1
|
Klem JR, Schwantes-An TH, Abreu M, Suttie M, Gray R, Vo H, Conley G, Foroud TM, Wetherill L, Lovely CB. Mutations in the Bone Morphogenetic Protein signaling pathway sensitize zebrafish and humans to ethanol-induced jaw malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.28.546932. [PMID: 37425959 PMCID: PMC10327032 DOI: 10.1101/2023.06.28.546932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describe ethanol-induced developmental defects including craniofacial malformations. While ethanol-sensitive genetic mutations contribute to facial malformations, the impacted cellular mechanisms remain unknown. Bmp signaling is a key regulator of epithelial morphogenesis driving facial development, providing a possible ethanol-sensitive mechanism. We found that zebrafish mutants for Bmp signaling components are ethanol-sensitive and affect anterior pharyngeal endoderm shape and gene expression, indicating ethanol-induced malformations of the anterior pharyngeal endoderm cause facial malformations. Integrating FASD patient data, we provide the first evidence that variants in the human Bmp receptor gene BMPR1B associate with ethanol-related differences in jaw volume. Our results show that ethanol exposure disrupts proper morphogenesis of, and tissue interactions between, facial epithelia that mirror overall viscerocranial shape changes and are predictive for Bmp-ethanol associations in human jaw development. Our data provide a mechanistic paradigm linking ethanol to disrupted epithelial cell behaviors that underlie facial defects in FASD. Summary Statement In this study, we apply a unique combination of zebrafish-based approaches and human genetic and facial dysmorphology analyses to resolve the cellular mechanisms driven by the ethanol-sensitive Bmp pathway.
Collapse
|
2
|
Fish EW, Boschen KE, Parnell SE. Efcab7 deletion sensitizes mice to the teratogenic effects of gastrulation-stage alcohol exposure. Reprod Toxicol 2024; 130:108729. [PMID: 39366525 DOI: 10.1016/j.reprotox.2024.108729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/06/2024]
Abstract
Alcohol exposure during the gastrulation stage of development can disrupt Sonic hedgehog (Shh) signaling and cause eye, craniofacial, and brain defects. One of the genes that regulates Shh signaling is Efcab7, which encodes a protein that facilitates the actions of Smoothened (Smo), a critical component of the Shh pathway. Previous work from our lab has demonstrated that Efcab7 is differentially expressed between two sub-strains of C57BL/6 mice that differ in their sensitivity to gastrulation-stage alcohol exposure. The more alcohol-sensitive C57BL/6 J mice express lower levels of Efcab7 during gastrulation than do the less alcohol-sensitive C57BL/6NHsd mice. The current study examined whether partial or full Efcab7 deletions render mice more sensitive to gastrulation-stage alcohol exposure and affect the sensitivity to other modulators of Shh signaling that cause craniofacial malformations. Efcab7+/- dams were mated with Efcab7+/- sires to produce Efcab7+/+, Efcab7+/-, and Efcab7-/- fetuses. On gestational day 7 (GD 7), they received either alcohol (two doses of 2.9 g/kg, i.p., given 4 hours apart), the Smo antagonist vismodegib (40 mg/kg, or vehicle, p.o.), the Smo agonist SAG (20 mg/kg) or the appropriate vehicles. GD 17 fetuses were collected and examined for ocular and craniofacial dysmorphology. As compared to Efcab7+/+ fetuses, Efcab7-/- fetuses exposed to alcohol or vismodegib treatment had more severe ocular and craniofacial malformations. In contrast, Efcab7-/- fetuses had less severe malformations induced by SAG. These results confirm that Efcab7 can modify responses to Shh agonists and antagonists and further identify Efcab7 as a gene important for the sensitivity to gastrulation-stage alcohol exposure.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
| | - Karen E Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, United States; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States; Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
3
|
Antón-Bolaños N, Faravelli I, Faits T, Andreadis S, Kastli R, Trattaro S, Adiconis X, Wei A, Sampath Kumar A, Di Bella DJ, Tegtmeyer M, Nehme R, Levin JZ, Regev A, Arlotta P. Brain Chimeroids reveal individual susceptibility to neurotoxic triggers. Nature 2024; 631:142-149. [PMID: 38926573 PMCID: PMC11338177 DOI: 10.1038/s41586-024-07578-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/17/2024] [Indexed: 06/28/2024]
Abstract
Interindividual genetic variation affects the susceptibility to and progression of many diseases1,2. However, efforts to study how individual human brains differ in normal development and disease phenotypes are limited by the paucity of faithful cellular human models, and the difficulty of scaling current systems to represent multiple people. Here we present human brain Chimeroids, a highly reproducible, multidonor human brain cortical organoid model generated by the co-development of cells from a panel of individual donors in a single organoid. By reaggregating cells from multiple single-donor organoids at the neural stem cell or neural progenitor cell stage, we generate Chimeroids in which each donor produces all cell lineages of the cerebral cortex, even when using pluripotent stem cell lines with notable growth biases. We used Chimeroids to investigate interindividual variation in the susceptibility to neurotoxic triggers that exhibit high clinical phenotypic variability: ethanol and the antiepileptic drug valproic acid. Individual donors varied in both the penetrance of the effect on target cell types, and the molecular phenotype within each affected cell type. Our results suggest that human genetic background may be an important mediator of neurotoxin susceptibility and introduce Chimeroids as a scalable system for high-throughput investigation of interindividual variation in processes of brain development and disease.
Collapse
Affiliation(s)
- Noelia Antón-Bolaños
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Irene Faravelli
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tyler Faits
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sophia Andreadis
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Rahel Kastli
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sebastiano Trattaro
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xian Adiconis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anqi Wei
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Abhishek Sampath Kumar
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniela J Di Bella
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew Tegtmeyer
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ralda Nehme
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Genentech, San Francisco, CA, USA
| | - Paola Arlotta
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
Bhadsavle SS, Scaturro KZ, Golding MC. Maternal 129S1/SvImJ background attenuates the placental phenotypes induced by chronic paternal alcohol exposure. Reprod Toxicol 2024; 126:108605. [PMID: 38735594 DOI: 10.1016/j.reprotox.2024.108605] [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: 01/04/2024] [Revised: 04/17/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024]
Abstract
Paternal alcohol use is emerging as a plausible driver of alcohol-related growth and patterning defects. Studies from our lab using an inbred C57Bl/6 J mouse model suggest that these paternally-inherited phenotypes result from paternally programmed deficits in the formation and function of the placenta. The 129S1/SvImJ genetic background is typically more susceptible to fetoplacental growth defects due to strain-specific differences in placental morphology. We hypothesized that these placental differences would sensitize 129S1/SvImJ-C57Bl/6 J hybrid offspring to paternally-inherited fetoplacental growth phenotypes induced by paternal alcohol exposure. Using a limited access model, we exposed C57Bl/6 J males to alcohol and bred them to naïve 129S1/SvImJ dams. We then assayed F1 hybrid offspring for alterations in fetoplacental growth and used micro-CT imaging to contrast placental histological patterning between the preconception treatments. F1 hybrid placentae exhibit larger placental weights than pure C57Bl/6 J offspring but display a proportionally smaller junctional zone with increased glycogen content. The male F1 hybrid offspring of alcohol-exposed sires exhibit modest placental hyperplasia but, unlike pure C57Bl/6 J offspring, do not display observable changes in placental histology, glycogen content, or measurable impacts on fetal growth. Although F1 hybrid female offspring do not exhibit any measurable alterations in fetoplacental growth, RT-qPCR analysis of placental gene expression reveals increased expression of genes participating in the antioxidant response. The reduced placental junctional zone but increased glycogen stores of 129S1/SvImJ-C57Bl/6 J F1 hybrid placentae ostensibly attenuate the previously observed placental patterning defects and fetal growth restriction induced by paternal alcohol use in the C57Bl/6 J strain.
Collapse
Affiliation(s)
- Sanat S Bhadsavle
- Department of Veterinary Physiology & Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Katherine Z Scaturro
- Department of Veterinary Physiology & Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Michael C Golding
- Department of Veterinary Physiology & Pharmacology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
5
|
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
|
6
|
May PA, Hasken JM, de Vries MM, Marais AS, Abdul-Rahman O, Robinson LK, Adam MP, Manning MA, Kalberg WO, Buckley D, Seedat S, Parry CD, Hoyme HE. Maternal and paternal risk factors for fetal alcohol spectrum disorders: Alcohol and other drug use as proximal influences. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:2090-2109. [PMID: 38226752 PMCID: PMC10792253 DOI: 10.1111/acer.15193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 01/17/2024]
Abstract
OBJECTIVE To explore and analyze the significance of proximal influences of maternal and paternal traits associated with bearing a child with a fetal alcohol spectrum disorder (FASD). METHODS Aggregated, maternal interview-collected data (N = 2515) concerning alcohol, tobacco, and other drug use were examined to determine risk for FASD from seven cross-sectional samples of mothers of first-grade students who were evaluated for a possible diagnosis of FASD. RESULTS Mothers of children with fetal alcohol syndrome (FAS) reported the highest alcohol use throughout pregnancy, proportion of binge drinking, drinks per drinking day (DDD), drinking days per week, and total drinks per week. Mothers of children with FAS also consumed significantly more alcohol than mothers of children with partial FAS (PFAS), alcohol-related neurodevelopmental disorder (ARND), or typically developing controls. Mothers of children with PFAS and ARND reported similar drinking patterns, which exposed fetuses to 3-4 times more alcohol than mothers of controls, but the PFAS group was more likely than the ARND group to abstain in latter trimesters. Fathers of all children were predominantly drinkers (70%-85%), but more fathers of children with FASD binged heavily on more days than fathers of controls. Compared to the few mothers of controls who used alcohol during pregnancy, the ARND group binge drank more (3+ DDD) throughout pregnancy and drank more DDD before pregnancy and first trimester. Regression analysis, controlling for tobacco use, indicated that mothers who reported drinking <1 DDD were significantly more likely than abstainers to bear a child with FASD (OR = 2.75) as were those reporting higher levels such as 5-5.9 DDD (OR = 32.99). Exclusive, first-trimester maternal drinking increased risk for FASD five times over that of abstinence (p < 0.001, OR = 5.05, 95% CI: 3.88-6.58), first- and second-trimester drinking by 12.4 times, and drinking all trimesters by 16 times (p < 0.001, OR = 15.69, 95% CI: 11.92-20.64). Paternal drinking during and prior to pregnancy, without adjustment, increased the likelihood of FASD significantly (OR = 1.06 and 1.11, respectively), but the significance of both relationships disappeared when maternal alcohol and tobacco use were controlled. CONCLUSIONS Differences in FASD risk emerged from the examination of multiple proximal variables of maternal alcohol and tobacco use, reflecting increased FASD risk at greater levels of maternal alcohol consumption.
