1
|
Hussain S, Day D, Ellenbroek BA. Preconceptual paternal ethanol drinking induces sexually dimorphic behavioural changes across 2 generations. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06807-w. [PMID: 40389584 DOI: 10.1007/s00213-025-06807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 05/03/2025] [Indexed: 05/21/2025]
Abstract
This study aimed to assess both the inter and transgenerational impacts of preconceptual paternal ethanol Exposure (PPEE) using a rat model. Sprague Dawley male rats (F0) underwent chronic voluntary ethanol intake and at the end of the drinking paradigm were kept for one spermatogenesis cycle before being mated with ethanol naïve females. The litters and matched controls were behaviourally assessed, and a cohort of F1 males mated to observe a F2 generation. PPEE caused behavioural changes in both the F1 and F2 generations, including altering litter sizes and delaying development. The F1 also show a reduction in sensitivity to the motor impairing effects of ethanol compared to controls. Sexually dimorphic effects were seen with female offspring having a reduced preference to ethanol in both the F1 and F2, while tolerance to ethanol induced motor coordination was seen in the F2 females but not F2 males. Likewise, F1 males presented reductions in locomotor activity but these effects did not persist in the F2. The findings show PPEE induces transgenerational changes in development, drinking behaviour and ethanol sensitivity in a sexually dimorphic manner. These changes may be protective to the female offspring of PPEE to modify their ethanol intake. The alterations demonstrate potential far-reaching consequences for the metabolism of xenotoxic substances extending beyond ethanol and provides evidence to support developmental and behavioural changes across generations due to paternal alcohol consumption.
Collapse
Affiliation(s)
- Sahir Hussain
- School of Psychological Sciences, Te Herenga Waka - Victoria University of Wellington, Wellington, New Zealand
| | - Darren Day
- School of Biological Sciences, Te Herenga Waka - Victoria University of Wellington, Wellington, New Zealand
| | - Bart A Ellenbroek
- School of Psychological Sciences, Te Herenga Waka - Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
2
|
Dunkley J, Deputy NP, Denny CH, Bertrand J, Godfred-Cato S, Kim SY. Assessing Prenatal Alcohol Exposure History for Pediatric Patients: Practices Among U.S. Clinicians. Matern Child Health J 2025; 29:449-456. [PMID: 39567462 PMCID: PMC12005974 DOI: 10.1007/s10995-024-04015-0] [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] [Accepted: 11/01/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVES The American Academy of Pediatrics recommends clinicians who treat pediatric patients screen for prenatal alcohol exposure (PAE) to facilitate the identification of children with fetal alcohol spectrum disorders and promote timely access to behavioral and cognitive interventions. We evaluated how frequently clinicians inquire about PAE in their pediatric patient interactions and the methods used to ascertain this information. METHODS We analyzed data from the Fall 2020 DocStyles survey, a web-based survey of primary healthcare professionals (n = 1754). Distributions for frequency of assessing PAE history for five pediatric populations and the methods used were calculated by clinician specialty (family practitioners [FP], pediatricians, and nurse practitioners/physician assistants [NP/PAs]) and overall. Chi-square and Bonferroni post-hoc tests determined whether frequency of assessing PAE history varied by specialty. RESULTS Among 779 clinicians serving pediatric patients, approximately 70.5%, 63.0%, and 60.7% reported often/always obtaining PAE history from parents of children with developmental/behavioral issues, adopted/foster children, and newborns, respectively. By contrast, less than half of respondents reported often/always collecting this information from parents of infants (47.6%) and new patients (38.2%). Most respondents reported collecting PAE history through interviews conducted by physicians or physician assistants (69.7%). Obtaining PAE history varied by specialty; pediatricians (71.5%) were more likely to collect PAE history for adopted/foster children when compared to FPs (57.7%, p = 0.003). CONCLUSIONS FOR PRACTICE PAE history is not routinely obtained for pediatric patients. These findings highlight the need for trainings and practice supports to aid clinicians in identifying and treating children at-risk of FASDs.
Collapse
Affiliation(s)
- Janae Dunkley
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA.
| | - Nicholas P Deputy
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA
- US Public Health Service Commissioned Corps, Rockville, MD, USA
| | - Clark H Denny
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA
| | - Jacquelyn Bertrand
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA
| | - Shana Godfred-Cato
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA
| | - Shin Y Kim
- Division of Birth Defects and Infant Disorders, National Center On Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, 4770 Buford Hwy NE, Atlanta, Chamblee, GA, 30341, USA
| |
Collapse
|
3
|
Hwang HM, Yamashita S, Matsumoto Y, Ito M, Edwards A, Sasaki J, Dutta DJ, Mohammad S, Yamashita C, Wetherill L, Schwantes-An TH, Abreu M, Mahnke AH, Mattson SN, Foroud T, Miranda RC, Chambers C, Torii M, Hashimoto-Torii K. Reduction of APOE accounts for neurobehavioral deficits in fetal alcohol spectrum disorders. Mol Psychiatry 2024; 29:3364-3380. [PMID: 38734844 PMCID: PMC11541007 DOI: 10.1038/s41380-024-02586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
A hallmark of fetal alcohol spectrum disorders (FASD) is neurobehavioral deficits that still do not have effective treatment. Here, we present that reduction of Apolipoprotein E (APOE) is critically involved in neurobehavioral deficits in FASD. We show that prenatal alcohol exposure (PAE) changes chromatin accessibility of Apoe locus, and causes reduction of APOE levels in both the brain and peripheral blood in postnatal mice. Of note, postnatal administration of an APOE receptor agonist (APOE-RA) mitigates motor learning deficits and anxiety in those mice. Several molecular and electrophysiological properties essential for learning, which are altered by PAE, are restored by APOE-RA. Our human genome-wide association study further reveals that the interaction of PAE and a single nucleotide polymorphism in the APOE enhancer which chromatin is closed by PAE in mice is associated with lower scores in the delayed matching-to-sample task in children. APOE in the plasma is also reduced in PAE children, and the reduced level is associated with their lower cognitive performance. These findings suggest that controlling the APOE level can serve as an effective treatment for neurobehavioral deficits in FASD.
Collapse
Grants
- F31AA027693 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA021886 NIAAA NIH HHS
- R01 AA026272 NIAAA NIH HHS
- U01 AA014834 NIAAA NIH HHS
- U01AA014834 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U24AA030169 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA014835 NIAAA NIH HHS
- P50HD105328 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA014835 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 AA025215 NIAAA NIH HHS
- R01AA026272 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AA026103 NIAAA NIH HHS
- R01AA025215 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA014809 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AA025103 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P50 HD105328 NICHD NIH HHS
- U24 AA030169 NIAAA NIH HHS
- U01 AA014809 NIAAA NIH HHS
- F31 AA027693 NIAAA NIH HHS
- District of Columbia Intellectual and Developmental Disabilities Research Center (DC-IDDRC) Award program
- Lilly Endowment
- Texas A&M University’s Accountability, Climate, Equity, and Scholarship (ACES) Faculty Fellows Program
Collapse
Affiliation(s)
- Hye M Hwang
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Yu Matsumoto
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Mariko Ito
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Alex Edwards
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Junko Sasaki
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Dipankar J Dutta
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Shahid Mohammad
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Chiho Yamashita
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Leah Wetherill
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marco Abreu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amanda H Mahnke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Sarah N Mattson
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Christina Chambers
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Masaaki Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, The Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Departments of Pediatrics, and Pharmacology & Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA.
| |
Collapse
|
4
|
Minozzi S, Ambrosi L, Saulle R, Uhm SS, Terplan M, Sinclair JM, Agabio R. Psychosocial and medication interventions to stop or reduce alcohol consumption during pregnancy. Cochrane Database Syst Rev 2024; 4:CD015042. [PMID: 38682758 PMCID: PMC11057221 DOI: 10.1002/14651858.cd015042.pub2] [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] [Indexed: 05/01/2024]
Abstract
BACKGROUND Despite the known harms, alcohol consumption is common in pregnancy. Rates vary between countries, and are estimated to be 10% globally, with up to 25% in Europe. OBJECTIVES To assess the efficacy of psychosocial interventions and medications to reduce or stop alcohol consumption during pregnancy. SEARCH METHODS We searched the Cochrane Drugs and Alcohol Group Specialised Register (via CRSLive), Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, CINAHL, Web of Science, and PsycINFO, from inception to 8 January 2024. We also searched for ongoing and unpublished studies via ClinicalTrials.gov and the World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP). All searches included non-English language literature. We handsearched references of topic-related systematic reviews and included studies. SELECTION CRITERIA We included randomised controlled trials that compared medications or psychosocial interventions, or both, to placebo, no intervention, usual care, or other medications or psychosocial interventions used to reduce or stop alcohol use during pregnancy. Our primary outcomes of interest were abstinence from alcohol, reduction in alcohol consumption, retention in treatment, and women with any adverse event. DATA COLLECTION AND ANALYSIS We used standard Cochrane methodological procedures. MAIN RESULTS We included eight studies (1369 participants) in which pregnant women received an intervention to stop or reduce alcohol use during pregnancy. In one study, almost half of participants had a current diagnosis of alcohol use disorder (AUD); in another study, 40% of participants had a lifetime diagnosis of AUD. Six studies took place in the USA, one in Spain, and one in the Netherlands. All included studies evaluated the efficacy of psychosocial interventions; we did not find any study that evaluated the efficacy of medications for the treatment of AUD during pregnancy. Psychosocial interventions were mainly brief interventions ranging from a single session of 10 to 60 minutes to five sessions of 10 minutes each. Pregnant women received the psychosocial intervention approximately at the end of the first trimester of pregnancy, and the outcome of alcohol use was reassessed 8 to 24 weeks after the psychosocial intervention. Women in the control group received treatment as usual (TAU) or similar treatments such as comprehensive assessment of alcohol use and advice to stop drinking during pregnancy. Globally, we found that, compared to TAU, psychosocial interventions may increase the rate of continuously abstinent participants (risk ratio (RR) 1.34, 95% confidence interval (CI) 1.14 to 1.57; I2 =0%; 3 studies; 378 women; low certainty evidence). Psychosocial interventions may have little to no effect on the number of drinks per day, but the evidence is very uncertain (mean difference -0.42, 95% CI -1.13 to 0.28; I2 = 86%; 2 studies; 157 women; very low certainty evidence). Psychosocial interventions probably have little to no effect on the number of women who completed treatment (RR 0.98, 95% CI 0.94 to 1.02; I2 = 0%; 7 studies; 1283 women; moderate certainty evidence). None of the included studies assessed adverse events of treatments. We downgraded the certainty of the evidence due to risk of bias and imprecision of the estimates. AUTHORS' CONCLUSIONS Brief psychosocial interventions may increase the rate of continuous abstinence among pregnant women who report alcohol use during pregnancy. Further studies should be conducted to investigate the efficacy and safety of psychosocial interventions and other treatments (e.g. medications) for women with AUD. These studies should provide detailed information on alcohol use before and during pregnancy using consistent measures such as the number of drinks per drinking day. When heterogeneous populations are recruited, more detailed information on alcohol use during pregnancy should be provided to allow future systematic reviews to be conducted. Other important information that would enhance the usefulness of these studies would be the presence of other comorbid conditions such as anxiety, mood disorders, and the use of other psychoactive substances.
Collapse
Affiliation(s)
- Silvia Minozzi
- Department of Epidemiology, Lazio Regional Health Service, Rome, Italy
| | - Ludovico Ambrosi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Rosella Saulle
- Department of Epidemiology, Lazio Regional Health Service, Rome, Italy
| | - Seilin S Uhm
- School of Medicine, University of Southampton, Southampton, UK
| | - Mishka Terplan
- Friends Research Institute, Baltimore, USA
- Family and Community Medicine, University of California, San Francisco, San Francisco, USA
| | | | - Roberta Agabio
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| |
Collapse
|
5
|
Rockhold MN, Gimbel BA, Richardson AA, Kautz-Turnbull C, Speybroeck EL, de Water E, Myers J, Hargrove E, May M, Abdi SS, Petrenko CLM. Racial and ethnic disparities in psychological care for individuals with FASD: a dis/ability studies and critical race theory perspective toward improving prevention, assessment/diagnosis, and intervention. Front Public Health 2024; 12:1355802. [PMID: 38544727 PMCID: PMC10965703 DOI: 10.3389/fpubh.2024.1355802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/16/2024] [Indexed: 05/14/2024] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are among the most common neurodevelopmental disorders and substantially impact public health. FASD can affect people of all races and ethnicities; however, there are important racial and ethnic disparities in alcohol-exposed pregnancy prevention, assessment and diagnosis of FASD, and interventions to support individuals with FASD and their families. In this article we use the Dis/Ability Studies and Critical Race Theory (Dis/Crit) framework to structure the exploration of disparities and possible solutions within these three areas (prevention, diagnosis, intervention). Dis/Crit provides a guide to understanding the intersection of dis/ability and race, while framing both as social constructs. Following the Dis/Crit framework, the systemic, historical, and contemporary racism and ableism present in psychological care is further discussed. We aim to elucidate these racial and ethnic disparities within the fields of psychology and neuropsychology through the Dis/Crit framework and provide potential points of action to reduce these disparities.
