), post-implantation loss (G0: [3.95±1.34], G1: [13.75±3.62]) and reduced fetal viability: [97.30±1.00]; G1: [81.09±6.22]). Regarding fetal formation, the treated group had visceral malformations with a significant frequency: cleft palate (G0: [1.0%], G1: [11.86%]) and reduced kidneys (G0: [0%]; G1: [10.17%]). Regarding skeletal malformations, a higher frequency was observed in the following parameters: incomplete supraoccipital ossification (G0: [0%], G1: [15.25]), absence of ribs (G0: [0%], G1 (G0: [0%], G1 [15.25%]) and absence of one or more of the foot phalanges (G0: [1.0%]; 64%]). CONCLUSION Results indicate that ESC is an embryotoxic and teratogenic drug. IMPLICATIONS Until further studies are performed, greater caution is necessary in prescribing the drug to pregnant women.
Collapse
Affiliation(s)
- Camila Salvador Sestario
- Programa de Pós-graduação em Ciências da Saúde, Universidade Estadual de Londrina, Av. Robert Koch, 60, CEP: 86038-350 Londrina, Brasil; and Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, CEP: 86057-970 Londrina, Brasil
| | - Viviane de Fátima Mestre
- Programa de Pós-graduação em Ciências da Saúde, Universidade Estadual de Londrina, Av. Robert Koch, 60, CEP: 86038-350 Londrina, Brasil; and Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, CEP: 86057-970 Londrina, Brasil
| | - Caio Cezar Nantes Martins
- Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, CEP: 86057-970 Londrina, Brasil
| | - Aline Campos Zeffa
- Programa de Pós-graduação em Ciências da Saúde, Universidade Estadual de Londrina, Av. Robert Koch, 60, CEP: 86038-350 Londrina, Brasil
| | - Márjori Frítola
- Programa de Pós-graduação em Ciências da Saúde, Universidade Estadual de Londrina, Av. Robert Koch, 60, CEP: 86038-350 Londrina, Brasil; and Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, CEP: 86057-970 Londrina, Brasil
| | - Maria José Sparça Salles
- Departamento de Biologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, Km 380, CEP: 86057-970 Londrina, Brasil
| |
Collapse
|
25
|
Ormazabal V, Nair S, Carrión F, Mcintyre HD, Salomon C. The link between gestational diabetes and cardiovascular diseases: potential role of extracellular vesicles. Cardiovasc Diabetol 2022; 21:174. [PMID: 36057662 PMCID: PMC9441052 DOI: 10.1186/s12933-022-01597-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Extracellular vesicles are critical mediators of cell communication. They encapsulate a variety of molecular cargo such as proteins, lipids, and nucleic acids including miRNAs, lncRNAs, circular RNAs, and mRNAs, and through transfer of these molecular signals can alter the metabolic phenotype in recipient cells. Emerging studies show the important role of extracellular vesicle signaling in the development and progression of cardiovascular diseases and associated risk factors such as type 2 diabetes and obesity. Gestational diabetes mellitus (GDM) is hyperglycemia that develops during pregnancy and increases the future risk of developing obesity, impaired glucose metabolism, and cardiovascular disease in both the mother and infant. Available evidence shows that changes in maternal metabolism and exposure to the hyperglycemic intrauterine environment can reprogram the fetal genome, leaving metabolic imprints that define life-long health and disease susceptibility. Understanding the factors that contribute to the increased susceptibility to metabolic disorders of children born to GDM mothers is critical for implementation of preventive strategies in GDM. In this review, we discuss the current literature on the fetal programming of cardiovascular diseases in GDM and the impact of extracellular vesicle (EV) signaling in epigenetic programming in cardiovascular disease, to determine the potential link between EV signaling in GDM and the development of cardiovascular disease in infants.
Collapse
Affiliation(s)
- Valeska Ormazabal
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia.,Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile
| | - Soumyalekshmi Nair
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - H David Mcintyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine + Biomedical Sciences, The University of Queensland, Building 71/918, Herston, QLD, 4029, Australia. .,Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
26
|
A hypothesis: Retrotransposons as a relay of epigenetic marks in intergenerational epigenetic inheritance. Gene 2022; 817:146229. [PMID: 35063571 DOI: 10.1016/j.gene.2022.146229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/13/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Epigenetic marks in gametes, which both respond to the parental environmental factors and shape offspring phenotypes, are usually positioned to mediate intergenerational or transgenerational epigenetic inheritance. Nonetheless, the mechanisms through which gametic epigenetic signatures encode parental acquired phenotypes, and further initiate a cascade of molecular events to affect offspring phenotypes during early embryonic development, remain unclear. Retrotransposons are mobile DNA elements that could resist to genomic epigenetic reprogramming at specific loci and rewire the core regulatory networks of embryogenesis. Increasing evidences show that retrotransposons in the embryonic genome could interact with gametic epigenetic marks, which provides a tentative possibility that retrotransposons may serve as a relay of gametic epigenetic marks to transmit parental acquired traits. Here, we summarize the recent progress in exploring the crosstalk between gametic epigenetic marks and retrotransposons, and the regulation of gene expression and early embryonic development by retrotransposons. Accordingly, deciphering the mystery of interactions between gametic epigenetic marks and retrotransposons during early embryonic development will provide valuable insights into the intergenerational or transgenerational transmission of acquired traits.