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
- Department of Psychiatry, 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
| | - Julie M. Hasken
- Nutrition Research Institute, The University of North Carolina at Chapel Hill, 500 Laureate Way, Kannapolis, NC 28081, United States
| | - Marlene M. de Vries
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
| | - Anna-Susan Marais
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
| | - Omar Abdul-Rahman
- Department of Pediatrics, New York- Presbyterian Weill Cornell Medicine, Columbia University, 505 E 70 St, New York, NY 10021
| | - Luther K. Robinson
- Department of Pediatrics, State University of New York, 1001 Main Street, Buffalo, NY 14203, United States
| | - Margaret P. Adam
- Department of Pediatrics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98175, USA
| | - Melanie A. Manning
- Department of Pathology and Pediatrics, Stanford University School of Medicine, 291 Campus Drive, Stanford, CA 94305, 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
| | - David Buckley
- Center on Alcoholism, Substance Abuse and Addictions, The University of New Mexico, 2650 Yale SE, Albuquerque, NM 87106, United States
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
| | - Charles D.H. Parry
- Department of Psychiatry, 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
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Francie van Zijl Drive, Tygerberg, Cape Town 7505, South Africa
- Sanford Children’s Genomic Medicine Consortium, Sanford Health, 1600 W. 22 St. Sioux Falls, SD, 57117, United States
| |
Collapse
|
7
|
Collier AD, Abdulai AR, Leibowitz SF. Utility of the Zebrafish Model for Studying Neuronal and Behavioral Disturbances Induced by Embryonic Exposure to Alcohol, Nicotine, and Cannabis. Cells 2023; 12:2505. [PMID: 37887349 PMCID: PMC10605371 DOI: 10.3390/cells12202505] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
It is estimated that 5% of pregnant women consume drugs of abuse during pregnancy. Clinical research suggests that intake of drugs during pregnancy, such as alcohol, nicotine and cannabis, disturbs the development of neuronal systems in the offspring, in association with behavioral disturbances early in life and an increased risk of developing drug use disorders. After briefly summarizing evidence in rodents, this review focuses on the zebrafish model and its inherent advantages for studying the effects of embryonic exposure to drugs of abuse on behavioral and neuronal development, with an emphasis on neuropeptides known to promote drug-related behaviors. In addition to stimulating the expression and density of peptide neurons, as in rodents, zebrafish studies demonstrate that embryonic drug exposure has marked effects on the migration, morphology, projections, anatomical location, and peptide co-expression of these neurons. We also describe studies using advanced methodologies that can be applied in vivo in zebrafish: first, to demonstrate a causal relationship between the drug-induced neuronal and behavioral disturbances and second, to discover underlying molecular mechanisms that mediate these effects. The zebrafish model has great potential for providing important information regarding the development of novel and efficacious therapies for ameliorating the effects of early drug exposure.
Collapse
Affiliation(s)
| | | | - Sarah F. Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
8
|
Kukowka A, Brzuchalski B, Kurzawski M, Malinowski D, Białecka MA. ADH1B, ADH1B/C and CYP2E1 Gene Polymorphism and the Risk of Fetal Alcohol Spectrum Disorder. Genes (Basel) 2023; 14:1392. [PMID: 37510297 PMCID: PMC10379323 DOI: 10.3390/genes14071392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Increasing alcohol consumption by women of childbearing age contributes to more frequent cases of fetal alcohol spectrum disorder. The cause of the syndrome is fetal alcohol exposure, particularly what is referred to as high prenatal alcohol exposure. Low metabolic activity of fetal enzymes shifts the burden of ethanol removal to maternal metabolism. One of the factors influencing the pathogenesis of FASD is the genetic background. It can determine the rate of elimination of ethanol, thus increasing or decreasing the time of fetal exposure to ethanol and also decreasing its concentration. Genetic polymorphisms could potentially play a significant role in these processes. In the present study, we considered three polymorphisms of genes implicated in the synthesis of enzymes involved in ethanol metabolism, i.e., ADH1b (rs1229984), ADH1b/c (rs1789891), and CYP2E1 (rs3813867). The studied group consisted of 303 children and 251 mothers. Both mothers' and children's genotypes were considered in our analysis. There were no statistically significant differences between the respective groups of genotypes of the studied polymorphisms. However, the genetic background of FASD is still elusive.
Collapse
Affiliation(s)
- Arnold Kukowka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Bogusław Brzuchalski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Mateusz Kurzawski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland;
| | - Damian Malinowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| | - Monika Anna Białecka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstanców Wielkopolskich 72 St., 70-111 Szczecin, Poland (D.M.)
| |
Collapse
|
9
|
Chaudoin TR, Bonasera SJ, Dunaevsky A, Padmashri R. Exploring behavioral phenotypes in a mouse model of fetal alcohol spectrum disorders. Dev Neurobiol 2023; 83:184-204. [PMID: 37433012 PMCID: PMC10546278 DOI: 10.1002/dneu.22922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/18/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Fetal alcohol spectrum disorders are one of the leading causes of developmental abnormalities worldwide. Maternal consumption of alcohol during pregnancy leads to a diverse range of cognitive and neurobehavioral deficits. Although moderate-to-heavy levels of prenatal alcohol exposure (PAE) have been associated with adverse offspring outcomes, there is limited data on the consequences of chronic low-level PAE. Here, we use a model of maternal voluntary alcohol consumption throughout gestation in a mouse model to investigate the effects of PAE on behavioral phenotypes during late adolescence and early adulthood in male and female offspring. Body composition was measured by dual-energy X-ray absorptiometry. Baseline behaviors, including feeding, drinking, and movement, were examined by performing home cage monitoring studies. The impact of PAE on motor function, motor skill learning, hyperactivity, acoustic reactivity, and sensorimotor gating was investigated by performing a battery of behavioral tests. PAE was found to be associated with altered body composition. No differences in overall movement, food, or water consumption were observed between control and PAE mice. Although PAE offspring of both sexes exhibited deficits in motor skill learning, no differences were observed in basic motor skills such as grip strength and motor coordination. PAE females exhibited a hyperactive phenotype in a novel environment. PAE mice exhibited increased reactivity to acoustic stimuli, and PAE females showed disrupted short-term habituation. Sensorimotor gating was not altered in PAE mice. Collectively, our data show that chronic low-level exposure to alcohol in utero results in behavioral impairments.
Collapse
Affiliation(s)
- Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
10
|
Schriml LM, Lichenstein R, Bisordi K, Bearer C, Baron JA, Greene C. Modeling the enigma of complex disease etiology. J Transl Med 2023; 21:148. [PMID: 36829165 PMCID: PMC9957692 DOI: 10.1186/s12967-023-03987-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Complex diseases often present as a diagnosis riddle, further complicated by the combination of multiple phenotypes and diseases as features of other diseases. With the aim of enhancing the determination of key etiological factors, we developed and tested a complex disease model that encompasses diverse factors that in combination result in complex diseases. This model was developed to address the challenges of classifying complex diseases given the evolving nature of understanding of disease and interaction and contributions of genetic, environmental, and social factors. METHODS Here we present a new approach for modeling complex diseases that integrates the multiple contributing genetic, epigenetic, environmental, host and social pathogenic effects causing disease. The model was developed to provide a guide for capturing diverse mechanisms of complex diseases. Assessment of disease drivers for asthma, diabetes and fetal alcohol syndrome tested the model. RESULTS We provide a detailed rationale for a model representing the classification of complex disease using three test conditions of asthma, diabetes and fetal alcohol syndrome. Model assessment resulted in the reassessment of the three complex disease classifications and identified driving factors, thus improving the model. The model is robust and flexible to capture new information as the understanding of complex disease improves. CONCLUSIONS The Human Disease Ontology's Complex Disease model offers a mechanism for defining more accurate disease classification as a tool for more precise clinical diagnosis. This broader representation of complex disease, therefore, has implications for clinicians and researchers who are tasked with creating evidence-based and consensus-based recommendations and for public health tracking of complex disease. The new model facilitates the comparison of etiological factors between complex, common and rare diseases and is available at the Human Disease Ontology website.
Collapse
Affiliation(s)
- Lynn M. Schriml
- grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD USA
| | - Richard Lichenstein
- grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Baltimore, MD USA
| | - Katharine Bisordi
- grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Baltimore, MD USA
| | - Cynthia Bearer
- grid.67105.350000 0001 2164 3847Case Western Reserve University, Cleveland, OH USA
| | - J. Allen Baron
- grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD USA
| | - Carol Greene
- grid.411024.20000 0001 2175 4264University of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
11
|
Popova S, Charness ME, Burd L, Crawford A, Hoyme HE, Mukherjee RAS, Riley EP, Elliott EJ. Fetal alcohol spectrum disorders. Nat Rev Dis Primers 2023; 9:11. [PMID: 36823161 DOI: 10.1038/s41572-023-00420-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2023] [Indexed: 02/25/2023]
Abstract
Alcohol readily crosses the placenta and may disrupt fetal development. Harm from prenatal alcohol exposure (PAE) is determined by the dose, pattern, timing and duration of exposure, fetal and maternal genetics, maternal nutrition, concurrent substance use, and epigenetic responses. A safe dose of alcohol use during pregnancy has not been established. PAE can cause fetal alcohol spectrum disorders (FASD), which are characterized by neurodevelopmental impairment with or without facial dysmorphology, congenital anomalies and poor growth. FASD are a leading preventable cause of birth defects and developmental disability. The prevalence of FASD in 76 countries is >1% and is high in individuals living in out-of-home care or engaged in justice and mental health systems. The social and economic effects of FASD are profound, but the diagnosis is often missed or delayed and receives little public recognition. Future research should be informed by people living with FASD and be guided by cultural context, seek consensus on diagnostic criteria and evidence-based treatments, and describe the pathophysiology and lifelong effects of FASD. Imperatives include reducing stigma, equitable access to services, improved quality of life for people with FASD and FASD prevention in future generations.
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada.
| | - Michael E Charness
- VA Boston Healthcare System, West Roxbury, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.,Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Larry Burd
- North Dakota Fetal Alcohol Syndrome Center, Department of Pediatrics, University of North Dakota School of Medicine and Health Sciences, Pediatric Therapy Services, Altru Health System, Grand Forks, ND, USA
| | - Andi Crawford
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - H Eugene Hoyme
- Sanford Children's Genomic Medicine Consortium, Sanford Health, and University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Raja A S Mukherjee
- National UK FASD Clinic, Surrey and Borders Partnership NHS Foundation Trust, Redhill, Surrey, UK
| | - Edward P Riley
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, USA
| | - Elizabeth J Elliott
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,New South Wales FASD Assessment Service, CICADA Centre for Care and Intervention for Children and Adolescents affected by Drugs and Alcohol, Sydney Children's Hospitals Network, Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Genetic Influences on Fetal Alcohol Spectrum Disorder. Genes (Basel) 2023; 14:genes14010195. [PMID: 36672936 PMCID: PMC9859092 DOI: 10.3390/genes14010195] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
13
|
Rodríguez-Sastre N, Shapiro N, Hawkins DY, Lion AT, Peyreau M, Correa AE, Dionne K, Bradham CA. Ethanol exposure perturbs sea urchin development and disrupts developmental timing. Dev Biol 2023; 493:89-102. [PMID: 36368523 DOI: 10.1016/j.ydbio.2022.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Ethanol is a known vertebrate teratogen that causes craniofacial defects as a component of fetal alcohol syndrome (FAS). Our results show that sea urchin embryos treated with ethanol similarly show broad skeletal patterning defects, potentially analogous to the defects associated with FAS. The sea urchin larval skeleton is a simple patterning system that involves only two cell types: the primary mesenchymal cells (PMCs) that secrete the calcium carbonate skeleton and the ectodermal cells that provide migratory, positional, and differentiation cues for the PMCs. Perturbations in RA biosynthesis and Hh signaling pathways are thought to be causal for the FAS phenotype in vertebrates. Surprisingly, our results indicate that these pathways are not functionally relevant for the teratogenic effects of ethanol in developing sea urchins. We found that developmental morphology as well as the expression of some ectodermal and PMC genes was delayed by ethanol exposure. Temporal transcriptome analysis revealed significant impacts of ethanol on signaling and metabolic gene expression, and a disruption in the timing of GRN gene expression that includes both delayed and precocious gene expression throughout the specification network. We conclude that the skeletal patterning perturbations in ethanol-treated embryos likely arise from a loss of temporal synchrony within and between the instructive and responsive tissues.