Collapse
Affiliation(s)
| | - Blake A. Gimbel
- Department of Pediatrics, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | | | | | - Emily L. Speybroeck
- Mt. Hope Family Center, University of Rochester, Rochester, NY, United States
| | - Erik de Water
- Great Lakes Neurobehavioral Center, Edina, MN, United States
| | - Julianne Myers
- Mt. Hope Family Center, University of Rochester, Rochester, NY, United States
| | - Emily Hargrove
- International Adult Leadership Collaborative of FASD Changemakers
| | - Maggie May
- International Adult Leadership Collaborative of FASD Changemakers
| | - Samia S. Abdi
- Department of Pediatrics, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | | |
Collapse
|
6
|
Darbinian N, Merabova N, Tatevosian G, Morrison M, Darbinyan A, Zhao H, Goetzl L, Selzer ME. Biomarkers of Affective Dysregulation Associated with In Utero Exposure to EtOH. Cells 2023; 13:2. [PMID: 38201206 PMCID: PMC10778368 DOI: 10.3390/cells13010002] [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: 11/04/2023] [Revised: 12/13/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
INTRODUCTION Children with fetal alcohol spectrum disorders (FASD) exhibit behavioral and affective dysregulation, including hyperactivity and depression. The mechanisms are not known, but they could conceivably be due to postnatal social or environmental factors. However, we postulate that, more likely, the affective dysregulation is associated with the effects of EtOH exposure on the development of fetal serotonergic (5-HT) and/or dopaminergic (DA) pathways, i.e., pathways that in postnatal life are believed to regulate mood. Many women who use alcohol (ethanol, EtOH) during pregnancy suffer from depression and take selective serotonin reuptake inhibitors (SSRIs), which might influence these monoaminergic pathways in the fetus. Alternatively, monoaminergic pathway abnormalities might reflect a direct effect of EtOH on the fetal brain. To distinguish between these possibilities, we measured their expressions in fetal brains and in fetal brain-derived exosomes (FB-Es) isolated from the mothers' blood. We hypothesized that maternal use of EtOH and/or SSRIs during pregnancy would be associated with impaired fetal neural development, detectable as abnormal levels of monoaminergic and apoptotic biomarkers in FB-Es. METHODS Fetal brain tissues and maternal blood were collected at 9-23 weeks of pregnancy. EtOH groups were compared with unexposed controls matched for gestational age (GA). The expression of 84 genes associated with the DA and 5-HT pathways was analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) on microarrays. FB-Es also were assayed for serotonin transporter protein (SERT) and brain-derived neurotrophic factor (BDNF) by enzyme-linked immunosorbent assay (ELISA). RESULTS Six EtOH-exposed human fetal brain samples were compared to SSRI- or polydrug-exposed samples and to unexposed controls. EtOH exposure was associated with significant upregulation of DA receptor D3 and 5-HT receptor HTR2C, while HTR3A was downregulated. Monoamine oxidase A (MAOA), MAOB, the serine/threonine kinase AKT3, and caspase-3 were upregulated, while mitogen-activated protein kinase 1 (MAPK1) and AKT2 were downregulated. ETOH was associated with significant upregulation of the DA transporter gene, while SERT was downregulated. There were significant correlations between EtOH exposure and (a) caspase-3 activation, (b) reduced SERT protein levels, and (c) reduced BDNF levels. SSRI exposure independently increased caspase-3 activity and downregulated SERT and BDNF. Early exposure to EtOH and SSRI together was associated synergistically with a significant upregulation of caspase-3 and a significant downregulation of SERT and BDNF. Reduced SERT and BDNF levels were strongly correlated with a reduction in eye diameter, a somatic manifestation of FASD. CONCLUSIONS Maternal use of EtOH and SSRI during pregnancy each was associated with changes in fetal brain monoamine pathways, consistent with potential mechanisms for the affective dysregulation associated with FASD.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
- Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
| | - Mary Morrison
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
- Department of Psychiatry, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Huaqing Zhao
- Center for Biostatistics and Epidemiology, Department of Biomedical Education and Data Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA;
| | - Michael Edgar Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
7
|
Mazumdar R, Eberhart JK. Loss of Nicotinamide nucleotide transhydrogenase sensitizes embryos to ethanol-induced neural crest and neural apoptosis via generation of reactive oxygen species. Front Neurosci 2023; 17:1154621. [PMID: 37360166 PMCID: PMC10289183 DOI: 10.3389/fnins.2023.1154621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/10/2023] [Indexed: 06/28/2023] Open
Abstract
Fetal alcohol spectrum disorders (FASD) are a continuum of birth defects caused by prenatal alcohol exposure. FASD are the most common environmentally induced birth defect and are highly variable. The genetics of an individual influence the severity of their FASD phenotype. However, the genes that sensitize an individual to ethanol-induced birth defects are largely unknown. The ethanol-sensitive mouse substrain, C57/B6J, carries several known mutations including one in Nicotinamide nucleotide transhydrogenase (Nnt). Nnt is a mitochondrial transhydrogenase thought to have an important role in detoxifying reactive oxygen species (ROS) and ROS has been implicated in ethanol teratogenesis. To directly test the role of Nnt in ethanol teratogenesis, we generated zebrafish nnt mutants via CRISPR/Cas9. Zebrafish embryos were dosed with varying concentrations of ethanol across different timepoints and assessed for craniofacial malformations. We utilized a ROS assay to determine if this could be a contributing factor of these malformations. We found that exposed and unexposed mutants had higher levels of ROS compared to their wildtype counterparts. When treated with ethanol, nnt mutants experienced elevated apoptosis in the brain and neural crest, a defect that was rescued by administration of the antioxidant, N-acetyl cysteine (NAC). NAC treatment also rescued most craniofacial malformations. Altogether this research demonstrates that ethanol-induced oxidative stress leads to craniofacial and neural defects due to apoptosis in nnt mutants. This research further supports the growing body of evidence implicating oxidative stress in ethanol teratogenesis. These findings suggest that antioxidants can be used as a potential therapeutic in the treatment of FASD.
Collapse
Affiliation(s)
- Rayna Mazumdar
- Department of Molecular Biosciences, School of Natural Sciences, University of Texas at Austin, Austin, TX, United States
- Waggoner Center for Alcohol and Addiction Research, School of Pharmacy, University of Texas at Austin, Austin, TX, United States
| | - Johann K. Eberhart
- Department of Molecular Biosciences, School of Natural Sciences, University of Texas at Austin, Austin, TX, United States
- Waggoner Center for Alcohol and Addiction Research, School of Pharmacy, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
8
|
Taylor A, Whittaker A, Chandler A, Carnegie E. Accounts of women identified as drinking at 'high risk' during pregnancy: A meta-ethnography of missing voices. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2023; 117:104061. [PMID: 37245246 DOI: 10.1016/j.drugpo.2023.104061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/25/2023] [Accepted: 05/07/2023] [Indexed: 05/30/2023]
Abstract
BACKGROUND Drinking alcohol during pregnancy is associated with Foetal Alcohol Spectrum Disorders (FASD), and women who drink at higher levels are more likely to have a baby with FASD. Public health responses focus on population-level approaches to FASD prevention such as promoting abstinence and alcohol brief interventions. Efforts to better understand and respond to 'high risk' drinking during pregnancy have been largely ignored. This meta-ethnography of qualitative research aims to inform this policy and practice agenda. METHODS Ten health, social care, and social sciences databases were searched for qualitative studies published since 2000 exploring drinking during pregnancy. Studies that included accounts of women who described themselves, or were diagnosed as, alcohol-dependent during pregnancy, or reported drinking during pregnancy at levels considered by the Word Health Organisation to constitute 'high risk' drinking, were eligible. Noblit and Hare's analytic approach to meta-ethnography was used to synthesise the studies and eMERGe reporting guidance was followed. RESULTS Nine diverse studies were included. All explored the impact of social norms and relationships, women's knowledge about the risks involved in drinking during pregnancy, the behaviour of women, and the advice they received. Three key themes were identified: drinking is social and relational, knowledge is not enough, and multiple adversities matter. Multiple adversities were interconnected and primarily related to structural inequalities and oppression. The complex needs of women and the wider context in which their drinking occurred were rarely explored or responded to during pregnancy. CONCLUSION This meta-ethnography provides a more nuanced understanding of the complex dynamics involved in women's 'high risk' drinking during pregnancy, the contexts in which they drink and their unmet needs. These findings can inform future policy and practice responses to 'high risk' drinking during pregnancy. Further research should explore women's experiences in a UK context and consider how services could meet women's needs.
Collapse
Affiliation(s)
- Annie Taylor
- School of Health and Social Care, Edinburgh Napier University, Sighthill Campus, Sighthill, Edinburgh EH11 4BN, UK.
| | - Anne Whittaker
- NMAHP Research Unit, Faculty of Health Sciences and Sport, Pathfoot Building, University of Stirling, Stirling, Scotland, FK9 4LA, UK
| | - Amy Chandler
- School of Health in Social Science, University of Edinburgh, EH8 9AG, UK
| | - Elaine Carnegie
- School of Health and Social Care, Edinburgh Napier University, Sighthill Campus, Sighthill, Edinburgh EH11 4BN, UK
| |
Collapse
|
9
|
Cox LV. The Eastern Door Center: re-balancing the wheel-a Two-Eyed Seeing approach to FASD and other disorders related to transgenerational adversity. FRONTIERS IN SOCIOLOGY 2023; 8:910153. [PMID: 37214599 PMCID: PMC10196138 DOI: 10.3389/fsoc.2023.910153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/03/2023] [Indexed: 05/24/2023]
Abstract
In 2015, the Canadian Truth and Reconciliation Commission (TRC) called for immediate action to address the lack of access to health services for Fetal Alcohol Spectrum Disorder (FASD) in Indigenous communities. They called for the provision of culturally safe, community-based, FASD diagnostic, intervention and prevention services. FASD is a neurodevelopmental condition that can affect all aspects of functioning. The term refers to a spectrum of conditions occurring as a result of prenatal alcohol exposure (PAE) and associated risk factors. PAE can affect both physical and mental health leading to problems with learning, memory, attention, language, social behavior, executive functioning, sleep, and affect regulation. According to Elders in Mi'kmaq First Nations (FN) communities, FASD is a condition that is rooted in transgenerational trauma and the loss of relationship to their land, their language and the traditional community culture. The Elsipogtog Eastern Door (ED) Center opened in 2006 to provide culturally informed diagnosis, intervention and prevention for FASD and related conditions. The ED was the first FASD diagnostic team in Atlantic Canada and it served as a demonstration model for the New Brunswick FASD Center of Excellence as well as for Indigenous communities regionally and nationally. In this article, we outline the history and evolution of the Eastern Door Center and its programs and describe some of the successes of this model as well as some of its limitations in practice.
Collapse
|
10
|
Steane SE, Cuffe JSM, Moritz KM. The role of maternal choline, folate and one-carbon metabolism in mediating the impact of prenatal alcohol exposure on placental and fetal development. J Physiol 2023; 601:1061-1075. [PMID: 36755527 PMCID: PMC10952912 DOI: 10.1113/jp283556] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Prenatal alcohol consumption (PAE) may be associated with a broad spectrum of impacts, ranging from no overt effects, to miscarriage, fetal growth restriction and fetal alcohol spectrum disorder. A major mechanism underlying the effects of PAE is considered to be altered DNA methylation and gene expression. Maternal nutritional status may be an important factor in determining the extent to which PAE impacts pregnancy outcomes, particularly the dietary micronutrients folate and choline because they provide methyl groups for DNA methylation via one carbon metabolism. This review summarises the roles of folate and choline in development of the blastocyst, the placenta and the fetal brain, and examines the evidence that maternal intake of these micronutrients can modify the effects of PAE on development. Studies of folate or choline deficiency have found reduced blastocyst development and implantation, reduced placental invasion, vascularisation and nutrient transport capability, impaired fetal brain development, and abnormal neurodevelopmental outcomes. PAE has been shown to reduce absorption and/or metabolism of folate and choline and to produce similar outcomes to maternal choline/folate deficiency. A few studies have demonstrated that the effects of PAE on brain development can be ameliorated by folate or choline supplementation; however, there is very limited evidence on the effects of supplementation in early pregnancy on the blastocyst and placenta. Further studies are required to support these findings and to determine optimal supplementation parameters.