Collapse
|
27
|
Smith SM, Pjetri E, Friday WB, Presswood BH, Ricketts DK, Walter KR, Mooney SM. Aging-Related Behavioral, Adiposity, and Glucose Impairments and Their Association following Prenatal Alcohol Exposure in the C57BL/6J Mouse. Nutrients 2022; 14:1438. [PMID: 35406051 PMCID: PMC9002573 DOI: 10.3390/nu14071438] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
People that experience prenatal alcohol exposure (PAE) may have behavioral and metabolic impairments, and it is unclear whether these remain stable or change with age. We assessed behavioral and metabolic endpoints across the lifespan in a mouse model of fetal alcohol spectrum disorder (FASD). Pregnant C57BL/6J mice received alcohol (ALC; 3 g/kg) or maltose-dextrin (control, CON) daily from embryonic day 8.5 to 17.5. Offspring were tested on accelerating rotarod, Y-maze, novel object recognition, and fear conditioning at 6 weeks and 10 and 17 months; females were also tested at 24 months. Body composition, fasting glucose, and glucose clearance were assessed at 18 months. Female but not male ALC mice had greater adiposity than age-matched CON from 7 months onward. At 18 months, male but not female ALC mice had reduced glucose clearance and ALC mice were more likely to have elevated fasting glucose. In the rotarod training session, ALC females performed worse than CON. In the Y-maze, significant exposure-age interactions affected ALC performance in both sexes versus age-match CON. For fear conditioning, all animals acquired the task and froze more at older ages. In both the context and cued tasks, there were exposure-age interactions and ALC animals frozen less than CON at 10 months. Correlation analysis revealed that fasting glucose and glucose clearance correlated with % of body fat in ALC but not in CON mice. Additionally, glucose intolerance and % body fat negatively correlated with performance in the rotarod, context learning, and novel object recognition tasks in ALC but not CON mice. All mice exhibit worsening of behavioral performance as they age, and PAE did not further exacerbate this. ALC but not CON mice displayed adiposity and glucose intolerance that correlate with their cognitive impairments, suggesting that these may be mechanistically related in PAE. Findings emphasize that FASD should be considered a whole-body disorder.
Collapse
Affiliation(s)
- Susan M. Smith
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Eneda Pjetri
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
| | - Walter B. Friday
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
| | - Brandon H. Presswood
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
| | - Dane K. Ricketts
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
| | - Kathleen R. Walter
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
| | - Sandra M. Mooney
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (E.P.); (W.B.F.); (B.H.P.); (D.K.R.); (K.R.W.)
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| |
Collapse
|
28
|
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: 6] [Impact Index Per Article: 2.0] [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
|
29
|
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: 9] [Impact Index Per Article: 3.0] [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
|
30
|
Gutherz OR, Deyssenroth M, Li Q, Hao K, Jacobson JL, Chen J, Jacobson SW, Carter RC. Potential roles of imprinted genes in the teratogenic effects of alcohol on the placenta, somatic growth, and the developing brain. Exp Neurol 2021; 347:113919. [PMID: 34752786 DOI: 10.1016/j.expneurol.2021.113919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
Despite several decades of research and prevention efforts, fetal alcohol spectrum disorders (FASD) remain the most common preventable cause of neurodevelopmental disabilities worldwide. Animal and human studies have implicated fetal alcohol-induced alterations in epigenetic programming as a chief mechanism in FASD. Several studies have demonstrated fetal alcohol-related alterations in methylation and expression of imprinted genes in placental, brain, and embryonic tissue. Imprinted genes are epigenetically regulated in a parent-of-origin-specific manner, in which only the maternal or paternal allele is expressed, and the other allele is silenced. The chief functions of imprinted genes are in placental development, somatic growth, and neurobehavior-three domains characteristically affected in FASD. In this review, we summarize the growing body of literature characterizing prenatal alcohol-related alterations in imprinted gene methylation and/or expression and discuss potential mechanistic roles for these alterations in the teratogenic effects of prenatal alcohol exposure. Future research is needed to examine potential physiologic mechanisms by which alterations in imprinted genes disrupt development in FASD, which may, in turn, elucidate novel targets for intervention. Furthermore, mechanistic alterations in imprinted gene expression and/or methylation in FASD may inform screening assays that identify individuals with FASD neurobehavioral deficits who may benefit from early interventions.
Collapse
Affiliation(s)
- Olivia R Gutherz
- Institute of Human Nutrition, Columbia University Medical Center, United States of America
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, United States of America
| | - Qian Li
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Ke Hao
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Joseph L Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, United States of America; Department of Human Biology, University of Cape Town Faculty of Health Sciences, South Africa
| | - Jia Chen
- Department of Environmental Medicine & Public Health, Icahn School of Medicine at Mount Sinai, United States of America
| | - Sandra W Jacobson
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, United States of America; Department of Human Biology, University of Cape Town Faculty of Health Sciences, South Africa
| | - R Colin Carter
- Institute of Human Nutrition, Columbia University Medical Center, United States of America; Departments of Emergency Medicine and Pediatrics, Columbia University Medical Center, United States of America.