Collapse
Affiliation(s)
| | | | | | - Alexandra T Lion
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA
| | | | - Andrea E Correa
- Universidad de Puerto Rico-Recinto Aguadilla, Puerto Rico, USA
| | | | - Cynthia A Bradham
- Biology Department, Boston University, Boston, MA, USA; MCBB Program, Boston University, Boston, MA, USA; Biological Design Center, Boston University, Boston, MA, USA.
| |
Collapse
|
14
|
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: 2.0] [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
|
15
|
Manikandan P, Sarmah S, Marrs JA. Ethanol Effects on Early Developmental Stages Studied Using the Zebrafish. Biomedicines 2022; 10:2555. [PMID: 36289818 PMCID: PMC9599251 DOI: 10.3390/biomedicines10102555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Fetal alcohol spectrum disorder (FASD) results from prenatal ethanol exposure. The zebrafish (Danio rerio) is an outstanding in vivo FASD model. Early development produced the three germ layers and embryonic axes patterning. A critical pluripotency transcriptional gene circuit of sox2, pou5f1 (oct4; recently renamed pou5f3), and nanog maintain potency and self-renewal. Ethanol affects sox2 expression, which functions with pou5f1 to control target gene transcription. Various genes, like elf3, may interact and regulate sox2, and elf3 knockdown affects early development. Downstream of the pluripotency transcriptional circuit, developmental signaling activities regulate morphogenetic cell movements and lineage specification. These activities are also affected by ethanol exposure. Hedgehog signaling is a critical developmental signaling pathway that controls numerous developmental events, including neural axis specification. Sonic hedgehog activities are affected by embryonic ethanol exposure. Activation of sonic hedgehog expression is controlled by TGF-ß family members, Nodal and Bmp, during dorsoventral (DV) embryonic axis establishment. Ethanol may perturb TGF-ß family receptors and signaling activities, including the sonic hedgehog pathway. Significantly, experiments show that activation of sonic hedgehog signaling rescues some embryonic ethanol exposure effects. More research is needed to understand how ethanol affects early developmental signaling and morphogenesis.
Collapse
Affiliation(s)
| | | | - James A. Marrs
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
16
|
Todd D, Clapp M, Dains P, Karacay B, Bonthius DJ. Purkinje cell-specific deletion of CREB worsens alcohol-induced cerebellar neuronal losses and motor deficits. Alcohol 2022; 101:27-35. [PMID: 35378204 PMCID: PMC9783827 DOI: 10.1016/j.alcohol.2022.02.005] [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: 08/19/2021] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Exposure to alcohol during pregnancy can kill developing fetal neurons and lead to fetal alcohol spectrum disorder (FASD) in the offspring. However, not all fetuses are equally vulnerable to alcohol toxicity. These differences in vulnerability among individuals are likely due, at least in part, to genetic differences. Some genes encode neuroprotective molecules that act through signaling pathways to protect neurons against alcohol's toxic effects. One signaling pathway that can protect cultured neurons against alcohol-induced cell death in vitro is the cAMP pathway. A goal of this study was to determine whether the cAMP pathway can exert a similar neuroprotective effect against alcohol in vivo. A key molecule within the cAMP pathway is cAMP response element binding protein (CREB). In this study, CREB was specifically disrupted in cerebellar Purkinje cells to study its role in protection of cerebellar neurons against alcohol toxicity. METHODS Mice with Purkinje cell-specific knockout of CREB were generated with the Cre-lox system. A 2 × 2 design was used in which Cre-negative and Cre-positive mice received either 0.0 or 2.2 mg/g ethanol by intraperitoneal (i.p.) injection daily over postnatal day (PD) 4-9. Stereological cell counts of cerebellar Purkinje cells and granule cells were performed on PD 10. Motor function was assessed on PD 40 using the rotarod. RESULTS Purkinje cell-specific disruption of CREB alone (in the absence of alcohol) induced only a small reduction in Purkinje cell number. However, the loss of CREB function from Purkinje cells greatly increased the vulnerability of Purkinje cells to alcohol-induced cell death. While alcohol killed 20% of Purkinje cells in the Cre-negative (CREB-expressing) mice, alcohol killed 57% of Purkinje cells in the Cre-positive (CREB-nonexpressing) mice. This large loss of Purkinje cells did not lead to similar alcohol-induced losses of granule cells. In the absence of alcohol, lack of CREB function in Purkinje cells had no effect on rotarod performance. However, in the presence of alcohol, disruption of CREB in Purkinje cells substantially worsened rotarod performance. DISCUSSION Disruption of a single gene (CREB) in a single neuronal population (Purkinje cells) greatly increases the vulnerability of that cell population to alcohol-induced cell death and worsens alcohol-induced brain dysfunction. The results suggest that the cAMP pathway can protect cells in vivo against alcohol toxicity and underline the importance of genetics in determining the neuropathology and behavioral deficits of FASD.
Collapse
Affiliation(s)
- Dylan Todd
- Neuroscience Program, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Michael Clapp
- Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Parker Dains
- Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Bahri Karacay
- Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA
| | - Daniel J. Bonthius
- Neuroscience Program, University of Iowa Carver College of Medicine, Iowa City, IA,Department of Pediatrics, University of Iowa, Carver College of Medicine, Iowa City, IA,Atrium Health/Levine Children’s Hospital, Charlotte, NC
| |
Collapse
|
17
|
Yoon B, Yeung P, Santistevan N, Bluhm LE, Kawasaki K, Kueper J, Dubielzig R, VanOudenhove J, Cotney J, Liao EC, Grinblat Y. Zebrafish models of alx-linked frontonasal dysplasia reveal a role for Alx1 and Alx3 in the anterior segment and vasculature of the developing eye. Biol Open 2022; 11:bio059189. [PMID: 35142342 PMCID: PMC9167625 DOI: 10.1242/bio.059189] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/28/2022] [Indexed: 11/18/2022] Open
Abstract
The cellular and genetic mechanisms that coordinate formation of facial sensory structures with surrounding skeletal and soft tissue elements remain poorly understood. Alx1, a homeobox transcription factor, is a key regulator of midfacial morphogenesis. ALX1 mutations in humans are linked to severe congenital anomalies of the facial skeleton (frontonasal dysplasia, FND) with malformation or absence of eyes and orbital contents (micro- and anophthalmia). Zebrafish with loss-of-function alx1 mutations develop with craniofacial and ocular defects of variable penetrance, likely due to compensatory upregulation in expression of a paralogous gene, alx3. Here we show that zebrafish alx1;alx3 mutants develop with highly penetrant cranial and ocular defects that resemble human ALX1-linked FND. alx1 and alx3 are expressed in anterior cranial neural crest (aCNC), which gives rise to the anterior neurocranium (ANC), anterior segment structures of the eye and vascular pericytes. Consistent with a functional requirement for alx genes in aCNC, alx1; alx3 mutants develop with nearly absent ANC and grossly aberrant hyaloid vasculature and ocular anterior segment, but normal retina. In vivo lineage labeling identified a requirement for alx1 and alx3 during aCNC migration, and transcriptomic analysis suggested oxidative stress response as a key target mechanism of this function. Oxidative stress is a hallmark of fetal alcohol toxicity, and we found increased penetrance of facial and ocular malformations in alx1 mutants exposed to ethanol, consistent with a protective role for alx1 against ethanol toxicity. Collectively, these data demonstrate a conserved role for zebrafish alx genes in controlling ocular and facial development, and a novel role in protecting these key midfacial structures from ethanol toxicity during embryogenesis. These data also reveal novel roles for alx genes in ocular anterior segment formation and vascular development and suggest that retinal deficits in alx mutants may be secondary to aberrant ocular vascularization and anterior segment defects. This study establishes robust zebrafish models for interrogating conserved genetic mechanisms that coordinate facial and ocular development, and for exploring gene--environment interactions relevant to fetal alcohol syndrome.
Collapse
Affiliation(s)
- Baul Yoon
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Pan Yeung
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Nicholas Santistevan
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| | - Lauren E. Bluhm
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Kenta Kawasaki
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Janina Kueper
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
- Institute of Human Genetics, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Richard Dubielzig
- Comparative Ocular Pathology Laboratory of Wisconsin (COPLOW), University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer VanOudenhove
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Justin Cotney
- University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, CT 06030, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, Boston, 02114, USA
| | - Yevgenya Grinblat
- Departments of Integrative Biology and Neuroscience, University of Wisconsin, Madison, WI 53706, USA
- Genetics Ph.D. Training Program, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
18
|
Abstract
This article is part of a Festschrift commemorating the 50th anniversary of the National Institute on Alcohol Abuse and Alcoholism (NIAAA). Established in 1970, first as part of the National Institute of Mental Health and later as an independent institute of the National Institutes of Health, NIAAA today is the world’s largest funding agency for alcohol research. In addition to its own intramural research program, NIAAA supports the entire spectrum of innovative basic, translational, and clinical research to advance the diagnosis, prevention, and treatment of alcohol use disorder and alcohol-related problems. To celebrate the anniversary, NIAAA hosted a 2-day symposium, “Alcohol Across the Lifespan: 50 Years of Evidence-Based Diagnosis, Prevention, and Treatment Research,” devoted to key topics within the field of alcohol research. This article is based on Dr. Charness’ presentation at the event. NIAAA Director George F. Koob, Ph.D., serves as editor of the Festschrift.
Collapse
Affiliation(s)
- Michael E Charness
- VA Boston Healthcare System, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts. Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
19
|
Nishimura Y, Kurosawa K. Analysis of Gene-Environment Interactions Related to Developmental Disorders. Front Pharmacol 2022; 13:863664. [PMID: 35370658 PMCID: PMC8969575 DOI: 10.3389/fphar.2022.863664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/03/2022] [Indexed: 11/21/2022] Open
Abstract
Various genetic and environmental factors are associated with developmental disorders (DDs). It has been suggested that interaction between genetic and environmental factors (G × E) is involved in the etiology of DDs. There are two major approaches to analyze the interaction: genome-wide and candidate gene-based approaches. In this mini-review, we demonstrate how these approaches can be applied to reveal the G × E related to DDs focusing on zebrafish and mouse models. We also discuss novel approaches to analyze the G × E associated with DDs.