Collapse
Affiliation(s)
- Sarah E. Steane
- School of Biomedical SciencesThe University of QueenslandSt LuciaQLDAustralia
| | - James S. M. Cuffe
- School of Biomedical SciencesThe University of QueenslandSt LuciaQLDAustralia
| | - Karen M. Moritz
- School of Biomedical SciencesThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
11
|
Genetic Influences on Fetal Alcohol Spectrum Disorder. Genes (Basel) 2023; 14:genes14010195. [PMID: 36672936 PMCID: PMC9859092 DOI: 10.3390/genes14010195] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Fetal alcohol spectrum disorder (FASD) encompasses the range of deleterious outcomes of prenatal alcohol exposure (PAE) in the affected offspring, including developmental delay, intellectual disability, attention deficits, and conduct disorders. Several factors contribute to the risk for and severity of FASD, including the timing, dose, and duration of PAE and maternal factors such as age and nutrition. Although poorly understood, genetic factors also contribute to the expression of FASD, with studies in both humans and animal models revealing genetic influences on susceptibility. In this article, we review the literature related to the genetics of FASD in humans, including twin studies, candidate gene studies in different populations, and genetic testing identifying copy number variants. Overall, these studies suggest different genetic factors, both in the mother and in the offspring, influence the phenotypic outcomes of PAE. While further work is needed, understanding how genetic factors influence FASD will provide insight into the mechanisms contributing to alcohol teratogenicity and FASD risk and ultimately may lead to means for early detection and intervention.
Collapse
|
12
|
Fish EW, Mendoza-Romero HN, Love CA, Dragicevich CJ, Cannizzo MD, Boschen KE, Hepperla A, Simon JM, Parnell SE. The pro-apoptotic Bax gene modifies susceptibility to craniofacial dysmorphology following gastrulation-stage alcohol exposure. Birth Defects Res 2022; 114:1229-1243. [PMID: 35396933 PMCID: PMC10103739 DOI: 10.1002/bdr2.2009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND During early development, alcohol exposure causes apoptotic cell death in discrete regions of the embryo which are associated with distinctive patterns of later-life abnormalities. In gastrulation, which occurs during the third week of human pregnancy, alcohol targets the ectoderm, the precursor of the eyes, face, and brain. This midline tissue loss leads to the craniofacial dysmorphologies, such as microphthalmia and a smooth philtrum, which define fetal alcohol syndrome (FAS). An important regulator of alcohol-induced cell death is the pro-apoptotic protein Bax. The current study determines if mice lacking the Bax gene are less susceptible to the pathogenic effects of gastrulation-stage alcohol exposure. METHODS Male and female Bax+/- mice mated to produce embryos with full (-/- ) or partial (+/- ) Bax deletions, or Bax+/+ wild-type controls. On Gestational Day 7 (GD 7), embryos received two alcohol (2.9 g/kg, 4 hr apart), or control exposures. A subset of embryos was collected 12 hr later and examined for the presence of apoptotic cell death, while others were examined on GD 17 for the presence of FAS-like facial features. RESULTS Full Bax deletion reduced embryonic apoptotic cell death and the incidence of fetal eye and face malformations, indicating that Bax normally facilitates the development of alcohol-induced defects. An RNA-seq analysis of GD 7 Bax+/+ and Bax-/- embryos revealed 63 differentially expressed genes, some of which may interact with the Bax deletion to further protect against apoptosis. CONCLUSIONS Overall, these experiments identify that Bax is a primary teratogenic mechanism of gastrulation-stage alcohol exposure.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Haley N Mendoza-Romero
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Charlotte A Love
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Constance J Dragicevich
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael D Cannizzo
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Karen E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Austin Hepperla
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina, USA.,Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jeremy M Simon
- Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina, USA.,Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Gualdoni GS, Pérez-Tito L, Barril C, Sobarzo C, Cebral E. Abnormal growth and morphogenesis of placenta at term is linked to adverse fetal development after perigestational alcohol consumption up to early gestation in mouse. Birth Defects Res 2022; 114:611-630. [PMID: 35775613 DOI: 10.1002/bdr2.2063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 05/20/2022] [Accepted: 06/13/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Gestation alcohol consumption produces fetal growth restriction and malformations by affecting the embryo-fetal development. Recently a relationship between abnormal placentation and fetal malformation and intrauterine growth retardation has been suggested. However, the effects of perigestational alcohol ingestion up to early pregnancy on the placenta at term and its association with fetal abnormalities are little known. METHODS In female mice, ethanol 10% in water was administered for 15 days previous and up to days 4 (D4), 8 (D8), or 10 (D10) of gestation (TF), and gestation continues without ethanol exposure. Control females (CF) received ethanol-free water. At day 18, feto-placental units and implantation sites were studied. RESULTS TF had increased resorptions and only fetuses from D8-TF and D10-TF had significantly increased weights versus CF. D4 and D10-TF-placentas had significantly reduced weights. All TF had increased junctional zone (JZ) and reduced labyrinth (Lab) areas (PAS-histology and morphometry) compared with CF. Fetuses with mainly with craniofacial abnormalities and skeletal defects (Alizarin red staining), significantly increase; while the fetal bone density (alizarin color intensity, ImageJ) was reduced in D4, D8 and D10-TF versus CF. Although all TF-placentas were histo-structural affected, TF-abnormal fetuses had the most severe placental anomalies, with junctional abundant glycogenic cells into the labyrinth, disorganized labyrinthine vascularization with signs of leukocyte infiltrates and feto-maternal blood mix. CONCLUSIONS Perigestational alcohol consumption up to early gestation induces at term fetal growth alterations, dysmorphology and defective skeleton, linked to deficient growth and abnormal morphogenesis of placenta, highlighting insight into the prenatal etiology of FASD.
Collapse
Affiliation(s)
- Gisela Soledad Gualdoni
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biodiversidad y Biología Experimental (DBBE), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Leticia Pérez-Tito
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Camila Barril
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biodiversidad y Biología Experimental (DBBE), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cristian Sobarzo
- Facultad de Medicina, CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Elisa Cebral
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Biodiversidad y Biología Experimental (DBBE), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
14
|
Davies KJM, Richmond S, Medeiros-Mirra RJ, Abbas HH, Wilson-Nagrani CE, Davis MG, Zhurov A. The effect of maternal smoking and alcohol consumption on lip morphology. J Orthod 2022; 49:403-411. [PMID: 35723071 DOI: 10.1177/14653125221094337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To determine whether maternal smoking and/or alcohol consumption has an influence on lip morphology. Maternal smoking is a known risk factor for orofacial clefts; however, its influence on normal lip variation is unknown. Recent research regarding normal lip morphology has been contradictory. DESIGN Retrospective cohort study. SETTING AND PARTICIPANTS A total of 4747 children from the Avon Longitudinal Study of Parents and Children (ALSPAC) who each had 3D facial scans carried out at 15 years of age were included in the study. METHODS Each of the participants was automatically categorised regarding predetermined lip morphological traits. Questionnaires completed by their mothers identified smoking and alcohol habits during pregnancy. Logistic regression analyses were applied to determine the effect of maternal smoking and alcohol consumption on lip morphology. RESULTS Maternal smoking has significant effects on upper and lower lip contours, Cupid's bow, lower lip-chin shape and lower lip tone (all P < 0.05). There was also an indication of a potential epigenetic effect of smoking pre-pregnancy on upper lip contour (P = 0.0573). Alcohol consumption is significantly associated with philtrum shape, particularly when >6 units of alcohol are consumed per week (P = 0.0149, 32 weeks). Overall results suggest a deeply grooved philtrum is more likely if alcohol is consumed. Investigating the combined effect of smoking and alcohol consumption, lower lip contour (P = 0.00923) and lower lip-chin shape (P = 0.0171) are statistically significant, with lower lip contour more likely to be narrow in the midline, and lower lip-chin shape more likely to be an angular concavity. CONCLUSION Maternal smoking influences a number of lip traits, including a possible epigenetic effect on upper lip contour. Maternal alcohol consumption, particularly at a high level, influences philtrum shape. Maternal smoking and alcohol consumption have a combined effect on lower lip contour and lower lip-chin shape.
Collapse
Affiliation(s)
| | - Stephen Richmond
- Department of Orthodontics, School of Dentistry, Cardiff University, Cardiff, UK
| | | | - Hawraa Hassan Abbas
- Department of Orthodontics, School of Dentistry, Cardiff University, Cardiff, UK
| | | | - Megan Gael Davis
- Department of Orthodontics, School of Dentistry, Cardiff University, Cardiff, UK
| | - Alexei Zhurov
- Department of Orthodontics, School of Dentistry, Cardiff University, Cardiff, UK
| |
Collapse
|
15
|
What Obstetrician–Gynecologists Should Know About Substance Use Disorders in the Perinatal Period. Obstet Gynecol 2022; 139:317-337. [DOI: 10.1097/aog.0000000000004657] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022]
|
16
|
Ford SM, Pedersen CJ, Ford MR, Kim JW, Karunamuni GH, McPheeters MT, Jawaid S, Jenkins MW, Rollins AM, Watanabe M. Folic acid prevents functional and structural heart defects induced by prenatal ethanol exposure. Am J Physiol Heart Circ Physiol 2021. [DOI: 10.1152/ajpheart.00817.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
State-of-the-art biophotonic tools captured blood flow and endocardial cushion volumes in tiny beating quail embryo hearts, an accessible model for studying four-chambered heart development. Both hemodynamic flow and endocardial cushion volumes were altered with ethanol exposure but normalized when folic acid was introduced with ethanol. Folic acid supplementation preserved hemodynamic function that is intimately involved in sculpting the heart from the earliest stages of heart development.
Collapse
Affiliation(s)
- Stephanie M. Ford
- Division of Neonatology, Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Cameron J. Pedersen
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew R. Ford
- Department of Ophthalmology, Cole Eye Institute, Cleveland Clinic, Cleveland Ohio
| | - Jun W. Kim
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ganga H. Karunamuni
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew T. McPheeters
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Safdar Jawaid
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michael W. Jenkins
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michiko Watanabe
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
17
|
Koren G, Ornoy A. Institutionalized Children and the Risk of Fetal Alcohol Spectrum Disorder (FASD); A Primer for Clinicians, Adoption Staff and Parents. Glob Pediatr Health 2021; 8:2333794X21989556. [PMID: 33644259 PMCID: PMC7890725 DOI: 10.1177/2333794x21989556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/22/2020] [Accepted: 12/31/2020] [Indexed: 12/05/2022] Open
Abstract
Objectives: Our objective was to estimate the likelihood of abnormal development among institutionalized children, addressing either the risk in general, or the risk for fetal alcohol spectrum disorder (FASD). Methods: Narrative review of studies measuring developmental effects of these populations. We identified all systematic reviews and meta analyses dealing with the associations between institutionalization of children and their neurodevelopment in general, or between institunalization of children and their likelihood of suffering from FASD. Results: a) In a published meta-analysis the mean IQ/DQ was 84 among institutionalized children, as compared to 104 among children raised in families. Favorable caregiver-child ratios appeared to have a protective effect, whereas longer stays in institutions had a detrimental effect on IQ/DQ. b) A further meta- analysis has shown a positive impact of adoption on children’s cognitive development with adopted children’s displaying remarkably normal cognitive competence as compared to their non-adopted peers. c) The overall pooled prevalence was 6% (60 per 1,000, 95% CI 38-85) for full blown fetal alcohol syndrome (FAS), and 16.9% (95% CI 109-238 per 1,000) for the whole range of FASD. d) The estimated prevalence of FASD was 10-40 fold higher than the 7.7 per 1000 in the general population. Conclusions: A large proportion of adopted institutionalized children may not follow a normal developmental trajectory. If not afflicted by FASD, there is a positive impact of adoption on children’s cognitive development and in general they are comparable to their non- adopted peers.