| |
Collapse
|
31
|
Liu Y, Xu Q, Kang X, Wang K, Wang J, Feng D, Bai Y, Fang M. Dynamic changes of genomic methylation profiles at different growth stages in Chinese Tan sheep. J Anim Sci Biotechnol 2021; 12:118. [PMID: 34727982 PMCID: PMC8561971 DOI: 10.1186/s40104-021-00632-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 08/31/2021] [Indexed: 01/02/2023] Open
Abstract
Background Tan sheep, an important local sheep breed in China, is famous for their fur quality. One-month-old Tan sheep have white, curly hair with beautiful flower spikes, commonly known as “nine bends”, which has high economic value. However, the “nine bends” characteristic gradually disappears with age; consequently, the economic value of the Tan sheep decreases. Age-related changes in DNA methylation have been reported and may be responsible for age-induced changes in gene expression. Until now, no genome-wide surveys have been conducted to identify potential DNA methylation sites involved in different sheep growth stages. In this study we investigated the dynamic changes of genome-wide DNA methylation profiles in Tan sheep using DNA from skin and deep whole-genome bisulfite sequencing, and compared the DNA methylation levels at three different growth stages: 1, 24, and 48 months old (mon1, mon24, and mon48, respectively). Results In this study, 11 skin samples from three growth stages (four for mon1, four for mon24, and three for mon48) were used for DNA methylation analysis and gene expression profiling. There were 52, 288 and 236 differentially methylated genes (DMGs) identified between mon1 and mon24, mon1 and mon48, and mon24 and mon48, respectively. Of the differentially methylated regions, 1.11%, 7.61%, and 7.65% were in the promoter in mon1 vs. mon24, mon24 vs. mon48, and mon1 vs. mon48, respectively. DMGs were enriched in the MAPK and WNT signaling pathways, which are related to age growth and hair follicle morphogenesis processes. There were 51 DMGs associated with age growth and curly fleece formation. Four DMGs between mon1 and mon48 (KRT71, CD44, ROR2 and ZDHHC13) were further validated by bisulfite sequencing. Conclusions This study revealed dynamic changes in the genomic methylation profiles of mon1, mon24, and mon48 sheep, and the percentages of methylated cytosines were 3.38%, 2.85% and 4.17%, respectively. Of the DMGs, KRT71 and CD44 were highly methylated in mon1, and ROR2 and ZDHHC13 were highly methylated in mon48. These findings provide foundational information that may be used to develop strategies for potentially retaining the lamb fur and thus improving the economic value of Tan sheep. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00632-9.
Collapse
Affiliation(s)
- Yufang Liu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Rd, Beijing, 100193, People's Republic of China.,College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, 056021, People's Republic of China
| | - Qiao Xu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Rd, Beijing, 100193, People's Republic of China.,Biotechnology Institute, Nanchang Normal University, Nanchang, 330029, People's Republic of China
| | - Xiaolong Kang
- College of Agriculture, Ningxia University, Yinchuan, 750021, People's Republic of China
| | - Kejun Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, People's Republic of China
| | - Jve Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Rd, Beijing, 100193, People's Republic of China
| | - Dengzhen Feng
- Biotechnology Institute, Nanchang Normal University, Nanchang, 330029, People's Republic of China
| | - Ying Bai
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, 056021, People's Republic of China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Rd, Beijing, 100193, People's Republic of China. .,Beijing Key Laboratory for Animal Genetic Improvement, Beijing, 100193, People's Republic of China.
| |
Collapse
|
32
|
Chen ACH, Huang W, Fong SW, Chan C, Lee KC, Yeung WSB, Lee YL. Hyperglycemia Altered DNA Methylation Status and Impaired Pancreatic Differentiation from Embryonic Stem Cells. Int J Mol Sci 2021; 22:10729. [PMID: 34639069 PMCID: PMC8509790 DOI: 10.3390/ijms221910729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 11/16/2022] Open
Abstract
The prevalence of type 2 diabetes (T2D) is rapidly increasing across the globe. Fetal exposure to maternal diabetes was correlated with higher prevalence of impaired glucose tolerance and T2D later in life. Previous studies showed aberrant DNA methylation patterns in pancreas of T2D patients. However, the underlying mechanisms remained largely unknown. We utilized human embryonic stem cells (hESC) as the in vitro model for studying the effects of hyperglycemia on DNA methylome and early pancreatic differentiation. Culture in hyperglycemic conditions disturbed the pancreatic lineage potential of hESC, leading to the downregulation of expression of pancreatic markers PDX1, NKX6-1 and NKX6-2 after in vitro differentiation. Genome-wide DNA methylome profiling revealed over 2000 differentially methylated CpG sites in hESC cultured in hyperglycemic condition when compared with those in control glucose condition. Gene ontology analysis also revealed that the hypermethylated genes were enriched in cell fate commitment. Among them, NKX6-2 was validated and its hypermethylation status was maintained upon differentiation into pancreatic progenitor cells. We also established mouse ESC lines at both physiological glucose level (PG-mESC) and conventional hyperglycemia glucose level (HG-mESC). Concordantly, DNA methylome analysis revealed the enrichment of hypermethylated genes related to cell differentiation in HG-mESC, including Nkx6-1. Our results suggested that hyperglycemia dysregulated the epigenome at early fetal development, possibly leading to impaired pancreatic development.
Collapse
Affiliation(s)
- Andy Chun Hang Chen
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Wen Huang
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Chris Chan
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Kai Chuen Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - William Shu Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| | - Yin Lau Lee
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong, Shenzhen Hospital, Shenzhen 518000, China;
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong; (W.H.); (S.W.F.); (C.C.); (K.C.L.)
| |
Collapse
|
33
|
Burton NO, Greer EL. Multigenerational epigenetic inheritance: Transmitting information across generations. Semin Cell Dev Biol 2021; 127:121-132. [PMID: 34426067 DOI: 10.1016/j.semcdb.2021.08.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/07/2023]
Abstract
Inherited epigenetic information has been observed to regulate a variety of complex organismal phenotypes across diverse taxa of life. This continually expanding body of literature suggests that epigenetic inheritance plays a significant, and potentially fundamental, role in inheritance. Despite the important role these types of effects play in biology, the molecular mediators of this non-genetic transmission of information are just now beginning to be deciphered. Here we provide an intellectual framework for interpreting these findings and how they can interact with each other. We also define the different types of mechanisms that have been found to mediate epigenetic inheritance and to regulate whether epigenetic information persists for one or many generations. The field of epigenetic inheritance is entering an exciting phase, in which we are beginning to understand the mechanisms by which non-genetic information is transmitted to, and deciphered by, subsequent generations to maintain essential environmental information without permanently altering the genetic code. A more complete understanding of how and when epigenetic inheritance occurs will advance our understanding of numerous different aspects of biology ranging from how organisms cope with changing environments to human pathologies influenced by a parent's environment.