Collapse
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kenji Kurosawa
- Division of Medical Genetics, Kanagawa Children’s Medical Center, Yokohama, Japan
- Department of Clinical Dysmorphology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
20
|
Joseph JJ, Mela M, Pei J. Aggressive behaviour and violence in children and adolescents with FASD: A synthesizing review. Clin Psychol Rev 2022; 94:102155. [DOI: 10.1016/j.cpr.2022.102155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/09/2022] [Accepted: 03/23/2022] [Indexed: 11/03/2022]
|
21
|
Harvey DC, De Zoysa P, Toubat O, Choi J, Kishore J, Tsukamoto H, Kumar SR. Concomitant genetic defects potentiate the adverse impact of prenatal alcohol exposure on cardiac outflow tract maturation. Birth Defects Res 2022; 114:105-115. [PMID: 34859965 PMCID: PMC10033225 DOI: 10.1002/bdr2.1968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is associated with an increased incidence of congenital heart defects (CHD), in particular outflow tract (OFT) defects. However, the variability in the incidence of CHD following PAE has not been fully explored. We hypothesize that a concomitant, relevant genetic defect would potentiate the adverse effect of PAE and partially explain the variability of PAE-induced CHD incidence. METHODS The OFT is formed by the second heart field (SHF). Our PAE model consisted of two intraperitoneal injections (3 g/kg, separated by 6 hr) of 30% ethanol on E6.5 during SHF specification. The impact of genetic defects was studied by SHF-specific loss of Delta-like ligand 4 (Dll4), fibroblast growth factor 8 (Fgf8) and Islet1. RESULTS Acute PAE alone significantly increased CHD incidence (4% vs. 26%, p = .015) with a particular increase in OFT alignment defects, viz., double outlet right ventricle (0 vs. 9%, p = .02). In embryos with a SHF genetic defect, acute PAE significantly increased CHD incidence (14 vs. 63%, p < .001), including double outlet right ventricle (6 vs. 50%, p < .001) compared to controls. PAE (p = .01) and heterozygous loss of Dll4 (p = .04) were found to independently contribute to CHD incidence, while neither Islet1 nor Fgf8 defects were found to be significant. CONCLUSIONS Our model recapitulates the increased incidence of OFT alignment defects seen in the clinic due to PAE. The presence of a concomitant SHF genetic mutation increases the incidence of PAE-related OFT defects. An apparent synergistic interaction between PAE and the loss of DLL4-mediated Notch signaling in OFT alignment requires further analysis.
Collapse
Affiliation(s)
- Drayton C Harvey
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Prashan De Zoysa
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Omar Toubat
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Jongkyu Choi
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Jahnavi Kishore
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Hidekazu Tsukamoto
- Department of Pathology, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Southern California Research Center for ALPD and Cirrhosis, Los Angeles, California, USA
- Greater Los Angeles VA Healthcare System, Los Angeles, California, USA
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| |
Collapse
|
22
|
Bhatia S, Arslan E, Rodriguez-Hernandez L, Bonin R, Wells PG. DNA damage and repair and epigenetic modification in the role of oxoguanine glycosylase 1 (OGG1) in brain development. Toxicol Sci 2022; 187:93-111. [PMID: 35038743 DOI: 10.1093/toxsci/kfac003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Oxoguanine glycosylase 1 (OGG1) repairs the predominant reactive oxygen species (ROS)-initiated DNA lesion 8-oxoguanine (8-oxoG). Human OGG1 polymorphisms resulting in reduced DNA repair associate with an increased risk for disorders like cancer and diabetes, but the role of OGG1 in brain development is unclear. Herein, we show that Ogg1 knockout mice at 2-3 months of age exhibit enhanced gene- and sex-dependent DNA damage (strand breaks) and decreased epigenetic DNA methylation marks (5-methylcytosine, 5-hydroxymethylcytosine), both of which were associated with increased cerebellar calbindin levels, reduced hippocampal postsynaptic function, altered body weight with age and disorders of brain function reflected in behavioural tests for goal-directed repetitive behaviour, anxiety and fear, object recognition and spatial memory, motor coordination and startle response. These results suggest that OGG1 plays an important role in normal brain development, possibly via both its DNA repair activity and its role as an epigenetic modifier, with OGG1 deficiencies potentially contributing to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shama Bhatia
- Dept. of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Eliyas Arslan
- Dept. of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Luis Rodriguez-Hernandez
- Dept. of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Robert Bonin
- Dept. of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Peter G Wells
- Dept. of Pharmaceutical Sciences, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Centre for Pharmaceutical Oncology, Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Dept. of Pharmacology & Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Burton DF, Boa-Amponsem OM, Dixon MS, Hopkins MJ, Herbin TA, Toney S, Tarpley M, Rodriguez BV, Fish EW, Parnell SE, Cole GJ, Williams KP. Pharmacological activation of the Sonic hedgehog pathway with a Smoothened small molecule agonist ameliorates the severity of alcohol-induced morphological and behavioral birth defects in a zebrafish model of fetal alcohol spectrum disorder. J Neurosci Res 2022; 100:1585-1601. [PMID: 35014067 PMCID: PMC9271529 DOI: 10.1002/jnr.25008] [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: 07/19/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 12/17/2022]
Abstract
Ethanol exposure during the early stages of embryonic development can lead to a range of morphological and behavioral differences termed fetal alcohol spectrum disorders (FASDs). In a zebrafish model, we have shown that acute ethanol exposure at 8-10 hr postfertilization (hpf), a critical time of development, produces birth defects similar to those clinically characterized in FASD. Dysregulation of the Sonic hedgehog (Shh) pathway has been implicated as a molecular basis for many of the birth defects caused by prenatal alcohol exposure. We observed in zebrafish embryos that shh expression was significantly decreased by ethanol exposure at 8-10 hpf, while smo expression was much less affected. Treatment of zebrafish embryos with SAG or purmorphamine, small molecule Smoothened agonists that activate Shh signaling, ameliorated the severity of ethanol-induced developmental malformations including altered eye size and midline brain development. Furthermore, this rescue effect of Smo activation was dose dependent and occurred primarily when treatment was given after ethanol exposure. Markers of Shh signaling (gli1/2) and eye development (pax6a) were restored in embryos treated with SAG post-ethanol exposure. Since embryonic ethanol exposure has been shown to produce later-life neurobehavioral impairments, juvenile zebrafish were examined in the novel tank diving test. Our results further demonstrated that in zebrafish embryos exposed to ethanol, SAG treatment was able to mitigate long-term neurodevelopmental impairments related to anxiety and risk-taking behavior. Our results indicate that pharmacological activation of the Shh pathway at specific developmental timing markedly diminishes the severity of alcohol-induced birth defects.
Collapse
Affiliation(s)
- Derek F Burton
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Oswald M Boa-Amponsem
- Integrated Biosciences PhD Program, North Carolina Central University, Durham, North Carolina, USA.,Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, USA
| | - Maria S Dixon
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Michael J Hopkins
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Te-Andre Herbin
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Shiquita Toney
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Michael Tarpley
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA
| | - Blanca V Rodriguez
- Department of Biochemistry, Duke University, Durham, North Carolina, USA
| | - Eric W Fish
- Bowles Center for Alcohol Studies, 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.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gregory J Cole
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina, USA.,Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, North Carolina, USA
| | - Kevin P Williams
- Biomanufacturing Research Institute and Technology Enterprise, North Carolina Central University, Durham, North Carolina, USA.,Department of Pharmaceutical Sciences, North Carolina Central University, Durham, North Carolina, USA
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe recent findings on the clinical presentation, pathogenesis, and management of fetal alcohol spectrum disorders (FASDs). Alcohol causes a range of physical, developmental, and cognitive impairments on the developing fetus. Individuals exposed to alcohol prenatally have a wide variability in dysmorphic and neurologic features. Hence, a greater understanding of the mechanisms through which alcohol induces defects in the developing fetus is imperative in developing therapies that prevent alcohol-induced effects. RECENT FINDINGS Current research has focused on leveraging technology to developing tools that can aid in the diagnostic process, defining patterns of neurocognition and neuroimaging specific to FASD, developing neurobehavioral and pharmacologic interventions, and expanding access to care. SUMMARY FASDs are a common cause of neurodevelopmental impairment in school-age children, and their recognition is essential to provide early interventions in order to optimize the outcome for these individuals when they reach adulthood. Although previously thought to be the result of irreversible neurologic injury from prenatal alcohol exposure, recent evidence points to the benefits of applying principles regarding neuroplasticity in improving the lives for patients and their families.
Collapse
Affiliation(s)
- Diego A Gomez
- Mayo Clinic Alix School of Medicine, Phoenix, Arizona
| | - Omar A Abdul-Rahman
- Department of Genetic Medicine, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
25
|
Thomas KN, Zimmel KN, Roach AN, Basel A, Mehta NA, Bedi YS, Golding MC. Maternal background alters the penetrance of growth phenotypes and sex-specific placental adaptation of offspring sired by alcohol-exposed males. FASEB J 2021; 35:e22035. [PMID: 34748230 PMCID: PMC8713293 DOI: 10.1096/fj.202101131r] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/05/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022]
Abstract
Epigenetic mechanisms of paternal inheritance are an emerging area of interest in our efforts to understand fetal alcohol spectrum disorders. In rodent models examining maternal alcohol exposures, different maternal genetic backgrounds protect or sensitize offspring to alcohol‐induced teratogenesis. However, whether maternal background can mitigate sperm‐inherited alterations in developmental programming and modify the penetrance of growth defects induced by preconception paternal alcohol exposures remains unaddressed. In our previous studies examining pure C57Bl/6J crosses, the offspring of alcohol‐exposed sires exhibited fetal growth restriction, enlarged placentas, and decreased placental efficiency. Here, we find that in contrast to our previous studies, the F1 offspring of alcohol‐exposed C57Bl/6J sires and CD‐1 dams do not exhibit fetal growth restriction, with male fetuses developing smaller placentas and increased placental efficiencies. However, in these hybrid offspring, preconception paternal alcohol exposure induces sex‐specific changes in placental morphology. Specifically, the female offspring of alcohol‐exposed sires displayed structural changes in the junctional and labyrinth zones, along with increased placental glycogen content. These changes in placental organization are accompanied by female‐specific alterations in the expression of imprinted genes Cdkn1c and H19. Although male placentae do not display overt changes in placental histology, using RNA‐sequencing, we identified programmed alterations in genes regulating oxidative phosphorylation, mitochondrial function, and Sirtuin signaling. Collectively, our data reveal that preconception paternal alcohol exposure transmits a stressor to developing offspring, that males and females exhibit distinct patterns of placental adaptation, and that maternal genetic background can modulate the effects of paternal alcohol exposure.
Collapse
Affiliation(s)
- Kara N Thomas
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Katherine N Zimmel
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alexis N Roach
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alison Basel
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Nicole A Mehta
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Yudhishtar S Bedi
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Michael C Golding
- Department of Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
26
|
Fernandes Y, Lovely CB. Zebrafish models of fetal alcohol spectrum disorders. Genesis 2021; 59:e23460. [PMID: 34739740 DOI: 10.1002/dvg.23460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022]
Abstract
Fetal alcohol spectrum disorder (FASD) describes a wide range of structural deficits and cognitive impairments. FASD impacts up to 5% of children born in the United States each year, making ethanol one of the most common teratogens. Due to limitations and ethical concerns, studies in humans are limited in their ability to study FASD. Animal models have proven critical in identifying and characterizing the mechanisms underlying FASD. In this review, we will focus on the attributes of zebrafish that make it a strong model in which to study ethanol-induced developmental defects. Zebrafish have several attributes that make it an ideal model in which to study FASD. Zebrafish produced large numbers of externally fertilized, translucent embryos. With a high degree of genetic amenability, zebrafish are at the forefront of identifying and characterizing the gene-ethanol interactions that underlie FASD. Work from multiple labs has shown that embryonic ethanol exposures result in defects in craniofacial, cardiac, ocular, and neural development. In addition to structural defects, ethanol-induced cognitive and behavioral impairments have been studied in zebrafish. Building upon these studies, work has identified ethanol-sensitive loci that underlie the developmental defects. However, analyses show there is still much to be learned of these gene-ethanol interactions. The zebrafish is ideally suited to expand our understanding of gene-ethanol interactions and their impact on FASD. Because of the conservation of gene function between zebrafish and humans, these studies will directly translate to studies of candidate genes in human populations and allow for better diagnosis and treatment of FASD.