Collapse
Affiliation(s)
- Gideon Koren
- Adelson faculty of medicine, Ariel University, Ariel, Israel.,Motherisk Israel, Clinical Pharmacology Unit, Shamir Hospital, Zrifin, Israel
| | - Asher Ornoy
- Motherisk Israel, Clinical Pharmacology Unit, Shamir Hospital, Zrifin, Israel.,Hebrew University Hadassah Medical School
| |
Collapse
|
18
|
May PA, Marais AS, De Vries MM, Buckley D, Kalberg WO, Hasken JM, Stegall JM, Hedrick DM, Robinson LK, Manning MA, Tabachnick BG, Seedat S, Parry CDH, Hoyme HE. The prevalence, child characteristics, and maternal risk factors for the continuum of fetal alcohol spectrum disorders: A sixth population-based study in the same South African community. Drug Alcohol Depend 2021; 218:108408. [PMID: 33250379 PMCID: PMC7756187 DOI: 10.1016/j.drugalcdep.2020.108408] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Prevalence and characteristics of fetal alcohol spectrum disorders (FASD) have been described previously in this community. METHODS Active case ascertainment methods were employed in a new cross-sectional study with Revised Institute of Medicine criteria among first grade students (n = 735) via dysmorphology examinations and neurobehavioral assessments. Their mothers were interviewed regarding risk factors. Final diagnoses were assigned via structured case conferences. RESULTS Children with fetal alcohol syndrome (FAS), partial FAS (PFAS), and alcohol related-neurodevelopmental disorder (ARND) were significantly different from controls on all cardinal variables, multiple dysmorphology traits and neurobehavioral performance. Mothers of children with FASD reported significantly more drinking before and during pregnancy (mothers of children with FAS reported 7.8 (±6.1) drinks per drinking day (DDD) prior to pregnancy and 5.1 (±5.9) after pregnancy recognition). Distal risk variables for a diagnosis on the continuum of FASD were: lower maternal height, weight, and body mass index; higher gravidity; lower education and household income; and later pregnancy recognition. Alcohol and tobacco remain the only commonly used drugs. Women reporting first trimester drinking of two DDD were 13 times more likely (95 % CI:1.3-133.4) to have a child with FASD than non-drinkers; and those who reported drinking throughout pregnancy were 19.4 times more likely (95 % CI:8.2-46.0) to have a child with FASD. CONCLUSION Seventeen years after the first study in this community, FASD prevalence remains high at 16 %-31 %. The FAS rate may have declined somewhat, but rates of PFAS and ARND seemed to plateau, at a high rate.
Collapse
Affiliation(s)
- Philip A May
- Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, United States; Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, 7505, South Africa; Center on Alcoholism, Substance Abuse and Addictions, The University of New Mexico, 2650 Yale SE, Albuquerque, NM, 87106, United States.
| | - Anna-Susan Marais
- Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, 7505, South Africa
| | - Marlene M De Vries
- Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, 7505, South Africa
| | - David Buckley
- Center on Alcoholism, Substance Abuse and Addictions, The University of New Mexico, 2650 Yale SE, Albuquerque, NM, 87106, United States
| | - Wendy O Kalberg
- Center on Alcoholism, Substance Abuse and Addictions, The University of New Mexico, 2650 Yale SE, Albuquerque, NM, 87106, United States
| | - Julie M Hasken
- Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, United States
| | - Julie M Stegall
- Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, United States
| | - Dixie M Hedrick
- Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC, 28081, United States
| | - Luther K Robinson
- Department of Pediatrics, State University of New York, 1001 Main Street, Buffalo, NY, 14203, United States
| | - Melanie A Manning
- Department of Pathology and Pediatrics, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA, 94305, United States
| | - Barbara G Tabachnick
- California State University, 18111 Nordhoff Street, Northridge, CA, 91330, United States
| | - Soraya Seedat
- Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, 7505, South Africa
| | - Charles D H Parry
- Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town, 7505, South Africa; Alcohol, Tobacco and Other Drug Research Unit, South African Medical Research Council, Francie van Zijl Drive, Parowvallei, Cape Town, 7505, South Africa
| | - H Eugene Hoyme
- Sanford Children's Genomic Medicine Consortium, Sanford Health, and the University of South Dakota Sanford School of Medicine, Department of Pediatrics, 1600 W. 22nd St., Sioux Falls, SD, 57117, United States; Department of Pediatrics and the Center for Applied Genetics and Genomic Medicine, The University of Arizona College of Medicine, 1501 N. Campbell Avenue, Tucson, Arizona, 85724, United States
| |
Collapse
|
19
|
Popova S, Lange S, Temple V, Poznyak V, Chudley AE, Burd L, Murray M, Rehm J. Profile of Mothers of Children with Fetal Alcohol Spectrum Disorder: A Population-Based Study in Canada. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217986. [PMID: 33143108 PMCID: PMC7663482 DOI: 10.3390/ijerph17217986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
Objective: To compare the characteristics of mothers of children with Fetal Alcohol Spectrum Disorder (FASD) with mothers of typically developing control children. Methods: The study utilized a cross-sectional, observational design, using active case ascertainment. Biological mothers were interviewed using a standardized retrospective questionnaire to collect data on demographics, living environment, pregnancy history, nutrition, alcohol and other drug use prior to and following pregnancy recognition. Results: A total of 173 mothers were interviewed. Of these, 19 had a child who was diagnosed with FASD, five had a child who had received a deferred FASD diagnosis, and 37 had children who were selected into the control group as typically developing children. The remaining 112 mothers had children who did not meet diagnostic criteria for FASD. The mothers of children with FASD did not differ significantly from mothers of the control group children with respect to age, ethnicity, marital status, and employment status at the time of pregnancy. However, mothers of children with FASD had lower levels of education (p < 0.01) and were more likely to have received financial support (p < 0.05) at the time of pregnancy, to have smoked tobacco (p < 0.001), and to have used marijuana or hashish (p < 0.01) prior to pregnancy recognition, compared with mothers of control children. All mothers of children with FASD reported alcohol consumption prior to pregnancy recognition; however, only 10.5% reported alcohol consumption following pregnancy recognition. None of the mothers interviewed reported any drug use following pregnancy recognition. Conclusions: Population-based preventive interventions, including repeated screening, monitoring, and education regarding the effects of alcohol use, as well as other substances, before and during pregnancy, are needed to eliminate risk for FASD and other negative consequences on child and maternal health.
Collapse
Affiliation(s)
- Svetlana Popova
- Centre for Addiction and Mental Health, Institute for Mental Health Policy Research, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (S.L.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, ON M5T 3M7, Canada
- Factor-Inwentash Faculty of Social Work, University of Toronto, 246 Bloor Street W, Toronto, ON M5S 1V4, Canada
- Institute of Medical Science, University of Toronto, Faculty of Medicine, Medical Sciences, Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Correspondence:
| | - Shannon Lange
- Centre for Addiction and Mental Health, Institute for Mental Health Policy Research, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (S.L.); (J.R.)
| | - Valerie Temple
- Surrey Place, 2 Surrey Place, Toronto, ON M5S 2C2, Canada;
| | - Vladimir Poznyak
- Department of Mental Health and Substance Abuse, World Health Organization, 20 Avenue Appia, CH-1211 Geneva, Switzerland;
| | - Albert E. Chudley
- Department of Paediatrics and Child Health, University of Manitoba, 840 Sherbrook Street, Winnipeg, MB R3A 1S1, Canada;
| | - Larry Burd
- North Dakota Fetal Alcohol Syndrome Center, Pediatrics Department, 1301 N Columbia Rd, Stop 9037 Grand Forks, ND 58202-9037, Canada;
| | - Margaret Murray
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Jürgen Rehm
- Centre for Addiction and Mental Health, Institute for Mental Health Policy Research, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (S.L.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, ON M5T 3M7, Canada
- Institute of Medical Science, University of Toronto, Faculty of Medicine, Medical Sciences, Building, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
- Institute of Clinical Psychology and Psychotherapy & Center of Clinical Epidemiology and Longitudinal Studies, Technische Universität Dresden, Chemnitzer Str. 46, 01187 Dresden, Germany
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
20
|
Post-exposure environment modulates long-term developmental ethanol effects on behavior, neuroanatomy, and cortical oscillations. Brain Res 2020; 1748:147128. [PMID: 32950485 DOI: 10.1016/j.brainres.2020.147128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/25/2020] [Accepted: 09/12/2020] [Indexed: 11/23/2022]
Abstract
Developmental exposure to ethanol has a wide range of anatomical, cellular, physiological and behavioral impacts that can last throughout life. In humans, this cluster of effects is termed fetal alcohol spectrum disorder and is highly prevalent in western cultures. The ultimate expression of the effects of developmental ethanol exposure however can be influenced by post-exposure experience. Here we examined the effects of developmental binge exposure to ethanol (postnatal day 7) in C57BL/6By mice on a specific cohort of inter-related long-term outcomes including contextual memory, hippocampal parvalbumin-expressing neuron density, frontal cortex oscillations related to sleep-wake cycling including delta oscillation amplitude and sleep spindle density, and home-cage behavioral activity. When assessed in adults that were raised in standard housing, all of these factors were altered by early ethanol exposure compared to saline controls except home-cage activity. However, exposure to an enriched environment and exercise from weaning to postnatal day 90 reversed most of these ethanol-induced impairments including memory, CA1 but not dentate gyrus PV+ cell density, delta oscillations and sleep spindles, and enhanced home-cage behavioral activity in Saline- but not EtOH-treated mice. The results are discussed in terms of the inter-dependence of diverse developmental ethanol outcomes and potential mechanisms of post-exposure experiences to regulate those outcomes.
Collapse
|
21
|
Nayak MB, Kaskutas LA, Mericle AA. Randomized Trial of an Innovative Electronic Screening and Brief Intervention for Reducing Drinking Among Women of Childbearing Age. J Addict Med 2020; 13:450-459. [PMID: 30882553 PMCID: PMC6742588 DOI: 10.1097/adm.0000000000000518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To evaluate the efficacy of an innovative, self-administered, electronic Screening and Brief Intervention (e-SBI) in English and Spanish, "DrinkWise," for reducing drinking among nonpregnant women of childbearing age. METHODS A parallel design, phase 1 trial included 185 nonpregnant women reporting risky drinking (8 or more drinks in a week or 3 or more drinks in a day) who were recruited from 2 publicly funded Nutritional Assistance for Women, Infants and Children (WIC) program sites in the United States from 2016 to 2017. Participants were 18 to 44 years in age, 75% of Hispanic ethnicity, 44% Spanish speakers, 30% had not completed high school, and 15% were currently breastfeeding. Participants were randomized to receive (intervention condition, n = 99) or not receive (control condition, no intervention, n = 86) DrinkWise and followed at 3 and 6 months. RESULTS Women receiving DrinkWise had greater reductions in the odds of self-reported weekly alcohol use (odds ratio [OR] = 0.22, SE = 0.12, P < 0.01) and heavy alcohol use (OR = 0.23, SE = 0.14, P < 0.05) at 6-month follow-up than controls, with no group differences at 3-month follow-up. Compared with heavy drinking controls, heavy drinkers receiving DrinkWise showed a trend (P = 0.06) for greater reductions in drink (pour) size from 3- to 6-month follow-up. CONCLUSIONS DrinkWise may be efficacious in reducing drinking among low-income women of childbearing age and provides a low-cost tool for increasing access to recommended SBI among childbearing-age women. Studies should continue to build DrinkWise's evidence base. TRIAL REGISTRATION ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT02337361.