Collapse
Affiliation(s)
- Nicholas O Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| | - Eric L Greer
- Division of Newborn Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Harvard Medical School Initiative for RNA Medicine, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Bertozzi TM, Becker JL, Blake GET, Bansal A, Nguyen DK, Fernandez-Twinn DS, Ozanne SE, Bartolomei MS, Simmons RA, Watson ED, Ferguson-Smith AC. Variably methylated retrotransposons are refractory to a range of environmental perturbations. Nat Genet 2021; 53:1233-1242. [PMID: 34326545 PMCID: PMC7611517 DOI: 10.1038/s41588-021-00898-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/18/2021] [Indexed: 12/27/2022]
Abstract
The agouti viable yellow (Avy) allele is an insertional mutation in the mouse genome caused by a variably methylated intracisternal A particle (VM-IAP) retrotransposon. Avy expressivity is sensitive to a range of early-life chemical exposures and nutritional interventions, suggesting that environmental perturbations can have long-lasting effects on the methylome. However, the extent to which VM-IAP elements are environmentally labile with phenotypic implications is unknown. Using a recently identified repertoire of VM-IAPs, we assessed the epigenetic effects of different environmental contexts. A longitudinal aging analysis indicated that VM-IAPs are stable across the murine lifespan, with only small increases in DNA methylation detected for a subset of loci. No significant effects were observed after maternal exposure to the endocrine disruptor bisphenol A, an obesogenic diet or methyl donor supplementation. A genetic mouse model of abnormal folate metabolism exhibited shifted VM-IAP methylation levels and altered VM-IAP-associated gene expression, yet these effects are likely largely driven by differential targeting by polymorphic KRAB zinc finger proteins. We conclude that epigenetic variability at retrotransposons is not predictive of environmental susceptibility.
Collapse
Affiliation(s)
| | | | - Georgina E T Blake
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Amita Bansal
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Australian National University Medical School, John Curtin School of Medical Research, College of Health and Medicine, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Duy K Nguyen
- Department of Cell and Developmental Biology, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust Medical Research Council Institute of Metabolic Science, Cambridge, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust Medical Research Council Institute of Metabolic Science, Cambridge, UK
| | - Marisa S Bartolomei
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Erica D Watson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
35
|
|
36
|
Wallén E, Auvinen P, Kaminen-Ahola N. The Effects of Early Prenatal Alcohol Exposure on Epigenome and Embryonic Development. Genes (Basel) 2021; 12:genes12071095. [PMID: 34356111 PMCID: PMC8303887 DOI: 10.3390/genes12071095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure is one of the most significant causes of developmental disability in the Western world. Maternal alcohol consumption during pregnancy leads to an increased risk of neurological deficits and developmental abnormalities in the fetus. Over the past decade, several human and animal studies have demonstrated that alcohol causes alterations in epigenetic marks, including DNA methylation, histone modifications, and non-coding RNAs. There is an increasing amount of evidence that early pregnancy is a sensitive period for environmental-induced epigenetic changes. It is a dynamic period of epigenetic reprogramming, cell divisions, and DNA replication and, therefore, a particularly interesting period to study the molecular changes caused by alcohol exposure as well as the etiology of alcohol-induced developmental disorders. This article will review the current knowledge about the in vivo and in vitro effects of alcohol exposure on the epigenome, gene regulation, and the phenotype during the first weeks of pregnancy.
Collapse
|
37
|
Prenatal ethanol exposure increases maternal bile acids through placental transport pathway. Toxicology 2021; 458:152848. [PMID: 34217791 DOI: 10.1016/j.tox.2021.152848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022]
Abstract
High maternal serum bile acid level is common and sometimes harmful to the gravida. This study aimed to confirm the bile acid phenotypic change caused by prenatal ethanol exposure (PEE) and elucidate its placental mechanism. Pregnant Wistar rats were administered intragastrically with ethanol 4 g/kg⋅d from gestational day 9-20. Total bile acids (TBA) were detected in maternal, fetal serum and placental tissues, increasing significantly in the serum but no significant change in the placental tissues. Meta-analysis was performed and verified the efficacy of the PEE-induced model based on published data from several relevant studies. Mining of microarray data from human and rat placental sources identified the involvement of bile acid metabolism and its significant genes, which were verified by RT-qPCR and western blotting on tissues and treated BeWo cells with the administration of FXR/PXR siRNAs or FXR/PXR agonists. Our examination, consistent with microarray data and wet experiments, showed that organic anion transporter polypeptide-related protein 2B1 (Oatp2b1), multidrug resistance-associated proteins 3 (Mrp3) and breast cancer resistance protein (Bcrp) expression were increased, while nuclear receptor farnesoid X receptor (Fxr) was decreased but pregnane X receptor (Pxr) was increased. Furthermore, the interventional experiments confirmed that FXR regulated Bcrp while PXR regulated Oatp2b1 and Mrp3. In summary, PEE could induce high bile acid level in maternal serum and its mechanism is associated with the high expression of BCRP/MRP3/OATP2B1 in the placenta through up-regulating PXR and down-regulating FXR, thereby leading to an excessive bile acid transport to maternal blood via the placenta. Our study provides a novel perspective in terms of placenta, explaining the increased maternal blood bile acids under the toxicity of PEE.