Collapse
Affiliation(s)
- Yohaan Fernandes
- Department of Biology, University of South Dakota, Vermillion, South Dakota, USA
| | - C Ben Lovely
- Department of Biochemistry and Molecular Genetics, Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
27
|
Popova S, Dozet D, Shield K, Rehm J, Burd L. Alcohol's Impact on the Fetus. Nutrients 2021; 13:3452. [PMID: 34684453 PMCID: PMC8541151 DOI: 10.3390/nu13103452] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Alcohol is a teratogen and prenatal exposure may adversely impact the developing fetus, increasing risk for negative outcomes, including Fetal Alcohol Spectrum Disorder (FASD). Global trends of increasing alcohol use among women of childbearing age due to economic development, changing gender roles, increased availability of alcohol, peer pressure and social acceptability of women's alcohol use may put an increasing number of pregnancies at risk for prenatal alcohol exposure (PAE). This risk has been exacerbated by the ongoing COVID-19 pandemic in some countries. METHOD This literature review presents an overview on the epidemiology of alcohol use among childbearing age and pregnant women and FASD by World Health Organization regions; impact of PAE on fetal health, including FASD; associated comorbidities; and social outcomes. RESULTS/CONCLUSION The impact of alcohol on fetal health and social outcomes later in life is enormous, placing a huge economic burden on countries. Prevention of prenatal alcohol exposure and early identification of affected individuals should be a global public health priority.
Collapse
Affiliation(s)
- Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (D.D.); (K.S.); (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, Faculty of Medicine, Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Danijela Dozet
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (D.D.); (K.S.); (J.R.)
- Institute of Medical Science, Faculty of Medicine, Medical Sciences Building, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada
| | - Kevin Shield
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (D.D.); (K.S.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, ON M5T 3M7, Canada
| | - Jürgen Rehm
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON M5S 2S1, Canada; (D.D.); (K.S.); (J.R.)
- Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, ON M5T 3M7, Canada
- Institute of Medical Science, Faculty of Medicine, Medical Sciences Building, University of Toronto, 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 Street 46, 01187 Dresden, Germany
- Department of Psychiatry, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada
- Department of International Health Projects, Institute for Leadership and Health Management, I.M. Sechenov First Moscow State Medical University, Trubetskaya Street, 8, b. 2, 119992 Moscow, Russia
| | - Larry Burd
- Department of Pediatrics, University of North Dakota School of Medicine and Health Sciences, 1301 N Columbia Rd., Grand Forks, ND 58202, USA;
| |
Collapse
|
28
|
Addila AE, Azale T, Gete YK, Yitayal M. The effects of maternal alcohol consumption during pregnancy on adverse fetal outcomes among pregnant women attending antenatal care at public health facilities in Gondar town, Northwest Ethiopia: a prospective cohort study. Subst Abuse Treat Prev Policy 2021; 16:64. [PMID: 34446055 PMCID: PMC8390259 DOI: 10.1186/s13011-021-00401-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The teratogenic effect of fetal alcohol exposure may lead to actual and potential problems, instantly after birth, at infancy; or even later, and mental impairment in life. This study aimed to investigate the effects of maternal alcohol consumption during pregnancy on adverse fetal outcomes at Gondar town public health facilities, Northwest Ethiopia. METHODS A facility-based prospective cohort study was performed among 1778 pregnant women who were booked for antenatal care in selected public health facilities from 29 October 2019 to 7 May 2020 in Gondar town. We used a two-stage random sampling technique to recruit and include participants in the cohort. Data were collected using the Alcohol Use Disorders Identification Test - Consumption (AUDIT-C) standardized and pre-tested questionnaire. Multivariable analysis was performed to examine the association between reported prenatal alcohol exposure (non-hazardous and hazardous) and interested adverse birth outcomes using log-binomial regression modeling. The burden of outcomes was reported using the adjusted risk ratio and population-attributable risk (PAR). RESULTS A total of 1686 pregnant women were included in the analysis, which revealed that the incidences of low birth weight, preterm, and stillbirth were 12.63% (95% CI: 11.12, 14.31), 6.05% (95% CI: 5.00, 7.29) and 4.27% (95% CI: 3.4, 5.35), respectively. Non-hazardous and hazardous alcohol consumption during pregnancy was significantly associated with low birth weight (ARR = 1.50; 95% CI: 1.31, 1.98) and (ARR = 2.34; 95% CI: 1.66, 3.30), respectively. Hazardous alcohol consumption during pregnancy was also significantly associated with preterm birth (ARR = 2.06; 95% CI: 1.21, 3.52). The adjusted PAR of low birth weight related to non-hazardous and hazardous alcohol drinking during pregnancy was 11.72 and 8.44%, respectively. The adjusted PAR of hazardous alcohol consumption was 6.80% for preterm. CONCLUSIONS Our findings suggest that there is an increasing risk of adverse birth outcomes, particularly preterm delivery and low birth weight, with increasing levels of alcohol intake. This result showed that the prevention of maternal alcohol use during pregnancy has the potential to reduce low birth weight and preterm birth. Hence, screening women for alcohol use during antenatal care visits and providing advice with rigorous follow-up of women who used alcohol may save the fetus from the potential risks of adverse birth outcomes.
Collapse
Affiliation(s)
- Alemu Earsido Addila
- Department of Public Health, College of Medicine and Health Sciences, Wachemo University, Hossana, Ethiopia.
- Department of Epidemiology and Biostatistics, College of Medicine and Health Sciences, Institute of Public Health, University of Gondar, Gondar, Ethiopia.
| | - Telake Azale
- Department of Health Education and Behavioral Sciences, College of Medicine and Health Sciences, Institute of Public Health, University of Gondar, Gondar, Ethiopia
| | - Yigzaw Kebede Gete
- Department of Epidemiology and Biostatistics, College of Medicine and Health Sciences, Institute of Public Health, University of Gondar, Gondar, Ethiopia
| | - Mezgebu Yitayal
- Department of Health Systems and Policy, College of Medicine and Health Sciences, Institute of Public Health, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
29
|
Sidik A, Dixon G, Buckley DM, Kirby HG, Sun S, Eberhart JK. Exposure to ethanol leads to midfacial hypoplasia in a zebrafish model of FASD via indirect interactions with the Shh pathway. BMC Biol 2021; 19:134. [PMID: 34210294 PMCID: PMC8247090 DOI: 10.1186/s12915-021-01062-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 06/03/2021] [Indexed: 11/10/2022] Open
Abstract
Background Gene-environment interactions are likely to underlie most human birth defects. The most common known environmental contributor to birth defects is prenatal alcohol exposure. Fetal alcohol spectrum disorders (FASD) describe the full range of defects that result from prenatal alcohol exposure. Gene-ethanol interactions underlie susceptibility to FASD, but we lack a mechanistic understanding of these interactions. Here, we leverage the genetic tractability of zebrafish to address this problem. Results We first show that vangl2, a member of the Wnt/planar cell polarity (Wnt/PCP) pathway that mediates convergent extension movements, strongly interacts with ethanol during late blastula and early gastrula stages. Embryos mutant or heterozygous for vangl2 are sensitized to ethanol-induced midfacial hypoplasia. We performed single-embryo RNA-seq during early embryonic stages to assess individual variation in the transcriptional response to ethanol and determine the mechanism of the vangl2-ethanol interaction. To identify the pathway(s) that are disrupted by ethanol, we used these global changes in gene expression to identify small molecules that mimic the effects of ethanol via the Library of Integrated Network-based Cellular Signatures (LINCS L1000) dataset. Surprisingly, this dataset predicted that the Sonic Hedgehog (Shh) pathway inhibitor, cyclopamine, would mimic the effects of ethanol, despite ethanol not altering the expression levels of direct targets of Shh signaling. Indeed, we found that ethanol and cyclopamine strongly, but indirectly, interact to disrupt midfacial development. Ethanol also interacts with another Wnt/PCP pathway member, gpc4, and a chemical inhibitor of the Wnt/PCP pathway, blebbistatin, phenocopies the effect of ethanol. By characterizing membrane protrusions, we demonstrate that ethanol synergistically interacts with the loss of vangl2 to disrupt cell polarity required for convergent extension movements. Conclusions Our results show that the midfacial defects in ethanol-exposed vangl2 mutants are likely due to an indirect interaction between ethanol and the Shh pathway. Vangl2 functions as part of a signaling pathway that regulates coordinated cell movements during midfacial development. Ethanol exposure alters the position of a critical source of Shh signaling that separates the developing eye field into bilateral eyes, allowing the expansion of the midface. Collectively, our results shed light on the mechanism by which the most common teratogen can disrupt development. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01062-9.
Collapse
Affiliation(s)
- Alfire Sidik
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, 78712, USA.
| | - Groves Dixon
- Department of Integrative Biology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Desire M Buckley
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Hannah G Kirby
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Shuge Sun
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, 78712, USA
| | - Johann K Eberhart
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas, 78712, USA
| |
Collapse
|
30
|
Friedrich RE, Zustin J, Luebke AM, Rosenbaum T, Gosau M, Hagel C, Kohlrusch FK, Wieland I, Zenker M. Neurofibromatosis Type 1 With Cherubism-like Phenotype, Multiple Osteolytic Bone Lesions of Lower Extremities, and Alagille-syndrome: Case Report With Literature Survey. In Vivo 2021; 35:1711-1736. [PMID: 33910856 DOI: 10.21873/invivo.12431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND/AIM Neurofibromatosis type 1 (NF) is an autosomal dominant hereditary disease. The cardinal clinical findings include characteristic skeletal alterations. Difficulties in diagnosis and therapy can arise if an individual has further illnesses. CASE REPORT This is a case report of a 16-year-old patient affected by NF1. She also suffered from Alagille syndrome and the consequences of fetal alcohol exposure. The patient's facial phenotype showed findings that could be assigned to one or more of the known diseases. The patient was referred for treating a cherubism-like recurrent central giant cell granuloma (CGCG) of the jaw. The patient developed bilateral, multilocular non-ossifying fibromas (NOF) of the long bones of the lower extremity. Treatment of the skeletal lesions consisted of local curettage. While NOF regressed after surgery, the CGCG of the jaw remained largely unchanged. Extensive genetic tests confirmed a previously unknown germline mutation in the JAG1 gene, the germline mutation of the NF1 gene, and the somatic mutation in the NF1 gene in the diffuse plexiform neurofibroma, but not in the CGCG. CONCLUSION Assigning facial findings to a defined syndrome is ambiguous in many cases and especially difficult in patients who have multiple diseases that can affect the facial phenotype. Surgical therapy should be adapted to the individual findings.