Collapse
|
22
|
An interaction between fetal sex and placental weight and efficiency predicts intrauterine growth in response to maternal protein insufficiency and gestational exposure window in a mouse model of FASD. Biol Sex Differ 2020; 11:40. [PMID: 32690098 PMCID: PMC7372829 DOI: 10.1186/s13293-020-00320-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/13/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Individuals exposed to gestational stressors such as alcohol exhibit a spectrum of growth patterns, suggesting individualized responses to the stressors. We hypothesized that intrauterine growth responses to gestational alcohol are modified not only by the stressor's severity but by fetal sex and the placenta's adaptive capacity. METHODS Pregnant C57BL/6J mice were assigned to one of three groups. Group 1 consumed a normal protein diet (18% protein by weight) and received 4.5 g alcohol/kg body weight (NP-Alc-8) or isocaloric maltodextrin (NP-MD-8) daily from embryonic day (E) 8.5-E17.5. Group 2 consumed the same diet but received alcohol (NP-Alc-13) or maltodextrin (NP-MD-13) daily from E13.5-E17.5. Group 3 consumed the same diet but containing a lower protein content (12% protein by weight) from E0.5 and also received alcohol (LP-Alc-8) or maltodextrin (LP-MD-8) daily from E8.5-E17.5. Maternal, placental, and fetal outcomes were assessed on E17.5 using 2-way ANOVA or mixed linear model. RESULTS We found that intrauterine growth differed in the alcohol-exposed fetuses depending on sex and insult severity. Both NP-Alc-8 (vs. NP-MD-8) males and females had lower body weight and asymmetrical growth, but only NP-Alc-8 females had lower placental weight (P < 0.05). NP-Alc-13 (vs. NP-MD-13) females, but not their male littermates, had lower body weight (P = 0.019). Alcohol exposure beginning from E8.5 (vs. E13.5) decreased the ratio of fetal liver-to-body weight and increased the ratio of fetal brain-to-liver weight in both sexes (P < 0.05). LP-Alc-8 (vs. NP-MD-8) group had smaller litter size (P = 0.048), but the survivors had normal placental and body weight at E17.5. Nevertheless, LP-Alc-8 fetuses still showed asymmetrical growth. Correlation analyses reveal a relationship between litter size and placental outcomes, which were related to fetal outcomes in a sex-dependent manner, suggesting that the placenta may mediate the consequence of LP-Alc-altered litter size on fetal development. CONCLUSIONS Our data indicate that the placenta is strongly involved in the fetal stress response and adapts in a sex-dependent fashion to support fetal development under the alcohol stressor. These variables may further influence the spectrum of intrauterine growth outcomes observed in those diagnosed with fetal alcohol spectrum disorder.
Collapse
|
23
|
Harp KLH, Bunting AM. The Racialized Nature of Child Welfare Policies and the Social Control of Black Bodies. SOCIAL POLITICS 2020; 27:258-281. [PMID: 32714000 PMCID: PMC7372952 DOI: 10.1093/sp/jxz039] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Black women are disproportionately involved in the child welfare system. This state-level intervention occurs at two levels-a higher likelihood of being (i) screened for drug use during pregnancy and (ii) reported to child welfare authorities after delivery. Consequently, they face further enmeshment in state-systems, including custody loss and lower reunification odds. Using evidence from the past forty years of research and media reports, we argue that systemic forces and policies largely contribute to racial disproportionality in the child welfare system, and assert this state intervention serves as a mechanism to control black reproduction.
Collapse
Affiliation(s)
- Kathi L H Harp
- Department of Health Management and Policy, University of Kentucky College of Public Health, Lexington, KY, USA
| | - Amanda M Bunting
- Department of Sociology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
24
|
Sarmah S, Srivastava R, McClintick JN, Janga SC, Edenberg HJ, Marrs JA. Embryonic ethanol exposure alters expression of sox2 and other early transcripts in zebrafish, producing gastrulation defects. Sci Rep 2020; 10:3951. [PMID: 32127575 PMCID: PMC7054311 DOI: 10.1038/s41598-020-59043-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023] Open
Abstract
Ethanol exposure during prenatal development causes fetal alcohol spectrum disorder (FASD), the most frequent preventable birth defect and neurodevelopmental disability syndrome. The molecular targets of ethanol toxicity during development are poorly understood. Developmental stages surrounding gastrulation are very sensitive to ethanol exposure. To understand the effects of ethanol on early transcripts during embryogenesis, we treated zebrafish embryos with ethanol during pre-gastrulation period and examined the transcripts by Affymetrix GeneChip microarray before gastrulation. We identified 521 significantly dysregulated genes, including 61 transcription factors in ethanol-exposed embryos. Sox2, the key regulator of pluripotency and early development was significantly reduced. Functional annotation analysis showed enrichment in transcription regulation, embryonic axes patterning, and signaling pathways, including Wnt, Notch and retinoic acid. We identified all potential genomic targets of 25 dysregulated transcription factors and compared their interactions with the ethanol-dysregulated genes. This analysis predicted that Sox2 targeted a large number of ethanol-dysregulated genes. A gene regulatory network analysis showed that many of the dysregulated genes are targeted by multiple transcription factors. Injection of sox2 mRNA partially rescued ethanol-induced gene expression, epiboly and gastrulation defects. Additional studies of this ethanol dysregulated network may identify therapeutic targets that coordinately regulate early development.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Rajneesh Srivastava
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Jeanette N McClintick
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sarath C Janga
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Abstract
AbstractIt is well established that high-dose alcohol consumption during pregnancy increases the risk for a plethora of adverse offspring outcomes. These include neurodevelopmental, cognitive and social deficits, as well as psychiatric illnesses, such as depression and anxiety. However, much less evidence is available on the effects of low- and early-dose alcohol exposure on mental health outcomes, regardless of the accumulating evidence that mental health outcomes should be considered in the context of the Developmental Origins of Health and Disease hypothesis. This review will discuss the evidence that indicates low-dose and early prenatal alcohol exposure can increase the risk of mental illness in offspring and discuss the mechanistic pathways that may be involved.
Collapse
|
26
|
Wang R, Martin CD, Lei AL, Hausknecht KA, Ishiwari K, Richards JB, Haj-Dahmane S, Shen RY. Prenatal Ethanol Exposure Leads to Attention Deficits in Both Male and Female Rats. Front Neurosci 2020; 14:12. [PMID: 32038156 PMCID: PMC6992663 DOI: 10.3389/fnins.2020.00012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/08/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Prenatal ethanol exposure (PE) causes multiple behavioral and cognitive deficits, collectively referred to as fetal alcohol spectrum disorders (FASD). Studies show that 49-94% of FASD children exhibit attention deficits, even when they have normal IQs or lack severe facial deformities, suggesting that attention deficits could be caused by even moderate prenatal exposure to alcohol, of which the underlying neural mechanisms are still unclear. A valid rodent model could help elucidate this phenomenon. MATERIALS AND METHODS A second-trimester equivalent binge drinking PE model was utilized. Pregnant Sprague Dawley rats were administered with 15% (w/v) ethanol (6 g/kg/day, via gastric gavage) during gestational days 8-20, and their offspring were the subjects in the present study. A modified 2-choice reaction time (2-CRT) task was used to illustrate possible attention deficits, including increased action impulsivity and lapses of attention. Enhanced impulsivity was reflected by more premature responses while increased lapses of attention were manifested as more incorrect responses and/or greater variability of reaction time, demonstrated by more skewed distributions of reaction time. Ten-week-old male and female rats were tested for three sessions following 16-19 days of training. RESULTS Our PE paradigm caused no major teratogenic effects. PE led to increased impulsivity exhibited as greater premature responses and augmented lapses of attention shown by greater skewnesses of reaction time distributions, relative to controls. The deficits were observed in both PE male and female rats. Interestingly, in males, the attention deficits were detected only when the 2-CRT task was relatively difficult whereas in females they were detected even when the task was at a less demanding level. CONCLUSION We show that the binge drinking pattern of PE led to attention deficits in both sexes of rats even though no major teratogenic effects were observed. Therefore, this rodent model can be used to study neural mechanisms underlying attention deficits caused by PE and to explore effective intervention approaches for FASD.
Collapse
Affiliation(s)
- Ruixiang Wang
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Department of Psychology, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Connor D. Martin
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Anna L. Lei
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Kathryn A. Hausknecht
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Keita Ishiwari
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Jerry B. Richards
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
27
|
McQuire C, Daniel R, Hurt L, Kemp A, Paranjothy S. The causal web of foetal alcohol spectrum disorders: a review and causal diagram. Eur Child Adolesc Psychiatry 2020; 29:575-594. [PMID: 30648224 PMCID: PMC7250957 DOI: 10.1007/s00787-018-1264-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022]
Abstract
Foetal alcohol spectrum disorders (FASDs) are a leading cause of developmental disability. Prenatal alcohol use is the sole necessary cause of FASD, but it is not always sufficient. Multiple factors influence a child's susceptibility to FASD following prenatal alcohol exposure. Much of the FASD risk factor literature has been limited to discussions of association, rather than causation. While knowledge of predictor variables is important for identifying who is most at risk of FASD and for targeting interventions, causal knowledge is important for identifying effective mechanisms for prevention and intervention programmes. We conducted a systematic search and narrative synthesis of the evidence and used this to create a causal diagram (directed acyclic graph; DAG) to describe the causal pathways to FASD. Our results show that the aetiology of FASD is multifaceted and complex. FASD risk is determined by a range of lifestyle, sociodemographic, maternal, social, gestational, and genetic factors. The causal diagram that we present in this review provides a comprehensive summary of causal risk factors for FASD and can be used as a tool to inform data collection and statistical modelling strategies to minimise bias in future studies of FASD.
Collapse
Affiliation(s)
- Cheryl McQuire
- Population Health Sciences, Bristol Medical School, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol, BS8 2PS, UK.
| | - R. Daniel
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - L. Hurt
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - A. Kemp
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| | - S. Paranjothy
- Division of Population Medicine, Cardiff University, 3rd Floor, Neuadd Meirionnydd, Heath Park, Cardiff, CF14 4YS UK
| |
Collapse
|
28
|
Yin X, Li J, Li Y, Zou S. Maternal alcohol consumption and oral clefts: a meta-analysis. Br J Oral Maxillofac Surg 2019; 57:839-846. [DOI: 10.1016/j.bjoms.2019.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 08/16/2019] [Indexed: 11/16/2022]
|
29
|
Koren G, Cohen R. Quantifying fetal alcohol exposure by meconium fatty acid ethyl esters (FAEE); association with adverse fetal outcomes and population estimates of fetal alcohol exposure. Drug Metab Rev 2019; 51:524-532. [PMID: 31595802 DOI: 10.1080/03602532.2019.1671859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) describes the wide range of adverse physical, behavioral and cognitive effects resulting from ethanol exposure during embryonic and fetal development. Identification of children suffering from FASD is often difficult, as abuse of ethanol during pregnancy is a heavily stigmatized behavior that receives little prenatal screening attention in routine care. Over the last 3 decades, measurement of the ethanol metabolites fatty acid ethyl esters (FAEE) has emerged as a useful tool to detect in the neonatal period fetal alcohol exposure starting from mid gestation. This review aims at updating clinicians and researchers on the validity and utility of this biological marker in two aspects: The association with adverse fetal outcomes and in generating population estimates of fetal alcohol exposure.
Collapse
Affiliation(s)
- Gideon Koren
- Motherisk Israel Program, Division of Clinical Pharmacology, Shamir Hospital, Zrifin, Israel.,Adelson Faculty of Medicine, Ariel University, Ariel, Israel.,Department of Physiology/Pharmacology, Western University, Ontario, Canada
| | - Rana Cohen
- Motherisk Israel Program, Division of Clinical Pharmacology, Shamir Hospital, Zrifin, Israel
| |
Collapse
|
30
|
Scepanovic G, Stewart BA. Analysis of Drosophila nervous system development following an early, brief exposure to ethanol. Dev Neurobiol 2019; 79:780-793. [PMID: 31472090 DOI: 10.1002/dneu.22718] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/22/2019] [Accepted: 08/27/2019] [Indexed: 01/09/2023]
Abstract
The effects of ethanol on neural function and development have been studied extensively, motivated in part by the addictive properties of alcohol and the neurodevelopmental deficits that arise in children with fetal alcohol spectrum disorder (FASD). Absent from this research area is a genetically tractable system to study the effects of early ethanol exposure on later neurodevelopmental and behavioral phenotypes. Here, we used embryos of the fruit fly, Drosophila melanogaster, as a model system to investigate the neuronal defects that arise after an early exposure to ethanol. We found several disruptions of neural development and morphology following a brief ethanol exposure during embryogenesis and subsequent changes in larval behavior. Altogether, this study establishes a new system to examine the effects of alcohol exposure in embryos and the potential to conduct large-scale genetics screens to uncover novel factors that sensitize or protect neurons to the effects of alcohol.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Bryan A Stewart
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada.,Department of Biology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
31
|
Mattson SN, Bernes GA, Doyle LR. Fetal Alcohol Spectrum Disorders: A Review of the Neurobehavioral Deficits Associated With Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2019; 43:1046-1062. [PMID: 30964197 DOI: 10.1111/acer.14040] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/03/2019] [Indexed: 11/26/2022]
Abstract
In utero alcohol exposure can disrupt the development of the fetal brain and result in a wide range of neurobehavioral outcomes collectively known as fetal alcohol spectrum disorders (FASD). This paper provides a comprehensive review of the cognitive and behavioral outcomes of prenatal alcohol exposure, including domains of general intelligence, executive functioning, language development, learning and memory, adaptive functioning, academic performance, and concurrent psychopathology. In addition, the current status of the neurobehavioral profile of FASD and its potential as a diagnostic tool will be discussed.