Collapse
|
38
|
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: 0.8] [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
|
39
|
Oyaci Y, Aytac HM, Pasin O, Cetinay Aydin P, Pehlivan S. Detection of altered methylation of MB-COMT promotor and DRD2 gene in cannabinoid or synthetic cannabinoid use disorder regarding gene variants and clinical parameters. J Addict Dis 2021; 39:526-536. [PMID: 33781176 DOI: 10.1080/10550887.2021.1906618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study aims to investigate the association between cannabinoid use disorder (CUD) or synthetic cannabinoid use disorder (SCUD) and methylation status of MB-COMT (membrane-bound catechol-O-methyltransferase) promotor or DRD2 gene considering gene variants and clinical parameters. Based on the DSM-5 criteria, 218 CUD/SCUD patients' diagnoses were confirmed with a positive urine test, and a control group consisting of 102 participants without substance use disorders was included. Methylation-specific PCR was used to identify the methylation of the MB-COMT promotor and DRD2 gene. DRD2-141C Ins/Del and COMT Val158Met gene variants were evaluated by using PCR-RFLP. When the DRD2 and MB-COMT promoter methylation of CUD/SCUD patients were compared with the control group, there was a significant difference between the MB-COMT promoter methylation status of the two groups. When comparing DRD2 gene methylation due to clinical parameters and DRD2 genotype distribution in patients, the methylation status was significantly different between the groups due to the family history. Again, comparing the MB-COMT promotor methylation due to the COMT Val158Met genotype distribution and clinical parameters in patients, the MB-COMT promoter methylation status was significantly different between the groups due to the presence of alcohol usage. In summary, whereas the MB-COMT promoter methylation may be associated with the CUD/SCUD, the methylation of the DRD2 gene was not related to CUD/SCUD.
Collapse
Affiliation(s)
- Yasemin Oyaci
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hasan Mervan Aytac
- Department of Psychiatry, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Ozge Pasin
- Department of Biostatistics, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Pinar Cetinay Aydin
- Department of Psychiatry, University of Health Sciences, Psychiatry Clinic, Bakirkoy Research and Training Hospital for Psychiatry, Neurology and Neurosurgery, Istanbul, Turkey
| | - Sacide Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
40
|
Jawaid S, Strainic JP, Kim J, Ford MR, Thrane L, Karunamuni GH, Sheehan MM, Chowdhury A, Gillespie CA, Rollins AM, Jenkins MW, Watanabe M, Ford SM. Glutathione Protects the Developing Heart from Defects and Global DNA Hypomethylation Induced by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2021; 45:69-78. [PMID: 33206417 PMCID: PMC8865806 DOI: 10.1111/acer.14511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is caused by prenatal alcohol exposure (PAE), the intake of ethanol (C2 H5 OH) during pregnancy. Features of FASD cover a range of structural and functional defects including congenital heart defects (CHDs). Folic acid and choline, contributors of methyl groups to one-carbon metabolism (OCM), prevent CHDs in humans. Using our avian model of FASD, we have previously reported that betaine, another methyl donor downstream of choline, prevents CHDs. The CHD preventions are substantial but incomplete. Ethanol causes oxidative stress as well as depleting methyl groups for OCM to support DNA methylation and other epigenetic alterations. To identify more compounds that can safely and effectively prevent CHDs and other effects of PAE, we tested glutathione (GSH), a compound that regulates OCM and is known as a "master antioxidant." METHODS/RESULTS Quail embryos injected with a single dose of ethanol at gastrulation exhibited congenital defects including CHDs similar to those identified in FASD individuals. GSH injected simultaneously with ethanol not only prevented CHDs, but also improved survival and prevented other PAE-induced defects. Assays of hearts at 8 days (HH stage 34) of quail development, when the heart normally has developed 4-chambers, showed that this single dose of PAE reduced global DNA methylation. GSH supplementation concurrent with PAE normalized global DNA methylation levels. The same assays performed on quail hearts at 3 days (HH stage 19-20) of development, showed no difference in global DNA methylation between controls, ethanol-treated, GSH alone, and GSH plus ethanol-treated cohorts. CONCLUSIONS GSH supplementation shows promise to inhibit effects of PAE by improving survival, reducing the incidence of morphological defects including CHDs, and preventing global hypomethylation of DNA in heart tissues.