Collapse
Affiliation(s)
- Reinhard E Friedrich
- Oral and Craniomaxillofacial Surgery, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany;
| | - Jozef Zustin
- Institute of Osteology and Biomechanics, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany.,Institute of Pathology, Gemeinschaftspraxis Pathologie-Regensburg, Regensburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | | | - Martin Gosau
- Oral and Craniomaxillofacial Surgery, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | - Christian Hagel
- Institute of Neuropathology, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | - Felix K Kohlrusch
- Oral and Craniomaxillofacial Surgery, Eppendorf University Hospital, University of Hamburg, Hamburg, Germany
| | - Ilse Wieland
- Institute of Human Genetics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
31
|
Boschen KE, Ptacek TS, Berginski ME, Simon JM, Parnell SE. Transcriptomic analyses of gastrulation-stage mouse embryos with differential susceptibility to alcohol. Dis Model Mech 2021; 14:dmm049012. [PMID: 34137816 PMCID: PMC8246266 DOI: 10.1242/dmm.049012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 12/28/2022] Open
Abstract
Genetics are a known contributor to differences in alcohol sensitivity in humans with fetal alcohol spectrum disorders (FASDs) and in animal models. Our study profiled gene expression in gastrulation-stage embryos from two commonly used, genetically similar mouse substrains, C57BL/6J (6J) and C57BL/6NHsd (6N), that differ in alcohol sensitivity. First, we established normal gene expression patterns at three finely resolved time points during gastrulation and developed a web-based interactive tool. Baseline transcriptional differences across strains were associated with immune signaling. Second, we examined the gene networks impacted by alcohol in each strain. Alcohol caused a more pronounced transcriptional effect in the 6J versus 6N mice, matching the increased susceptibility of the 6J mice. The 6J strain exhibited dysregulation of pathways related to cell death, proliferation, morphogenic signaling and craniofacial defects, while the 6N strain showed enrichment of hypoxia and cellular metabolism pathways. These datasets provide insight into the changing transcriptional landscape across mouse gastrulation, establish a valuable resource that enables the discovery of candidate genes that may modify alcohol susceptibility that can be validated in humans, and identify novel pathogenic mechanisms of alcohol. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Karen E. Boschen
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Travis S. Ptacek
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew E. Berginski
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeremy M. Simon
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Scott E. Parnell
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
32
|
Kazemi T, Huang S, Avci NG, Akay YM, Akay M. Investigating the effects of chronic perinatal alcohol and combined nicotine and alcohol exposure on dopaminergic and non-dopaminergic neurons in the VTA. Sci Rep 2021; 11:8706. [PMID: 33888815 PMCID: PMC8062589 DOI: 10.1038/s41598-021-88221-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
The ventral tegmental area (VTA) is the origin of dopaminergic neurons and the dopamine (DA) reward pathway. This pathway has been widely studied in addiction and drug reinforcement studies and is believed to be the central processing component of the reward circuit. In this study, we used a well-established rat model to expose mother dams to alcohol, nicotine-alcohol, and saline perinatally. DA and non-DA neurons collected from the VTA of the rat pups were used to study expression profiles of miRNAs and mRNAs. miRNA pathway interactions, putative miRNA-mRNA target pairs, and downstream modulated biological pathways were analyzed. In the DA neurons, 4607 genes were differentially upregulated and 4682 were differentially downregulated following nicotine-alcohol exposure. However, in the non-DA neurons, only 543 genes were differentially upregulated and 506 were differentially downregulated. Cell proliferation, differentiation, and survival pathways were enriched after the treatments. Specifically, in the PI3K/AKT signaling pathway, there were 41 miRNAs and 136 mRNAs differentially expressed in the DA neurons while only 16 miRNAs and 20 mRNAs were differentially expressed in the non-DA neurons after the nicotine-alcohol exposure. These results depicted that chronic nicotine and alcohol exposures during pregnancy differentially affect both miRNA and gene expression profiles more in DA than the non-DA neurons in the VTA. Understanding how the expression signatures representing specific neuronal subpopulations become enriched in the VTA after addictive substance administration helps us to identify how neuronal functions may be altered in the brain.
Collapse
Affiliation(s)
- Tina Kazemi
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Shuyan Huang
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Naze G Avci
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Yasemin M Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA
| | - Metin Akay
- Department of Biomedical Engineering, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
33
|
Okazaki S, Otsuka I, Shinko Y, Horai T, Hirata T, Yamaki N, Sora I, Hishimoto A. Epigenetic Clock Analysis in Children With Fetal Alcohol Spectrum Disorder. Alcohol Clin Exp Res 2021; 45:329-337. [PMID: 33296097 DOI: 10.1111/acer.14532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is characterized by severe clinical impairment, considerable social burden, and high mortality and morbidity, which are due to various malformations, sepsis, and cancer. As >50% of deaths from FASD occur during the first year of life, we hypothesized that there is the acceleration of biological aging in FASD. Several recent studies have established genome-wide DNA methylation (DNAm) profiles as "epigenetic clocks" that can estimate biological aging, and FASD has been associated with differential DNAm patterns. Therefore, we tested this hypothesis using epigenetic clocks. METHODS We investigated 5 DNAm-based measures of epigenetic age (HorvathAge, HannumAge, SkinBloodAge, PhenoAge, and GrimAge) and telomere length (DNAmTL) using 4 independent publicly available DNAm datasets; 2 datasets were derived from buccal epithelium, and the other 2 datasets were derived from peripheral blood. RESULTS Compared with controls, children with FASD exhibited an acceleration of GrimAge in 1 buccal and 2 blood datasets. No significant difference was found in other DNAm ages and DNAmTL. Meta-analyses showed a significant acceleration of GrimAge in the blood samples but not in the buccal samples. CONCLUSIONS This study provides novel evidence regarding accelerated epigenetic aging in children with FASD.
Collapse
Affiliation(s)
- Satoshi Okazaki
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ikuo Otsuka
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yutaka Shinko
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tadasu Horai
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takashi Hirata
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naruhisa Yamaki
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ichiro Sora
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akitoyo Hishimoto
- From, Department of Psychiatry, (SO, IO, YS, THo, THi, NY, IS, AH), Kobe University Graduate School of Medicine, Kobe, Japan.,Department of Psychiatry, (AH), Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
34
|
Brown J, Asp E, Carter MN, Spiller V, Bishop-Deaton D. Suggestibility and confabulation among individuals with Fetal Alcohol Spectrum Disorder: A review for criminal justice, forensic mental health, and legal interviewers. INTERNATIONAL JOURNAL OF LAW AND PSYCHIATRY 2020; 73:101646. [PMID: 33246222 DOI: 10.1016/j.ijlp.2020.101646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/19/2020] [Accepted: 11/03/2020] [Indexed: 06/11/2023]
Abstract
Fetal Alcohol Spectrum Disorders (FASD) are conditions arising from prenatal alcohol exposure which results in a range of neurodevelopmental deficits in areas including cognition, memory, language, executive functioning, emotional regulation, and adaptive functioning. Deficits in various neurodevelopmental areas can range from mild to severe, depending on many factors including the quantity and timing of alcohol exposure during the prenatal development period. During interviews in criminal justice, forensic mental health, and legal contexts (e.g., criminal investigations, cross examination, victim interviews, interviews for lawsuits, forensic evaluations, pre-sentence investigations), deficits associated with FASD may elevate the risk of suggestibility and confabulation. These issues can result in negative jurisprudence-related outcomes, including impulsive Miranda rights waivers, incorrect assumptions of competency, inaccurate or incomplete information gathering, misinterpretation of intent, witness reliability issues, court ordered treatment completion problems, probation and parole violations, false confessions, and false accusations. The aim of the present article is to explain the context in which these issues can arise and provide criminal justice, forensic mental health, and legal professionals with key guidelines that can assist in minimizing suggestibility and confabulation when interviewing persons with FASD. We hope that the suggestions and strategies presented in this article will reduce potential obstructions of justice and enhance the quality of information obtained from individuals impacted by FASD. A brief discussion is also provided to identify additional research and training opportunities needed to clarify "best practices" for professionals tasked with evaluating the challenges facing this unique population.
Collapse
Affiliation(s)
- Jerrod Brown
- American Institute for the Advancement of Forensic Studies, St. Paul, MN, USA.
| | - Erik Asp
- Department of Neurology, University of Iowa, Iowa City, IA, USA; Department of Psychology, Hamline University, St. Paul, MN, USA; Wesley & Lorene Artz Cognitive Neuroscience Research Center, Hamline University, St. Paul, MN, USA
| | - Megan N Carter
- University of Washington, Seattle, WA, USA; Department of Social and Health Services, Special Commitment Center, Steilacoom, WA, USA
| | | | - Deanna Bishop-Deaton
- College of Social and Behavioral Sciences, School of Forensic Psychology, Walden University, USA
| |
Collapse
|
35
|
Everson JL, Batchu R, Eberhart JK. Multifactorial Genetic and Environmental Hedgehog Pathway Disruption Sensitizes Embryos to Alcohol-Induced Craniofacial Defects. Alcohol Clin Exp Res 2020; 44:1988-1996. [PMID: 32767777 PMCID: PMC7692922 DOI: 10.1111/acer.14427] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/28/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) is perhaps the most common environmental cause of human birth defects. These exposures cause a range of structural and neurological defects, including facial dysmorphologies, collectively known as fetal alcohol spectrum disorders (FASD). While PAE causes FASD, phenotypic outcomes vary widely. It is thought that multifactorial genetic and environmental interactions modify the effects of PAE. However, little is known of the nature of these modifiers. Disruption of the Hedgehog (Hh) signaling pathway has been suggested as a modifier of ethanol teratogenicity. In addition to regulating the morphogenesis of craniofacial tissues commonly disrupted in FASD, a core member of the Hh pathway, Smoothened, is susceptible to modulation by structurally diverse chemicals. These include environmentally prevalent teratogens like piperonyl butoxide (PBO), a synergist found in thousands of pesticide formulations. METHODS Here, we characterize multifactorial genetic and environmental interactions using a zebrafish model of craniofacial development. RESULTS We show that loss of a single allele of shha sensitized embryos to both alcohol- and PBO-induced facial defects. Co-exposure of PBO and alcohol synergized to cause more frequent and severe defects. The effects of this co-exposure were even more profound in the genetically susceptible shha heterozygotes. CONCLUSIONS Together, these findings shed light on the multifactorial basis of alcohol-induced craniofacial defects. In addition to further implicating genetic disruption of the Hh pathway in alcohol teratogenicity, our findings suggest that co-exposure to environmental chemicals that perturb Hh signaling may be important variables in FASD and related craniofacial disorders.