Collapse
Affiliation(s)
- Sarah N Mattson
- Center for Behavioral Teratology and Department of Psychology, San Diego State University, San Diego, California
| | - Gemma A Bernes
- Center for Behavioral Teratology and Department of Psychology, San Diego State University, San Diego, California
| | - Lauren R Doyle
- Center for Behavioral Teratology and Department of Psychology, San Diego State University, San Diego, California
| |
Collapse
|
32
|
Théberge ET, Baker JA, Dubose C, Boyle JK, Balce K, Goldowitz D, Hamre KM. Genetic Influences on the Amount of Cell Death in the Neural Tube of BXD Mice Exposed to Acute Ethanol at Midgestation. Alcohol Clin Exp Res 2019; 43:439-452. [PMID: 30589433 DOI: 10.1111/acer.13947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) have a strong genetic component although the genes that underlie this are only beginning to be elucidated. In the present study, one of the most common phenotypes of FASD, cell death within the early developing neural tube, was examined across a genetic reference population in a reverse genetics paradigm with the goal of identifying genetic loci that could influence ethanol (EtOH)-induced apoptosis in the early developing neural tube. METHODS BXD recombinant inbred mice as well as the parental strains were used to evaluate genetic differences in EtOH-induced cell death after exposure on embryonic day 9.5. Dams were given either 5.8 g/kg EtOH or isocaloric maltose-dextrin in 2 doses via intragastric gavage. Embryos were collected 7 hours after the initial exposure and cell death evaluated via TUNEL staining in the brainstem and forebrain. Genetic loci were evaluated using quantitative trait locus (QTL) analysis at GeneNetwork.org. RESULTS Significant strain differences were observed in the levels of EtOH-induced cell death that were due to genetic effects and not confounding variables such as differences in developmental maturity or cell death kinetics. Comparisons between the 2 regions of the developing neural tube showed little genetic correlation with the QTL maps exhibiting no overlap. Significant QTLs were found on murine mid-chromosome 4 and mid-chromosome 14 only in the brainstem. Within these chromosomal loci, a number of interesting candidate genes were identified that could mediate this differential sensitivity including Nfia (nuclear factor I/A) and Otx2 (orthodenticle homeobox 2). CONCLUSIONS These studies demonstrate that the levels of EtOH-induced cell death occur in strain- and region-dependent manners. Novel QTLs on mouse Chr4 and Chr14 were identified that modulate the differential sensitivity to EtOH-induced apoptosis in the embryonic brainstem. The genes underlying these QTLs could identify novel molecular pathways that are critical in this phenotype.
Collapse
Affiliation(s)
- Emilie T Théberge
- Centre for Molecular Medicine and Therapeutics , British Columbia Children's Research Institution, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jessica A Baker
- Department of Anatomy and Neurobiology , University of Tennessee Health Science Center, Memphis, Tennessee
| | - Candis Dubose
- Department of Anatomy and Neurobiology , University of Tennessee Health Science Center, Memphis, Tennessee
| | - Julia K Boyle
- Centre for Molecular Medicine and Therapeutics , British Columbia Children's Research Institution, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristina Balce
- Centre for Molecular Medicine and Therapeutics , British Columbia Children's Research Institution, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dan Goldowitz
- Centre for Molecular Medicine and Therapeutics , British Columbia Children's Research Institution, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kristin M Hamre
- Department of Anatomy and Neurobiology , University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
33
|
Are We Justified in Introducing Carbon Monoxide Testing to Encourage Smoking Cessation in Pregnant Women? HEALTH CARE ANALYSIS 2018; 27:128-145. [DOI: 10.1007/s10728-018-0364-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
34
|
Sebastiani G, Borrás-Novell C, Casanova MA, Pascual Tutusaus M, Ferrero Martínez S, Gómez Roig MD, García-Algar O. The Effects of Alcohol and Drugs of Abuse on Maternal Nutritional Profile during Pregnancy. Nutrients 2018; 10:E1008. [PMID: 30072661 PMCID: PMC6116049 DOI: 10.3390/nu10081008] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/24/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
The consumption of alcohol and drugs of abuse among pregnant women has experienced a significant increase in the last decades. Suitable maternal nutritional status is crucial to maintain the optimal environment for fetal development but if consumption of alcohol or drugs of abuse disrupt the intake of nutrients, the potential teratogenic effects of these substances increase. Despite evidence of the importance of nutrition in addicted pregnant women, there is a lack of information on the effects of alcohol and drugs of abuse on maternal nutritional status; so, the focus of this review was to provide an overview on the nutritional status of addicted mothers and fetuses. Alcohol and drugs consumption can interfere with the absorption of nutrients, impairing the quality and quantity of proper nutrient and energy intake, resulting in malnutrition especially of micronutrients (vitamins, omega⁻3, folic acid, zinc, choline, iron, copper, selenium). When maternal nutritional status is compromised by alcohol and drugs of abuse the supply of essential nutrients are not available for the fetus; this can result in fetal abnormalities like Intrauterine Growth Restriction (IUGR) or Fetal Alcohol Spectrum Disorder (FASD). It is critical to find a strategy to reduce fetal physical and neurological impairment as a result of prenatal alcohol and drugs of abuse exposure combined with poor maternal nutrition. Prenatal nutrition interventions and target therapy are required that may reverse the development of such abnormalities.
Collapse
Affiliation(s)
- Giorgia Sebastiani
- Neonatology Unit, Hospital Clinic-Maternitat, ICGON, BCNatal, C/Sabino Arana 1, 08028 Barcelona, Spain.
| | - Cristina Borrás-Novell
- Neonatology Unit, Hospital Clinic-Maternitat, ICGON, BCNatal, C/Sabino Arana 1, 08028 Barcelona, Spain.
| | - Miguel Alsina Casanova
- Neonatology Unit, Hospital Clinic-Maternitat, ICGON, BCNatal, C/Sabino Arana 1, 08028 Barcelona, Spain.
| | - Mireia Pascual Tutusaus
- Neonatology Unit, Hospital Clinic-Maternitat, ICGON, BCNatal, C/Sabino Arana 1, 08028 Barcelona, Spain.
| | - Silvia Ferrero Martínez
- Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu, 08028 Barcelona, Spain.
| | - María Dolores Gómez Roig
- Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu, 08028 Barcelona, Spain.
| | - Oscar García-Algar
- Neonatology Unit, Hospital Clinic-Maternitat, ICGON, BCNatal, C/Sabino Arana 1, 08028 Barcelona, Spain.
| |
Collapse
|
35
|
McCormack C, Hutchinson D, Burns L, Youssef G, Wilson J, Elliott E, Allsop S, Najman J, Jacobs S, Rossen L, Olsson C, Mattick R. Maternal and partner prenatal alcohol use and infant cognitive development. Drug Alcohol Depend 2018; 185:330-338. [PMID: 29499553 DOI: 10.1016/j.drugalcdep.2017.12.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND Teratogenicity of heavy prenatal alcohol exposure is established, but uncertainty remains regarding the impact of moderate alcohol exposure on cognitive deficits in infants. Separating in utero effects from environmental confounding is a challenge for observational studies; consideration of alcohol use by partners as well as mothers may help clarify this. This study examined associations between prenatal alcohol use by both mothers and their partners and infant cognitive developmental outcomes at 12-months. METHODS Pregnant women (n = 1331) and their partners (n = 699) were recruited from antenatal clinics of three metropolitan public hospitals in Australia, and completed detailed interviews about alcohol consumptions throughout pregnancy. Infants were assessed with the Bayley Scales of Infant Development - Third edition (Bayley) at 12-months of age. RESULTS Alcohol use during pregnancy was reported by 65.7% of mothers and 84.1% of partners. Using multiple methods to adjust for confounding factors, no evidence for impaired cognitive ability associated with alcohol use by mothers or their partners was observed. Children born to women who drank low-levels of alcohol had slightly higher Bayley cognitive scores than those born to abstaining women. There was some evidence for an interaction between sociodemographic factors and prenatal alcohol exposure on infant cognitive outcomes. CONCLUSION This finding corroborates existing evidence to suggest there are no detrimental effects to infant cognitive development at 12-months of age following low-level prenatal alcohol exposure. Future prospective studies involving families of a broad range of backgrounds would be informative to clarify interaction between alcohol exposure and environmental factors on developmental outcomes.
Collapse
Affiliation(s)
- Clare McCormack
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia; Department of Psychiatry, Columbia University Medical Center, 622 West 168th St., New York, NY 10032, USA.
| | - Delyse Hutchinson
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia; Centre for Social and Early Emotional Development, Deakin University, 221 Burwood Hwy., Burwood, VIC 3125, Australia; Centre for Adolescent Health, Royal Children's Hospital, 50 Flemington Rd., Parkville, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, 50 Flemington Rd., Parkville, VIC 3052, Australia
| | - Lucy Burns
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia
| | - George Youssef
- Centre for Social and Early Emotional Development, Deakin University, 221 Burwood Hwy., Burwood, VIC 3125, Australia; Centre for Adolescent Health, Royal Children's Hospital, 50 Flemington Rd., Parkville, VIC 3052, Australia
| | - Judy Wilson
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia
| | - Elizabeth Elliott
- Paediatrics and Child Health, University of Sydney, NSW 2006, Australia
| | - Steve Allsop
- National Drug Research Institute, Curtin University, Kent St., Bentley, Perth 6102, Australia
| | - Jake Najman
- Queensland Alcohol and Drug Research Education Centre, University of Queensland, Brisbane, St. Lucia, QLD 4072, Australia
| | - Sue Jacobs
- Dept of Obstetrics, Royal Prince Alfred Hospital,Missenden Rd., Camperdown, NSW 2050, Australia
| | - Larissa Rossen
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia
| | - Craig Olsson
- Centre for Social and Early Emotional Development, Deakin University, 221 Burwood Hwy., Burwood, VIC 3125, Australia; Centre for Adolescent Health, Royal Children's Hospital, 50 Flemington Rd., Parkville, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, 50 Flemington Rd., Parkville, VIC 3052, Australia
| | - Richard Mattick
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St., Randwick, NSW 2031, Australia
| |
Collapse
|
36
|
Zarrei M, Hicks GG, Reynolds JN, Thiruvahindrapuram B, Engchuan W, Pind M, Lamoureux S, Wei J, Wang Z, Marshall CR, Wintle RF, Chudley AE, Scherer SW. Copy number variation in fetal alcohol spectrum disorder. Biochem Cell Biol 2018. [PMID: 29533680 DOI: 10.1139/bcb-2017-0241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is characterized by a combination of neurological, developmental, and congenital defects that may occur as a consequence of prenatal alcohol exposure. Earlier reports showed that large chromosomal anomalies may link to FASD. Here, we examined the prevalence and types of copy number variations (CNVs) in FASD cases previously diagnosed by a multidisciplinary FASD team in sites across Canada. We genotyped 95 children with FASD and 87 age-matched, typically developing controls on the Illumina Human Omni2.5 SNP (single nucleotide polymorphisms) array platform. We compared their CNVs with those of 10 851 population controls to identify rare CNVs (<0.1% frequency), which may include large unbalanced chromosomal abnormalities, that might be relevant to FASD. In 12/95 (13%) of the FASD cases, rare CNVs were found that impact potentially clinically relevant developmental genes, including the CACNA1H involved in epilepsy and autism, the 3q29 deletion disorder, and others. Our results show that a subset of children diagnosed with FASD have chromosomal deletions and duplications that may co-occur or explain the neurodevelopmental impairments in a diagnosed cohort of FASD individuals. Children suspected to have FASD with or without sentinel facial features of fetal alcohol syndrome and neurodevelopmental delays should potentially be evaluated by a clinical geneticist and possibly have genetic investigations as appropriate to exclude other etiologies.