Collapse
Affiliation(s)
- Safdar Jawaid
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - James P. Strainic
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Jun Kim
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Matthew R. Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Lars Thrane
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Ganga H. Karunamuni
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Megan M. Sheehan
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Amrin Chowdhury
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Brecksville-Broadview Heights High School, Broadview Heights OH 44147
| | - Caitlyn A. Gillespie
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Fisk University, Nashville TN 37208
| | - Andrew M. Rollins
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michael W. Jenkins
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University, Cleveland OH 44106
| | - Michiko Watanabe
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| | - Stephanie M Ford
- Department of Pediatrics, Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland OH 44106
| |
Collapse
|
41
|
Mohammad S, Page SJ, Sasaki T, Ayvazian N, Rakic P, Kawasawa YI, Hashimoto-Torii K, Torii M. Long-term spatial tracking of cells affected by environmental insults. J Neurodev Disord 2020; 12:38. [PMID: 33327938 PMCID: PMC7745478 DOI: 10.1186/s11689-020-09339-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Harsh environments surrounding fetuses and children can induce cellular damage in the developing brain, increasing the risk of intellectual disability and other neurodevelopmental disorders such as schizophrenia. However, the mechanisms by which early damage leads to disease manifestation in later life remain largely unknown. Previously, we demonstrated that the activation of heat shock (HS) signaling can be utilized as a unique reporter to label the cells that undergo specific molecular/cellular changes upon exposure to environmental insults throughout the body. Since the activation of HS signaling is an acute and transient event, this approach was not intended for long-term tracing of affected cells after the activation has diminished. In the present study, we generated new reporter transgenic mouse lines as a novel tool to achieve systemic and long-term tracking of affected cells and their progeny. Methods The reporter transgenic mouse system was designed so that the activation of HS signaling through HS response element (HSE) drives flippase (FLPo)-flippase recognition target (FRT) recombination-mediated permanent expression of the red fluorescent protein (RFP), tdTomato. With a priority on consistent and efficient assessment of the reporter system, we focused on intraperitoneal (i.p.) injection models of high-dose, short prenatal exposure to alcohol (ethanol) and sodium arsenite (ethanol at 4.0 g/kg/day and sodium arsenite at 5.0 mg/kg/day, at embryonic day (E) 12 and 13). Long-term reporter expression was examined in the brain of reporter mice that were prenatally exposed to these insults. Electrophysiological properties were compared between RFP+ and RFP− cortical neurons in animals prenatally exposed to arsenite. Results We detected RFP+ neurons and glia in the brains of postnatal mice that had been prenatally exposed to alcohol or sodium arsenite. In animals prenatally exposed to sodium arsenite, we also detected reduced excitability in RFP+ cortical neurons. Conclusion The reporter transgenic mice allowed us to trace the cells that once responded to prenatal environmental stress and the progeny derived from these cells long after the exposure in postnatal animals. Tracing of these cells indicates that the impact of prenatal exposure on neural progenitor cells can lead to functional abnormalities in their progeny cells in the postnatal brain. Further studies using more clinically relevant exposure models are warranted to explore this mechanism. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-020-09339-w.
Collapse
Affiliation(s)
- Shahid Mohammad
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Stephen J Page
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Toru Sasaki
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo, Japan
| | - Nicholas Ayvazian
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.,Institute of Biomedical Sciences, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, CT, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA. .,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
42
|
Jiang Q, Lu D, Wang F, Zhang Y, Cao L, Gui Y, Sun S. Folic acid supplement rescues ethanol-induced developmental defects in the zebrafish embryos. Acta Biochim Biophys Sin (Shanghai) 2020; 52:536-545. [PMID: 32369106 DOI: 10.1093/abbs/gmaa030] [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] [Received: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Fetal alcohol syndrome (FASD) describes a range of birth defects. Mechanisms of FASD-associated defects are not well understood. It has great significance to investigate whether nutrient supplements like folic acid (FA) can effectively rescue ethanol-induced defects. Moreover, it is very important to determine the optimal time for FA supplementation when it can most effectively antagonize the teratogenic effects of ethanol during embryonic development. Our results indicated that ethanol exposure interrupted the development of zebrafish embryos and induced multiple defects in cardiac function, pharyngeal arch arteries, vessel, craniofacial cartilage, pharyngeal arches, brain, somite and hemoglobin formation. The expressions of critical genes that play important roles in above organs such as tbx1, flk-1, hand2, ngn1, huc, titin, gata-1 and c-myb were reduced, and the apoptosis was increased in ethanol-treated group. FA supplementation could reverse ethanol-induced defects, improve the decreased expressions of above genes and reduce the apoptosis. We also found that giving FA at 6-12 h post-fertilization (hpf), which is at the gastrula period (5.25-10 hpf), can obviously prevent the teratogenicity of ethanol. This research provides clues for elucidating the mechanism of fetal abnormalities caused by alcohol intake and for preventing FASD.
Collapse
Affiliation(s)
- Qiu Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ding Lu
- Department of Pediatrics, Shanghai Municipal Eighth People's Hospital, Shanghai 200235, China
| | - Feng Wang
- Children’s Hospital, Fudan University, Shanghai 201102, China
| | - Yawen Zhang
- Children’s Hospital, Fudan University, Shanghai 201102, China
| | - Li Cao
- Department of Ultrasound, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Yonghao Gui
- Children’s Hospital, Fudan University, Shanghai 201102, China
| | - Shuna Sun
- Children’s Hospital, Fudan University, Shanghai 201102, China
| |
Collapse
|
43
|
Kaminen-Ahola N. Fetal alcohol spectrum disorders: Genetic and epigenetic mechanisms. Prenat Diagn 2020; 40:1185-1192. [PMID: 32386259 DOI: 10.1002/pd.5731] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/26/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are a consequence of prenatal alcohol exposure (PAE). The etiology of the complex FASD phenotype with growth deficit, birth defects, and neurodevelopmental impairments is under extensive research. Both genetic and environmental factors contribute to the wide phenotype: chromosomal rearrangements, risk and protective alleles, environmental-induced epigenetic alterations as well as gene-environment interactions are all involved. Understanding the molecular mechanisms of PAE can provide tools for prevention or intervention of the alcohol-induced developmental disorders in the future. By revealing the alcohol-induced genetic and epigenetic alterations which associate with the variable FASD phenotypes, it is possible to identify biomarkers for the disorder. This would enable early diagnoses and personalized support for development of the affected child.