Collapse
Affiliation(s)
- Joshua L. Everson
- From the Department of Molecular BiosciencesSchool of Natural SciencesUniversity of Texas at AustinAustinTexasUSA
- Waggoner Center for Alcohol and Addiction ResearchSchool of PharmacyUniversity of Texas at AustinAustinTexasUSA
| | - Rithik Batchu
- From the Department of Molecular BiosciencesSchool of Natural SciencesUniversity of Texas at AustinAustinTexasUSA
| | - Johann K. Eberhart
- From the Department of Molecular BiosciencesSchool of Natural SciencesUniversity of Texas at AustinAustinTexasUSA
- Waggoner Center for Alcohol and Addiction ResearchSchool of PharmacyUniversity of Texas at AustinAustinTexasUSA
| |
Collapse
|
36
|
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: 1.0] [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
|
37
|
Boschen KE, Ptacek TS, Simon JM, Parnell SE. Transcriptome-Wide Regulation of Key Developmental Pathways in the Mouse Neural Tube by Prenatal Alcohol Exposure. Alcohol Clin Exp Res 2020; 44:1540-1550. [PMID: 32557641 DOI: 10.1111/acer.14389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/02/2020] [Accepted: 05/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Early gestational alcohol exposure is associated with severe craniofacial and CNS dysmorphologies and behavioral abnormalities during adolescence and adulthood. Alcohol exposure during the formation of the neural tube (gestational day [GD] 8 to 10 in mice; equivalent to4th week of human pregnancy) disrupts development of ventral midline brain structures such as the pituitary, septum, and ventricles. This study identifies transcriptomic changes in the rostroventral neural tube (RVNT), the region of the neural tube that gives rise to the midline structures sensitive to alcohol exposure during neurulation. METHODS Female C57BL/6J mice were administered 2 doses of alcohol (2.9 g/kg) or vehicle 4 hours apart on GD 9.0. The RVNTs of embryos were collected 6 or 24 hours after the first dose and processed for RNA-seq. RESULTS Six hours following GD 9.0 alcohol exposure (GD 9.25), over 2,300 genes in the RVNT were determined to be differentially regulated by alcohol. Enrichment analysis determined that PAE affected pathways related to cell proliferation, p53 signaling, ribosome biogenesis, and immune activation. In addition, over 100 genes involved in primary cilia formation and function and regulation of morphogenic pathways were altered 6 hours after alcohol exposure. The changes to gene expression were largely transient, as only 91 genes identified as differentially regulated by prenatal alcohol at GD 10 (24 hours postexposure). Functionally, the differentially regulated genes at GD 10 were related to organogenesis and cell migration. CONCLUSIONS These data give a comprehensive view of the changing landscape of the embryonic transcriptome networks in regions of the neural tube that give rise to brain structures impacted by a neurulation-stage alcohol exposure. Identification of gene networks dysregulated by alcohol will help elucidate the pathogenic mechanisms of alcohol's actions.
Collapse
Affiliation(s)
- Karen E Boschen
- From the Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Travis S Ptacek
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeremy M Simon
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Scott E Parnell
- From the Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
38
|
Kahila H, Marjonen H, Auvinen P, Avela K, Riikonen R, Kaminen‐Ahola N. 18q12.3-q21.1 microdeletion detected in the prenatally alcohol-exposed dizygotic twin with discordant fetal alcohol syndrome phenotype. Mol Genet Genomic Med 2020; 8:e1192. [PMID: 32096599 PMCID: PMC7196488 DOI: 10.1002/mgg3.1192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A pair of dizygotic twins discordantly affected by heavy prenatal alcohol exposure (PAE) was reported previously by Riikonen, suggesting the role of genetic risk or protective factors in the etiology of alcohol-induced developmental disorders. Now, we have re-examined these 25-year-old twins and explored genetic origin of the phenotypic discordancy reminiscent with fetal alcohol syndrome (FAS). Furthermore, we explored alterations in DNA methylation profile of imprinting control region at growth-related insulin-like growth factor 2 (IGF2)/H19 locus in twins' white blood cells (WBC), which have been associated earlier with alcohol-induced genotype-specific changes in placental tissue. METHODS Microarray-based comparative genomic hybridization (aCGH) was used to detect potential submicroscopic chromosomal abnormalities, and developmental as well as phenotypic information about twins were collected. Traditional bisulfite sequencing was used for DNA methylation analysis. RESULTS Microarray-based comparative genomic hybridization revealed a microdeletion 18q12.3-q21.1. in affected twin, residing in a known 18q deletion syndrome region. This syndrome has been associated with growth restriction, developmental delay or intellectual deficiency, and abnormal facial features in previous studies, and thus likely explains the phenotypic discordancy between the twins. We did not observe association between WBCs' DNA methylation profile and PAE, but interestingly, a trend of decreased DNA methylation at the imprinting control region was seen in the twin with prenatal growth retardation at birth. CONCLUSIONS The microdeletion emphasizes the importance of adequate chromosomal testing in examining the etiology of complex alcohol-induced developmental disorders. Furthermore, the genotype-specific decreased DNA methylation at the IGF2/H19 locus cannot be considered as a biological mark for PAE in adult WBCs.
Collapse
Affiliation(s)
- Hanna Kahila
- Department of Obstetrics and GynecologyHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Heidi Marjonen
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| | - Pauliina Auvinen
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| | - Kristiina Avela
- Department of Clinical GeneticsHelsinki University HospitalHUSLABHelsinkiFinland
| | - Raili Riikonen
- Children's HospitalKuopio University HospitalUniversity of Eastern FinlandKuopioFinland
| | - Nina Kaminen‐Ahola
- Department of Medical and Clinical GeneticsMedicumUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
39
|
McDonald BW, Watson PE. Maternal alcohol intakes before and during pregnancy: Impact on the mother and infant outcome to 18 months. NORDIC STUDIES ON ALCOHOL AND DRUGS 2020; 37:153-171. [PMID: 32934599 PMCID: PMC7434174 DOI: 10.1177/1455072520905404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
Aim: To investigate maternal alcohol intakes before and during pregnancy, their impact on mothers and infants to 18 months. Method: Prospective study of 504 New Zealand volunteers visited in months 4 and 7 of pregnancy, measurements taken, lifestyle details recorded including alcohol intake before and during pregnancy. Eighteen months after birth, 370 infants were measured, and infant development recorded. Results: Nineteen per cent of mothers never drank, 53% stopped when they knew they were pregnant, 29% continued to drink. Twenty-two per cent of drinkers binge drank (over 50 g alcohol per session) before pregnancy and 10% during pregnancy. Daily drinking was associated with increased obesity in mothers. Alcohol consumption before or during pregnancy was not associated with infant motor development, had a slight negative effect on growth, and a significant association with vocal ability to 18 months. Energy intake appeared to partially moderate this effect. Conclusion: Maternal alcohol consumption exceeding 50 g per session both before and during pregnancy was associated with decreasing vocal ability in the 18-month old infant.
Collapse
|
40
|
Abozaid A, Trzuskot L, Najmi Z, Paul I, Tsang B, Gerlai R. Developmental stage and genotype dependent behavioral effects of embryonic alcohol exposure in zebrafish larvae. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109774. [PMID: 31655157 DOI: 10.1016/j.pnpbp.2019.109774] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/29/2019] [Accepted: 10/02/2019] [Indexed: 01/01/2023]
Abstract
Fetal Alcohol Spectrum Disorders (FASD) represent a worldwide problem. The severity and types of symptoms of FASD vary, which may be due to the genotype of the fetus and the developmental stage at which the fetus is exposed to alcohol. The most prevalent forms of FASD present less severe symptoms, including behavioral and cognitive abnormalities, and arise from exposure to low amounts of alcohol consumed infrequently. Treating or diagnosing FASD patients has been difficult because we do not understand the mechanisms underlying FASD. Animal models, including the zebrafish, have been suggested to answer this question. Here, we present a proof of concept analysis studying the behavioral effects of embryonic alcohol exposure in one-week old juvenile zebrafish. We exposed zebrafish embryos at one of five developmental stages (8, 16, 24, 32, or 40 hour post-fertilization) to 0% (control) or 1% (vol/vol) ethanol for 2 h, and tested the behavior of these fish at their age of 7-9 days post-fertilization. We employed two genetically distinct zebrafish populations, a quasi-inbred AB derivative strain, and a genetically variable WT population. We report significant developmental time and genotype dependent effects of alcohol on certain measures of motor function and/or anxiety-like responses. For example, we found embryonic alcohol exposed AB fish to swim faster, vary their speed more, stop moving more often and turn less compared to control fish, alcohol induced changes that were absent or less robust in WT fish. We conclude that our results open new avenues to the identification of genetic mechanisms that mediate or influence alcohol induced developmental alteration of brain function and behavior, which, on the long run, may allow us to identify diagnostic biomarkers and treatment options for human FASD.
Collapse
Affiliation(s)
- Amira Abozaid
- Department of Psychology, University of Toronto Mississauga, Canada
| | - Lidia Trzuskot
- Department of Psychology, University of Toronto Mississauga, Canada
| | - Zelaikha Najmi
- Department of Biology, University of Toronto Mississauga, Canada
| | - Ishti Paul
- Department of Biology, University of Toronto Mississauga, Canada
| | - Benjamin Tsang
- Department of Psychology, University of Toronto Mississauga, Canada
| | - Robert Gerlai
- Department of Psychology, University of Toronto Mississauga, Canada; Department of Cell & System Biology, University of Toronto, Canada.
| |
Collapse
|
41
|
Lovely CB. Quantification of Ethanol Levels in Zebrafish Embryos Using Head Space Gas Chromatography. J Vis Exp 2020. [PMID: 32116298 DOI: 10.3791/60766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describe a highly variable continuum of ethanol-induced developmental defects, including facial dysmorphologies and neurological impairments. With a complex pathology, FASD affects approximately 1 in 100 children born in the United States each year. Due to the highly variable nature of FASD, animal models have proven critical in our current mechanistic understanding of ethanol-induced development defects. An increasing number of laboratories has focused on using zebrafish to examine ethanol-induced developmental defects. Zebrafish produce large numbers of externally fertilized, genetically tractable, translucent embryos. This allows researchers to precisely control timing and dosage of ethanol exposure in multiple genetic contexts and quantify the impact of embryonic ethanol exposure through live imaging techniques. This, combined with the high degree of conservation of both genetics and development with humans, has proven zebrafish to be a powerful model in which to study the mechanistic basis of ethanol teratogenicity. However, ethanol exposure regimens have varied between different zebrafish studies, which has confounded the interpretation of zebrafish data across these studies. Here is a protocol to quantify ethanol concentrations in zebrafish embryos using head space gas chromatography.
Collapse
Affiliation(s)
- C Ben Lovely
- Department of Biochemistry and Molecular Genetics, Alcohol Research Center, University of Louisville;
| |
Collapse
|
42
|
Fainsod A, Bendelac-Kapon L, Shabtai Y. Fetal Alcohol Spectrum Disorder: Embryogenesis Under Reduced Retinoic Acid Signaling Conditions. Subcell Biochem 2020; 95:197-225. [PMID: 32297301 DOI: 10.1007/978-3-030-42282-0_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a complex set of developmental malformations, neurobehavioral anomalies and mental disabilities induced by exposing human embryos to alcohol during fetal development. Several experimental models and a series of developmental and biochemical approaches have established a strong link between FASD and reduced retinoic acid (RA) signaling. RA signaling is involved in the regulation of numerous developmental decisions from patterning of the anterior-posterior axis, starting at gastrulation, to the differentiation of specific cell types within developing organs, to adult tissue homeostasis. Being such an important regulatory signal during embryonic development, mutations or environmental perturbations that affect the level, timing or location of the RA signal can induce multiple and severe developmental malformations. The evidence connecting human syndromes to reduced RA signaling is presented here and the resulting phenotypes are compared to FASD. Available data suggest that competition between ethanol clearance and RA biosynthesis is a major etiological component in FASD.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel.