Collapse
Affiliation(s)
- Mehdi Zarrei
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Geoffrey G Hicks
- b Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - James N Reynolds
- c Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada.,d Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Bhooma Thiruvahindrapuram
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Worrawat Engchuan
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Molly Pind
- b Regenerative Medicine Program, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Sylvia Lamoureux
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - John Wei
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Zhouzhi Wang
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Christian R Marshall
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Richard F Wintle
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada
| | - Albert E Chudley
- e Department of Pediatrics and Child Health, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.,f Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Stephen W Scherer
- a The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 04A, Canada.,g Department of Molecular Genetics and McLaughlin Centre, University of Toronto, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
37
|
Ophthalmologic Findings in Russian Children with Fetal Alcohol Syndrome. Eur J Ophthalmol 2018; 23:823-30. [DOI: 10.5301/ejo.5000296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2013] [Indexed: 11/20/2022]
|
38
|
Embryonic Ethanol Exposure Affects Early- and Late-Added Cardiac Precursors and Produces Long-Lasting Heart Chamber Defects in Zebrafish. TOXICS 2017; 5:toxics5040035. [PMID: 29194345 PMCID: PMC5750563 DOI: 10.3390/toxics5040035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 11/28/2022]
Abstract
Drinking mothers expose their fetuses to ethanol, which produces birth defects: craniofacial defects, cognitive impairment, sensorimotor disabilities and organ deformities, collectively termed as fetal alcohol spectrum disorder (FASD). Various congenital heart defects (CHDs) are present in FASD patients, but the mechanisms of alcohol-induced cardiogenesis defects are not completely understood. This study utilized zebrafish embryos and older larvae to understand FASD-associated CHDs. Ethanol-induced cardiac chamber defects initiated during embryonic cardiogenesis persisted in later zebrafish life. In addition, myocardial damage was recognizable in the ventricle of the larvae that were exposed to ethanol during embryogenesis. Our studies of the pathogenesis revealed that ethanol exposure delayed differentiation of first and second heart fields and reduced the number of early- and late-added cardiomyocytes in the heart. Ethanol exposure also reduced the number of endocardial cells. Together, this study showed that ethanol-induced heart defects were present in late-stage zebrafish larvae. Reduced numbers of cardiomyocytes partly accounts for the ethanol-induced zebrafish heart defects.
Collapse
|
39
|
Zhang P, Wang G, Lin Z, Wu Y, Zhang J, Liu M, Lee KKH, Chuai M, Yang X. Alcohol exposure induces chick craniofacial bone defects by negatively affecting cranial neural crest development. Toxicol Lett 2017; 281:53-64. [PMID: 28919490 DOI: 10.1016/j.toxlet.2017.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 10/25/2022]
Abstract
Excess alcohol consumption during pregnancy could lead to fetal alcohol syndrome (FAS). However, the molecular mechanism leading to craniofacial abnormality, a feature of FAS, is still poorly understood. The cranial neural crest cells (NCCs) contribute to the formation of the craniofacial bones. Therefore, NCCs exposed to ethanol was investigated - using chick embryos and in vitro explant culture as experimental models. We demonstrated that exposure to 2% ethanol induced craniofacial defects, which includes parietal defect, in the developing chick fetus. Immunofluorescent staining revealed that ethanol treatment downregulated Ap-2ɑ, Pax7 and HNK-1 expressions by cranial NCCs. Using double-immunofluorescent stainings for Ap-2ɑ/pHIS3 and Ap-2ɑ/c-Caspase3, we showed that ethanol treatment inhibited cranial NCC proliferation and increased NCC apoptosis, respectively. Moreover, ethanol treatment of the dorsal neuroepithelium increased Laminin, N-Cadherin and Cadherin 6B expressions while Cadherin 7 expression was repressed. In situ hybridization also revealed that ethanol treatment up-regulated Cadherin 6B expression but down-regulated slug, Msx1, FoxD3 and BMP4 expressions. In summary, our experimental results demonstrated that ethanol treatment interferes with the production of cranial NCCs by affecting the proliferation and apoptosis of these cells. In addition, ethanol affected the delamination, epithelial-mesenchymal transition (EMT) and cell migration of cranial NCCs, which may have contributed to the etiology of the craniofacial defects.
Collapse
Affiliation(s)
- Ping Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Zhuangling Lin
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Yushi Wu
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Jing Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Meng Liu
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Kenneth Ka Ho Lee
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee, DD1 5EH, UK
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
40
|
Balaraman S, Idrus NM, Miranda RC, Thomas JD. Postnatal choline supplementation selectively attenuates hippocampal microRNA alterations associated with developmental alcohol exposure. Alcohol 2017; 60:159-167. [PMID: 28433422 PMCID: PMC5559286 DOI: 10.1016/j.alcohol.2016.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 11/25/2022]
Abstract
Prenatal alcohol exposure can result in a range of physical, neuropathological, and behavioral alterations, collectively termed fetal alcohol spectrum disorders (FASD). We have shown that supplementation with the nutrient choline reduces the severity of developmental alcohol-associated deficits in hippocampal-dependent behaviors and normalizes some aspects of hippocampal cholinergic development and DNA methylation patterns. Alcohol's developmental effects may also be mediated, in part, by altering microRNAs (miRNAs) that serve as negative regulators of gene translation. To determine whether choline supplementation alters ethanol's long-lasting effects on miRNAs, Sprague-Dawley rats were exposed to 5.25 g/kg/day ethanol from postnatal days (PD) 4-9 via intubation; controls received sham intubations. Subjects were treated with choline chloride (100 mg/kg/day) or saline vehicle subcutaneously (s.c.) from PD 4-21. On PD 22, subjects were sacrificed, and RNA was isolated from the hippocampus. MiRNA expression was assessed with TaqMan Human MicroRNA Panel Low-Density Arrays. Ethanol significantly increased miRNA expression variance, an effect that was attenuated with choline supplementation. Cluster analysis of stably expressed miRNAs that exceeded an ANOVA p < 0.05 criterion indicated that for both male and female offspring, control and ethanol-exposed groups were most dissimilar from each other, with choline-supplemented groups in between. MiRNAs that expressed an average 2-fold change due to ethanol exposure were further analyzed to identify which ethanol-sensitive miRNAs were protected by choline supplementation. We found that at a false discovery rate (FDR)-adjusted criterion of p < 0.05, miR-200c was induced by ethanol exposure and that choline prevented this effect. Collectively, our data show that choline supplementation can normalize disturbances in miRNA expression following developmental alcohol exposure and can protect specific miRNAs from induction by ethanol. These findings have important implications for the mechanisms by which choline may serve as a potential treatment for FASD.
Collapse
Affiliation(s)
- Sridevi Balaraman
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Nirelia M Idrus
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, San Diego, CA 92120, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Women's Health in Neuroscience Program, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, Department of Psychology, San Diego State University, San Diego, CA 92120, USA.
| |
Collapse
|
41
|
Popova S, Lange S, Probst C, Gmel G, Rehm J. Estimation of national, regional, and global prevalence of alcohol use during pregnancy and fetal alcohol syndrome: a systematic review and meta-analysis. Lancet Glob Health 2017; 5:e290-e299. [PMID: 28089487 DOI: 10.1016/s2214-109x(17)30021-9] [Citation(s) in RCA: 555] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alcohol use during pregnancy is the direct cause of fetal alcohol syndrome (FAS). We aimed to estimate the prevalence of alcohol use during pregnancy and FAS in the general population and, by linking these two indicators, estimate the number of pregnant women that consumed alcohol during pregnancy per one case of FAS. METHODS We began by doing two independent comprehensive systematic literature searches using multiple electronic databases for original quantitative studies that reported the prevalence in the general population of the respective country of alcohol use during pregnancy published from Jan 1, 1984, to June 30, 2014, or the prevalence of FAS published from Nov 1, 1973, to June 30, 2015, in a peer-reviewed journal or scholarly report. Each study on the prevalence of alcohol use during pregnancy was critically appraised using a checklist for observational studies, and each study on the prevalence of FAS was critically appraised by use of a method specifically designed for systematic reviews addressing questions of prevalence. Studies on the prevalence of alcohol use during pregnancy and/or FAS were omitted if they used a sample population not generalisable to the general population of the respective country, reported a pooled estimate by combining several studies, or were published in iteration. Studies that excluded abstainers were also omitted for the prevalence of alcohol use during pregnancy. We then did country-specific random-effects meta-analyses to estimate the pooled prevalence of these indicators. For countries with one or no empirical studies, we predicted prevalence of alcohol use during pregnancy using fractional response regression modelling and prevalence of FAS using a quotient of the average number of women who consumed alcohol during pregnancy per one case of FAS. We used Monte Carlo simulations to derive confidence intervals for the country-specific point estimates of the prevalence of FAS. We estimated WHO regional and global averages of the prevalence of alcohol use during pregnancy and FAS, weighted by the number of livebirths per country. The review protocols for the prevalence of alcohol use during pregnancy (CRD42016033835) and FAS (CRD42016033837) are available on PROSPERO. FINDINGS Of 23 470 studies identified for the prevalence of alcohol use, 328 studies were retained for systematic review and meta-analysis; the search strategy for the prevalence of FAS yielded 11 110 studies, of which 62 were used in our analysis. The global prevalence of alcohol use during pregnancy was estimated to be 9·8% (95% CI 8·9-11·1) and the estimated prevalence of FAS in the general population was 14·6 per 10 000 people (95% CI 9·4-23·3). We also estimated that one in every 67 women who consumed alcohol during pregnancy would deliver a child with FAS, which translates to about 119 000 children born with FAS in the world every year. INTERPRETATION Alcohol use during pregnancy is common in many countries and as such, FAS is a relatively prevalent alcohol-related birth defect. More effective prevention strategies targeting alcohol use during pregnancy and surveillance of FAS are urgently needed. FUNDING Centre for Addiction and Mental Health (no external funding was sought).
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Shannon Lange
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Charlotte Probst
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, ON, Canada; Epidemiological Research Unit, Klinische Psychologie and Psychotherapie, Technische Universität Dresden, Dresden, Germany
| | - Gerrit Gmel
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, ON, Canada; School of Electrical Engineering and Telecommunications, Faculty of Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Jürgen Rehm
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, ON, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada; Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Epidemiological Research Unit, Klinische Psychologie and Psychotherapie, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
42
|
McCormack C, Hutchinson D, Burns L, Wilson J, Elliott E, Allsop S, Najman J, Jacobs S, Rossen L, Olsson C, Mattick R. Prenatal Alcohol Consumption Between Conception and Recognition of Pregnancy. Alcohol Clin Exp Res 2017; 41:369-378. [PMID: 28116821 DOI: 10.1111/acer.13305] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 11/21/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Current estimates of the rates of alcohol-exposed pregnancies may underestimate prenatal alcohol exposure if alcohol consumption in early trimester 1, prior to awareness of pregnancy, is not considered. Extant literature describes predictors of alcohol consumption during pregnancy; however, alcohol consumption prior to awareness of pregnancy is a distinct behavior from consumption after becoming aware of pregnancy and thus may be associated with different predictors. The purpose of this study was therefore to examine prevalence and predictors of alcohol consumption by women prior to awareness of their pregnancy, and trajectories of change to alcohol use following pregnancy recognition. METHODS Pregnant women (n = 1,403) were prospectively recruited from general antenatal clinics of 4 public hospitals in Australian metropolitan areas between 2008 and 2013. Women completed detailed interviews about alcohol use before and after recognition of pregnancy. RESULTS Most women (n = 850, 60.6%) drank alcohol between conception and pregnancy recognition. Binge and heavy drinking were more prevalent than low-level drinking. The proportion of women who drank alcohol reduced to 18.3% (n = 257) after recognition of pregnancy. Of women who drank alcohol, 70.5% ceased drinking, 18.3% reduced consumption, and 11.1% made no reduction following awareness of pregnancy. Socioeconomic status (SES) was the strongest predictor of alcohol use, with drinkers more likely to be of high rather than low SES compared with abstainers (OR = 3.30, p < 0.001). Factors associated with different trajectories (either cessation, reduction, or continuation of drinking) included level of alcohol use prior to pregnancy recognition, age, pregnancy planning, and illicit substance use. CONCLUSIONS In this sample of relatively high SES women, most women ceased or reduced drinking once aware of their pregnancy. However, the rate of alcohol-exposed pregnancies was higher than previous estimates when the period prior to pregnancy recognition was taken into account.