Collapse
Affiliation(s)
- Nina Kaminen-Ahola
- Environmental Epigenetics Laboratory, Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
44
|
Kahila H, Marjonen H, Auvinen P, Avela K, Riikonen R, Kaminen‐Ahola N. 18q12.3-q21.1 microdeletion detected in the prenatally alcohol-exposed dizygotic twin with discordant fetal alcohol syndrome phenotype. Mol Genet Genomic Med 2020; 8:e1192. [PMID: 32096599 PMCID: PMC7196488 DOI: 10.1002/mgg3.1192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A pair of dizygotic twins discordantly affected by heavy prenatal alcohol exposure (PAE) was reported previously by Riikonen, suggesting the role of genetic risk or protective factors in the etiology of alcohol-induced developmental disorders. Now, we have re-examined these 25-year-old twins and explored genetic origin of the phenotypic discordancy reminiscent with fetal alcohol syndrome (FAS). Furthermore, we explored alterations in DNA methylation profile of imprinting control region at growth-related insulin-like growth factor 2 (IGF2)/H19 locus in twins' white blood cells (WBC), which have been associated earlier with alcohol-induced genotype-specific changes in placental tissue. METHODS Microarray-based comparative genomic hybridization (aCGH) was used to detect potential submicroscopic chromosomal abnormalities, and developmental as well as phenotypic information about twins were collected. Traditional bisulfite sequencing was used for DNA methylation analysis. RESULTS Microarray-based comparative genomic hybridization revealed a microdeletion 18q12.3-q21.1. in affected twin, residing in a known 18q deletion syndrome region. This syndrome has been associated with growth restriction, developmental delay or intellectual deficiency, and abnormal facial features in previous studies, and thus likely explains the phenotypic discordancy between the twins. We did not observe association between WBCs' DNA methylation profile and PAE, but interestingly, a trend of decreased DNA methylation at the imprinting control region was seen in the twin with prenatal growth retardation at birth. CONCLUSIONS The microdeletion emphasizes the importance of adequate chromosomal testing in examining the etiology of complex alcohol-induced developmental disorders. Furthermore, the genotype-specific decreased DNA methylation at the IGF2/H19 locus cannot be considered as a biological mark for PAE in adult WBCs.
Collapse
Affiliation(s)
- Hanna Kahila
- Department of Obstetrics and GynecologyHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Heidi Marjonen
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| | - Pauliina Auvinen
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| | - Kristiina Avela
- Department of Clinical GeneticsHelsinki University HospitalHUSLABHelsinkiFinland
| | - Raili Riikonen
- Children's HospitalKuopio University HospitalUniversity of Eastern FinlandKuopioFinland
| | - Nina Kaminen‐Ahola
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
45
|
Vukic M, Wu H, Daxinger L. Making headway towards understanding how epigenetic mechanisms contribute to early-life effects. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180126. [PMID: 30966890 DOI: 10.1098/rstb.2018.0126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
It has become clear that in addition to the DNA sequence there is another layer of information, termed epigenetic modifications, that can influence phenotypes and traits. In particular, environmental epigenomics, which addresses the effect of the environment on the epigenome and human health, is becoming an area of great interest for many researchers working in different scientific fields. In this review, we will consider the current evidence that early-life environmental signals can have long-term effects on the epigenome. We will further evaluate how recent technological advances may enable us to unravel the molecular mechanisms underlying these phenomena, which will be crucial for understanding heritability in health and disease. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Maja Vukic
- Department of Human Genetics, Leiden University Medical Centre , Einthovenweg 20, 2333 ZC Leiden , The Netherlands
| | - Haoyu Wu
- Department of Human Genetics, Leiden University Medical Centre , Einthovenweg 20, 2333 ZC Leiden , The Netherlands
| | - Lucia Daxinger
- Department of Human Genetics, Leiden University Medical Centre , Einthovenweg 20, 2333 ZC Leiden , The Netherlands
| |
Collapse
|
46
|
Cerrizuela S, Vega-Lopez GA, Aybar MJ. The role of teratogens in neural crest development. Birth Defects Res 2020; 112:584-632. [PMID: 31926062 DOI: 10.1002/bdr2.1644] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/11/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
The neural crest (NC), discovered by Wilhelm His 150 years ago, gives rise to a multipotent migratory embryonic cell population that generates a remarkably diverse and important array of cell types during the development of the vertebrate embryo. These cells originate in the neural plate border (NPB), which is the ectoderm between the neural plate and the epidermis. They give rise to the neurons and glia of the peripheral nervous system, melanocytes, chondrocytes, smooth muscle cells, odontoblasts and neuroendocrine cells, among others. Neurocristopathies are a class of congenital diseases resulting from the abnormal induction, specification, migration, differentiation or death of NC cells (NCCs) during embryonic development and have an important medical and societal impact. In general, congenital defects affect an appreciable percentage of newborns worldwide. Some of these defects are caused by teratogens, which are agents that negatively impact the formation of tissues and organs during development. In this review, we will discuss the teratogens linked to the development of many birth defects, with a strong focus on those that specifically affect the development of the NC, thereby producing neurocristopathies. Although increasing attention is being paid to the effect of teratogens on embryonic development in general, there is a strong need to critically evaluate the specific role of these agents in NC development. Therefore, increased understanding of the role of these factors in NC development will contribute to the planning of strategies aimed at the prevention and treatment of human neurocristopathies, whose etiology was previously not considered.