Collapse
Affiliation(s)
- Clare McCormack
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia.,Australian Centre for Perinatal Science, University of New South Wales, Sydney, New South Wales, Australia.,Department of Psychiatry, Columbia University, New York, New York
| | - Delyse Hutchinson
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia.,Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Deakin University, Burwood, Victoria, Australia.,Department of Paediatrics, The Murdoch Childrens Research Institute and The University of Melbourne, Royal Children's Hospital, Victoria, Australia
| | - Lucy Burns
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Judy Wilson
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Elizabeth Elliott
- Paediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| | - Steve Allsop
- National Drug Research Institute, Curtin University, Perth, Western Australia, Australia
| | - Jake Najman
- Queensland Alcohol & Drug Research Education Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Sue Jacobs
- Department of Obstetrics, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Larissa Rossen
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Craig Olsson
- Centre for Social and Early Emotional Development, School of Psychology, Faculty of Health, Deakin University, Burwood, Victoria, Australia.,Department of Paediatrics, The Murdoch Childrens Research Institute and The University of Melbourne, Royal Children's Hospital, Victoria, Australia
| | - Richard Mattick
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Germanaud D, Toutain S. Exposition prénatale à l’alcool et troubles causés par l’alcoolisation fœtale. ACTA ACUST UNITED AC 2017. [DOI: 10.3917/cont.046.0039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
44
|
Goodlett CR, Horn KH, Zhou FC. Alcohol Teratogenesis: Mechanisms of Damage and Strategies for Intervention. Exp Biol Med (Maywood) 2016; 230:394-406. [PMID: 15956769 DOI: 10.1177/15353702-0323006-07] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There are multiple mechanisms by which alcohol can damage the developing brain, but the type of damage induced will depend on the amount and developmental timing of exposure, along with other maternal and genetic factors. This article reviews current perspectives on how ethanol can produce neuroteratogenic effects by its interactions with molecular regulators of brain development. The current evidence suggests that alcohol produces many of its damaging effects by exerting specific actions on molecules that regulate key developmental processes (e.g., L1 cell adhesion molecule, alcohol dehydrogenase, catalase), interfering with the early development of midline serotonergic neurons and disrupting their regulatory-signaling function for other target brain structures, interfering with trophic factors that regulate neurogenesis and cell survival, or inducing excessive cell death via oxidative stress or activation of caspase-3 proteases. The current understanding of pathogenesis mechanisms suggests several strategic approaches to develop rational molecular prevention. However, the development of behavioral and biologic treatments for alcohol-affected children is crucial because it is unlikely that effective delivery of preventative interventions can realistically be achieved in ways to prevent prenatal damage in at-risk pregnancies. Toward that end, behavioral training that promotes experience-dependent neuroplasticity has been effective in a rat model of cerebellar damage induced by alcohol exposure during the period of brain development that is comparable to that of the human third trimester.
Collapse
Affiliation(s)
- Charles R Goodlett
- Department of Psychology, Indiana University-Purdue University at Indianapolis, 402 North Blackford Street, Indianapolis, Indiana 46202, USA.
| | | | | |
Collapse
|
45
|
Sarmah S, Muralidharan P, Marrs JA. Common congenital anomalies: Environmental causes and prevention with folic acid containing multivitamins. ACTA ACUST UNITED AC 2016; 108:274-286. [PMID: 27718306 DOI: 10.1002/bdrc.21138] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Congenital anomalies, congenital defects, or birth defects are significant causes of death in infants. The most common congenital defects are congenital heart defects (CHDs) and neural tube defects (NTDs). Defects induced by genetic mutations, environmental exposure to toxins, or a combination of these effects can result in congenital malformations, leading to infant death or long-term disabilities. These defects produce significant mortality and morbidity in the affected individuals, and families are affected emotional and financially. Also, society is impacted on many levels. Congenital anomalies may be reduced by dietary supplements of folic acid and other vitamins. Here, we review the evidence for specific roles of toxins (alcohol, cigarette smoke) in causing common severe congenital anomalies like CHDs, NTDs, and ocular defects. We also review the evidence for beneficial effects for dietary supplementation, and highlight gaps in our knowledge, where research may contribute to additional benefits of intervention that can reduce birth defects. Extensive discussion of common severe congenital anomalies (CHDs, NTDs, and ocular defects) illustrates the effects of diet on the frequency and severity of these defects. Birth Defects Research (Part C) 108:274-286, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202.
| | - Pooja Muralidharan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, 46202.
| |
Collapse
|
46
|
Li S, Wang G, Gao LR, Lu WH, Wang XY, Chuai M, Lee KKH, Cao L, Yang X. Autophagy is involved in ethanol-induced cardia bifida during chick cardiogenesis. Cell Cycle 2016; 14:3306-17. [PMID: 26317250 DOI: 10.1080/15384101.2015.1087621] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Excess alcohol consumption during pregnancy has been acknowledged to increase the incidence of congenital disorders, especially the cardiovascular system. However, the mechanism involved in ethanol-induced cardiac malformation in prenatal fetus is still unknown. We demonstrated that ethanol exposure during gastrulation in the chick embryo increased the incidence of cardia bifida. Previously, we reported that autophagy was involved in heart tube formation. In this context, we demonstrated that ethanol exposure increased ATG7 and LC3 expression. mTOR was found to be inhibited by ethanol exposure. We activated autophagy using exogenous rapamycin (RAPA) and observed that it induced cardiac bifida and increased GATA5 expression. RAPA beads implantation experiments revealed that RAPA restricted ventricular myosin heavy chain (VMHC) expression. In vitro explant cultures of anterior primitive streak demonstrated that both ethanol and RAPA treatments could reduce cell differentiation and the spontaneous beating of cardiac precursor cells. In addition, the bead experiments showed that RAPA inhibited GATA5 expression during heart tube formation. Semiquantitative RT-PCR analysis indicated that BMP2 expression was increased while GATA4 expression was suppressed. In the embryos exposed to excess ethanol, BMP2, GATA4 and FGF8 expression was repressed. These genes are associated with cardiomyocyte differentiation, while heart tube fusion is associated with increased Wnt3a but reduced VEGF and Slit2 expression. Furthermore, the ethanol exposure also caused the production of excess ROS, which might damage the cardiac precursor cells of developing embryos. In sum, our results revealed that disrupting autophagy and excess ROS generation are responsible for inducing abnormal cardiogenesis in ethanol-treated chick embryos.
Collapse
Affiliation(s)
- Shuai Li
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| | - Guang Wang
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| | - Lin-Rui Gao
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| | - Wen-Hui Lu
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| | - Xiao-Yu Wang
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| | - Manli Chuai
- b Division of Cell and Developmental Biology ; University of Dundee ; Dundee , UK
| | - Kenneth Ka Ho Lee
- d Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong ; Shatin , Hong Kong
| | - Liu Cao
- c Key Laboratory of Medical Cell Biology, China Medical University ; Shenyang , China
| | - Xuesong Yang
- a Division of Histology and Embryology ; Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University ; Guangzhou , China
| |
Collapse
|
47
|
Li X, Gao A, Wang Y, Chen M, Peng J, Yan H, Zhao X, Feng X, Chen D. Alcohol exposure leads to unrecoverable cardiovascular defects along with edema and motor function changes in developing zebrafish larvae. Biol Open 2016; 5:1128-33. [PMID: 27422904 PMCID: PMC5004616 DOI: 10.1242/bio.019497] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Maternal alcohol consumption during pregnancy can cause a series of developmental disorders in the fetus called FAS (fetal alcohol syndrome). In the present study we exposed zebrafish embryos to 1% and 2% alcohol and observed the morphology of heart and blood vessels during and after exposure to investigate motor function alterations, and damage and recovery to the cardiovascular system. The results showed that alcohol exposure could induce heart deformation, slower heart rate, and incomplete blood vessels and pericardium. After stopping exposure, larvae exposed to 1% alcohol could recover only in heart morphology, but larvae in 2% alcohol could not recover either morphology or function of cardiovascular system. The edema-like characteristics in the 2% alcohol group became more conspicuous afterwards, with destruction in the dorsal aorta, coarctation in segmental arteries and a decrease in motor function, implying more serious unrecoverable cardiovascular defects in the 2% group. The damaged blood vessels in the 2% alcohol group resulted in an alteration in permeability and a decrease of blood volume, which were the causes of edema in pathology. These findings contribute towards a better understanding of ethanol-induced cardiovascular abnormalities and co-syndrome in patients with FAS, and warns against excessive maternal alcohol consumption during pregnancy.
Collapse
Affiliation(s)
- Xu Li
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Aiai Gao
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yanan Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China
| | - Man Chen
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jun Peng
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Huaying Yan
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xin Zhao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin 300071, China
| | - Xizeng Feng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University, Tianjin 300071, China
| | - Dongyan Chen
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
48
|
Li ZY, Ma ZL, Lu WH, Cheng X, Chen JL, Song XY, Chuai M, Lee KKH, Yang X. Ethanol exposure represses osteogenesis in the developing chick embryo. Reprod Toxicol 2016; 62:53-61. [DOI: 10.1016/j.reprotox.2016.04.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 04/07/2016] [Accepted: 04/21/2016] [Indexed: 01/02/2023]
|
49
|
Fan J, Jacobson SW, Taylor PA, Molteno CD, Dodge NC, Stanton ME, Jacobson JL, Meintjes EM. White matter deficits mediate effects of prenatal alcohol exposure on cognitive development in childhood. Hum Brain Mapp 2016; 37:2943-58. [PMID: 27219850 DOI: 10.1002/hbm.23218] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 04/01/2016] [Accepted: 04/05/2016] [Indexed: 11/09/2022] Open
Abstract
Fetal alcohol spectrum disorders comprise the spectrum of cognitive, behavioral, and neurological impairments caused by prenatal alcohol exposure (PAE). Diffusion tensor imaging (DTI) was performed on 54 children (age 10.1 ± 1.0 years) from the Cape Town Longitudinal Cohort, for whom detailed drinking histories obtained during pregnancy are available: 26 with full fetal alcohol syndrome (FAS) or partial FAS (PFAS), 15 nonsyndromal heavily exposed (HE), and 13 controls. Using voxelwise analyses, children with FAS/PFAS showed significantly lower fractional anisotropy (FA) in four white matter (WM) regions and higher mean diffusivity (MD) in seven; three regions of FA and MD differences (left inferior longitudinal fasciculus (ILF), splenium, and isthmus) overlapped, and the fourth FA cluster was located in the same WM bundle (right ILF) as an MD cluster. HE children showed lower FA and higher MD in a subset of these regions. Significant correlations were observed between three continuous alcohol measures and DTI values at cluster peaks, indicating that WM damage in several regions is dose dependent. Lower FA in the regions of interest was attributable primarily to increased radial diffusivity rather than decreased axonal diffusivity, suggesting poorer axon packing density and/or myelination. Multiple regression models indicated that this cortical WM impairment partially mediated adverse effects of PAE on information processing speed and eyeblink conditioning. Hum Brain Mapp 37:2943-2958, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jia Fan
- MRC/UCT Medical Imaging Research Unit, University of Cape Town, Cape Town, South Africa.,Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Sandra W Jacobson
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.,Department of Psychiatry and Mental Health, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Paul A Taylor
- MRC/UCT Medical Imaging Research Unit, University of Cape Town, Cape Town, South Africa.,Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.,African Institute for Mathematical Sciences, Muizenberg, Western Cape, South Africa
| | - Christopher D Molteno
- Department of Psychiatry and Mental Health, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Neil C Dodge
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Mark E Stanton
- Department of Psychology, University of Delaware, Newark, Delaware
| | - Joseph L Jacobson
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.,Department of Psychiatry and Mental Health, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Ernesta M Meintjes
- MRC/UCT Medical Imaging Research Unit, University of Cape Town, Cape Town, South Africa.,Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| |
Collapse
|
50
|
Eberhart JK, Parnell SE. The Genetics of Fetal Alcohol Spectrum Disorders. Alcohol Clin Exp Res 2016; 40:1154-65. [PMID: 27122355 DOI: 10.1111/acer.13066] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/04/2016] [Indexed: 12/29/2022]
Abstract
The term "fetal alcohol spectrum disorders" (FASD) defines the full range of ethanol (EtOH)-induced birth defects. Numerous variables influence the phenotypic outcomes of embryonic EtOH exposure. Among these variables, genetics appears to play an important role, yet our understanding of the genetic predisposition to FASD is still in its infancy. We review the current literature that relates to the genetics of FASD susceptibility and gene-EtOH interactions. Where possible, we comment on potential mechanisms of reported gene-EtOH interactions. Early indications of genetic sensitivity to FASD came from human and animal studies using twins or inbred strains, respectively. These analyses prompted searches for susceptibility loci involved in EtOH metabolism and analyses of candidate loci, based on phenotypes observed in FASD. More recently, genetic screens in animal models have provided an additional insight into the genetics of FASD. Understanding FASD requires that we understand the many factors influencing phenotypic outcome following embryonic EtOH exposure. We are gaining ground on understanding some of the genetics behind FASD, yet much work remains to be carried out. Coordinated analyses using human patients and animal models are likely to be highly fruitful in uncovering the genetics behind FASD.
Collapse
Affiliation(s)
- Johann K Eberhart
- Department of Molecular Biosciences, Institute for Cell and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|