Collapse
Affiliation(s)
- Santiago Cerrizuela
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Guillermo A Vega-Lopez
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Manuel J Aybar
- Área Biología Experimental, Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), Tucumán, Argentina.,Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
47
|
Collier AD, Min SS, Campbell SD, Roberts MY, Camidge K, Leibowitz SF. Maternal ethanol consumption before paternal fertilization: Stimulation of hypocretin neurogenesis and ethanol intake in zebrafish offspring. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109728. [PMID: 31394141 PMCID: PMC6815720 DOI: 10.1016/j.pnpbp.2019.109728] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 12/27/2022]
Abstract
There are numerous clinical and pre-clinical studies showing that exposure of the embryo to ethanol markedly affects neuronal development and stimulates alcohol drinking and related behaviors. In rodents and zebrafish, our studies show that embryonic exposure to low-dose ethanol, in addition to increasing voluntary ethanol intake during adolescence, increases the density of hypothalamic hypocretin (hcrt) neurons, a neuropeptide known to regulate reward-related behaviors. The question addressed here in zebrafish is whether maternal ethanol intake before conception also affects neuronal and behavioral development, phenomena suggested by clinical reports but seldom investigated. To determine if preconception maternal ethanol consumption also affects these hcrt neurons and behavior in the offspring, we first standardized a method of measuring voluntary ethanol consumption in AB strain adult and larval zebrafish given gelatin meals containing 10% or 0.1% ethanol, respectively. We found the number of bites of gelatin to be an accurate measure of intake in adults and a strong predictor of blood ethanol levels, and also to be a reliable indicator of intake in larval zebrafish. We then used this feeding paradigm and live imaging to examine the effects of preconception maternal intake of 10% ethanol-gelatin compared to plain-gelatin for 14 days on neuronal development in the offspring. Whereas ethanol consumption by adult female HuC:GFP transgenic zebrafish had no impact on the number of differentiated HuC+ neurons at 28 h post-fertilization (hpf), preconception ethanol consumption by adult female hcrt:EGFP zebrafish significantly increased the number of hcrt neurons in the offspring, an effect observed at 28 hpf and confirmed at 6 and 12 days post-fertilization (dpf). This increase in hcrt neurons was primarily present on the left side of the brain, indicating asymmetry in ethanol's actions, and it was accompanied by behavioral changes in the offspring, including a significant increase in novelty-induced locomotor activity but not thigmotaxis measured at 6 dpf and also in voluntary consumption of 0.1% ethanol-gelatin at 12 dpf. Notably, these measures of ethanol intake and locomotor activity stimulated by preconception ethanol were strongly, positively correlated with the number of hcrt neurons. These findings demonstrate that preconception maternal ethanol consumption affects the brain and behavior of the offspring, producing effects similar to those caused by embryonic ethanol exposure, and they provide further evidence that the ethanol-induced increase in hcrt neurogenesis contributes to the behavioral disturbances caused by ethanol.
Collapse
Affiliation(s)
- Adam D Collier
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - Soe S Min
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - Samantha D Campbell
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - Mia Y Roberts
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - Kaylin Camidge
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - Sarah F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
48
|
Park B, Khanam R, Vinayachandran V, Baqui AH, London SJ, Biswal S. Epigenetic biomarkers and preterm birth. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa005. [PMID: 32551139 PMCID: PMC7293830 DOI: 10.1093/eep/dvaa005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 05/06/2023]
Abstract
Preterm birth (PTB) is a major public health challenge, and novel, sensitive approaches to predict PTB are still evolving. Epigenomic markers are being explored as biomarkers of PTB because of their molecular stability compared to gene expression. This approach is also relatively new compared to gene-based diagnostics, which relies on mutations or single nucleotide polymorphisms. The fundamental principle of epigenome diagnostics is that epigenetic reprogramming in the target tissue (e.g. placental tissue) might be captured by more accessible surrogate tissue (e.g. blood) using biochemical epigenome assays on circulating DNA that incorporate methylation, histone modifications, nucleosome positioning, and/or chromatin accessibility. Epigenomic-based biomarkers may hold great potential for early identification of the majority of PTBs that are not associated with genetic variants or mutations. In this review, we discuss recent advances made in the development of epigenome assays focusing on its potential exploration for association and prediction of PTB. We also summarize population-level cohort studies conducted in the USA and globally that provide opportunities for genetic and epigenetic marker development for PTB. In addition, we summarize publicly available epigenome resources and published PTB studies. We particularly focus on ongoing genome-wide DNA methylation and epigenome-wide association studies. Finally, we review the limitations of current research, the importance of establishing a comprehensive biobank, and possible directions for future studies in identifying effective epigenome biomarkers to enhance health outcomes for pregnant women at risk of PTB and their infants.
Collapse
Affiliation(s)
- Bongsoo Park
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Vinesh Vinayachandran
- School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Abdullah H Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, International Center for Maternal and Newborn Health, Baltimore, MD 21205, USA
| | - Stephanie J London
- Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
49
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
50
|
Bertozzi TM, Ferguson-Smith AC. Metastable epialleles and their contribution to epigenetic inheritance in mammals. Semin Cell Dev Biol 2020; 97:93-105. [PMID: 31551132 DOI: 10.1016/j.semcdb.2019.08.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/02/2023]
Abstract
Many epigenetic differences between individuals are driven by genetic variation. Mammalian metastable epialleles are unusual in that they show variable DNA methylation states between genetically identical individuals. The occurrence of such states across generations has resulted in their consideration by many as strong evidence for epigenetic inheritance in mammals, with the classic Avy and AxinFu mouse models - each products of repeat element insertions - being the most widely accepted examples. Equally, there has been interest in exploring their use as epigenetic biosensors given their susceptibility to environmental compromise. Here we review the classic murine metastable epialleles as well as more recently identified candidates, with the aim of providing a more holistic understanding of their biology. We consider the extent to which epigenetic inheritance occurs at metastable epialleles and explore the limited mechanistic insights into the establishment of their variable epigenetic states. We discuss their environmental modulation and their potential relevance in genome regulation. In light of recent whole-genome screens for novel metastable epialleles, we point out the need to reassess their biological relevance in multi-generational studies and we highlight their value as a model to study repeat element silencing as well as the mechanisms and consequences of mammalian epigenetic stochasticity.
Collapse
Affiliation(s)
- Tessa M Bertozzi
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|