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| |
Collapse
|
43
|
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: 26] [Impact Index Per Article: 6.5] [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
|
44
|
Swartz ME, Lovely CB, McCarthy N, Kuka T, Eberhart JK. Novel Ethanol-Sensitive Mutants Identified in an F3 Forward Genetic Screen. Alcohol Clin Exp Res 2019; 44:56-65. [PMID: 31742718 PMCID: PMC6980918 DOI: 10.1111/acer.14240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/13/2019] [Indexed: 01/25/2023]
Abstract
Background Fetal alcohol spectrum disorders (FASD) collectively refer to all deleterious outcomes due to prenatal alcohol exposures. Alterations to the face are common phenotypes in FASD. While alcohol exposure is the underlying cause of FASD, many variables modify the outcomes of such exposures. Genetic risk is one such variable, yet we still have a limited understanding of the nature of the genetic loci mediating susceptibility to FASD. Methods We employed ENU‐based random mutagenesis in zebrafish to identify mutations that enhanced the teratogenicity of ethanol (EtOH). F3 embryos obtained from 126 inbred F2 families were exposed to 1% EtOH in the medium (approximately 41 mM tissue levels). Zebrafish stained with Alcian Blue and Alizarin Red were screened for qualitative alterations to the craniofacial skeleton between 4 and 7 days postfertilization (dpf). Results In all, we recovered 6 EtOH‐sensitive mutants, 5 from the genetic screen itself and one as a background mutation in one of our wild‐type lines. Each mutant has a unique EtOH‐induced phenotype relative to the other mutant lines. All but 1 mutation appears to be recessive in nature, and only 1 mutant, au29, has apparent craniofacial defects in the absence of EtOH. To validate the genetic screen, we genetically mapped au29 and found that it carries a mutation in a previously uncharacterized gene, si:dkey‐88l16.3. Conclusions The phenotypes of these EtOH‐sensitive mutants differ from those in previous characterizations of gene–EtOH interactions. Thus, each mutant is likely to provide novel insights into EtOH teratogenesis. Given that most of these mutants only have craniofacial defects in the presence of EtOH and our mapping of au29, it is also likely that many of the mutants will be previously uncharacterized. Collectively, our findings point to the importance of unbiased genetic screens in the identification, and eventual characterization, of risk alleles for FASD.
Collapse
Affiliation(s)
- Mary E Swartz
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Austin, Texas
| | - Charles Ben Lovely
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Austin, Texas
| | - Neil McCarthy
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Austin, Texas
| | - Tim Kuka
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Austin, Texas
| | - Johann K Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research, Austin, Texas
| |
Collapse
|
45
|
Montagud-Romero S, Cantacorps L, Valverde O. Histone deacetylases inhibitor trichostatin A reverses anxiety-like symptoms and memory impairments induced by maternal binge alcohol drinking in mice. J Psychopharmacol 2019; 33:1573-1587. [PMID: 31294671 DOI: 10.1177/0269881119857208] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Alcohol exposure during development has detrimental effects, including a wide range of physical, cognitive and neurobehavioural anomalies known as foetal alcohol spectrum disorders. However, alcohol consumption among pregnant woman is an ongoing latent health problem. AIM In the present study, the effects of trichostatin A (TSA) on emotional and cognitive impairments caused by prenatal and lactational alcohol exposure were assessed. TSA is an inhibitor of class I and II histone deacetylases enzymes (HDAC), and for that, HDAC4 activity was determined. We also evaluated mechanisms underlying the behavioural effects observed, including the expression of brain-derived neurotrophic factor (BDNF) in discrete brain regions and newly differentiated neurons in the dentate gyrus (DG). METHODS C57BL/6 female pregnant mice were used, with limited access to a 20% v/v alcohol solution as a procedure to model binge alcohol drinking during gestation and lactation. Male offspring were treated with TSA during the postnatal days (PD28-35) and behaviourally evaluated (PD36-55). RESULTS Early alcohol exposure mice presented increased anxiogenic-like responses and memory deterioration - effects that were partially reversed with TSA. Early alcohol exposure produces a decrease in BDNF levels in the hippocampus (HPC) and prefrontal cortex, a reduction of neurogenesis in the DG and increased activity levels of the HDAC4 in the HPC. CONCLUSIONS Such findings support the participation of HDAC enzymes in cognitive and emotional alterations induced by binge alcohol consumption during gestation and lactation and would indicate potential benefits of HDAC inhibitors for some aspects of foetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Sandra Montagud-Romero
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lídia Cantacorps
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC-NeuroBio), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM-Hospital del Mar Medical Research Institute, Neurosciences Programme, Barcelona, Spain
| |
Collapse
|
46
|
Lovely CB. Animal models of gene-alcohol interactions. Birth Defects Res 2019; 112:367-379. [PMID: 31774246 DOI: 10.1002/bdr2.1623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/09/2019] [Indexed: 12/16/2022]
Abstract
Most birth defects arise from complex interactions between multiple genetic and environmental factors. However, our current understanding of how these interactions and their contributions affect birth defects remains incomplete. Human studies are limited in their ability to identify the fundamental causes of birth defects due to ethical and practical limitations. Animal models provide a great number of resources not available to human studies and they have been critical in advancing our understanding of birth defects and the complex interactions that underlie them. In this review, we discuss the use of animal models in the context of gene-environment interactions that underlie birth defects. We focus on alcohol which is the most common environmental factor associated with birth defects. Prenatal alcohol exposure leads to a wide range of cognitive impairments and structural deficits broadly termed fetal alcohol spectrum disorders (FASD). We discuss the broad impact of prenatal alcohol exposure on the developing embryo and elaborate on the current state of gene-alcohol interactions. Additionally, we discuss how animal models have informed our understanding of the genetics of FASD. Ultimately, these topics will provide insight into the use of animal models in understanding gene-environment interactions and their subsequent impact on birth defects.
Collapse
Affiliation(s)
- Charles Benjamin Lovely
- Department of Biochemistry and Molecular Genetics, Alcohol Research Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
47
|
Cannabinoids Exacerbate Alcohol Teratogenesis by a CB1-Hedgehog Interaction. Sci Rep 2019; 9:16057. [PMID: 31690747 PMCID: PMC6831672 DOI: 10.1038/s41598-019-52336-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 10/16/2019] [Indexed: 01/20/2023] Open
Abstract
We tested whether cannabinoids (CBs) potentiate alcohol-induced birth defects in mice and zebrafish, and explored the underlying pathogenic mechanisms on Sonic Hedgehog (Shh) signaling. The CBs, Δ9-THC, cannabidiol, HU-210, and CP 55,940 caused alcohol-like effects on craniofacial and brain development, phenocopying Shh mutations. Combined exposure to even low doses of alcohol with THC, HU-210, or CP 55,940 caused a greater incidence of birth defects, particularly of the eyes, than did either treatment alone. Consistent with the hypothesis that these defects are caused by deficient Shh, we found that CBs reduced Shh signaling by inhibiting Smoothened (Smo), while Shh mRNA or a CB1 receptor antagonist attenuated CB-induced birth defects. Proximity ligation experiments identified novel CB1-Smo heteromers, suggesting allosteric CB1-Smo interactions. In addition to raising concerns about the safety of cannabinoid and alcohol exposure during early embryonic development, this study establishes a novel link between two distinct signaling pathways and has widespread implications for development, as well as diseases such as addiction and cancer.
Collapse
|
48
|
Blanck-Lubarsch M, Dirksen D, Feldmann R, Sauerland C, Kirschneck C, Hohoff A. 3D Analysis of Philtrum Depth in Children with Fetal Alcohol Syndrome. Alcohol Alcohol 2019; 54:152-158. [PMID: 30608520 DOI: 10.1093/alcalc/agy088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 11/13/2022] Open
Abstract
AIMS Diagnosis of fetal alcohol spectrum disorder (FASD) is complex and difficult. The estimated number of unreported FASD is thus assumed to be substantial. In our cross-sectional study, we aimed to identify possible metric differences in philtrum depth in children with fetal alcohol syndrome (FAS) compared to healthy controls based on non-invasive 3D facial scanning in order to provide an objective, metrical tool improving FASD diagnosis. METHODS Twenty-five children with confirmed FAS and 30 healthy school children without FAS, both in the mixed dentition, were prospectively recruited and 3D facial scans were performed after recording body length, weight and head circumference. Philtrum surface data were extracted and metric philtrum depth was determined at four geometrically defined measuring points (P1-P4) along the vertical length of the philtrum. RESULTS Philtrum depths at P1 (P = 0.025), P2 (P = 0.001), P3 (P < 0.001) and P4 (P = 0.001) as well as mean philtrum depth P1-P4 (P < 0.001) differed significantly between patients with and without FAS. Compared to controls, the philtrum was shallower in patients with FAS by on average 0.4 mm at each of the respective points. Whereas no differences could be determined for body height and weight, head circumference was significantly smaller in patients with FAS (P = 0.001), particularly in girls (P = 0.008). CONCLUSIONS Apart from head circumference, philtrum depth is significantly reduced in children with FAS and can thus be used as diagnostic indicator to aid and confirm FAS diagnosis. In contrast to visual assessments, 3D face scan methods allow a more objective quantification and can thus provide additional evidence in FAS diagnosis.
Collapse
Affiliation(s)
- Moritz Blanck-Lubarsch
- Department of Orthodontics, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Dieter Dirksen
- Department of Prosthodontics and Biomaterials, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Reinhold Feldmann
- Department of Pediatrics, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| | - Cristina Sauerland
- Institute of Biostatistics and Clinical Research, University of Münster, Schmeddingstraße 56, Münster, Germany
| | - Christian Kirschneck
- Department of Orthodontics, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, Germany
| | - Ariane Hohoff
- Department of Orthodontics, University of Münster, Albert-Schweitzer-Campus 1, Münster, Germany
| |
Collapse
|
49
|
Mahnke AH, Adams AM, Wang AZ, Miranda RC. Toxicant and teratogenic effects of prenatal alcohol. CURRENT OPINION IN TOXICOLOGY 2019; 14:29-34. [PMID: 32864517 DOI: 10.1016/j.cotox.2019.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Prenatal alcohol exposure can result in growth, cognitive, and behavioral deficits due to the toxicant and teratogenic effects of alcohol. Alcohol is an unusual toxicant, because, unlike other toxicants, it is consumed and has biological effects in the millimolar range. Cerebral cortical development is particularly vulnerable to both alcohol's acute and long-term reprogramming effects. Recent evidence suggests that neuroinflammation may be a persistent result of prenatal alcohol exposure and that modes of cellular communication capable of carrying miRNAs, such as extracellular vesicles, may be an integral part of long-term changes to cellular communication and inflammation following in utero alcohol exposure.
Collapse
Affiliation(s)
- Amanda H Mahnke
- Texas A&M University Health Science Center, Department of Neuroscience and Experimental Therapeutics, Bryan, TX 77807 USA
| | - Amy M Adams
- Texas A&M University Health Science Center, Department of Neuroscience and Experimental Therapeutics, Bryan, TX 77807 USA
| | - Andrew Z Wang
- Texas A&M University Health Science Center, Department of Neuroscience and Experimental Therapeutics, Bryan, TX 77807 USA
| | - Rajesh C Miranda
- Texas A&M University Health Science Center, Department of Neuroscience and Experimental Therapeutics, Bryan, TX 77807 USA
| |
Collapse
|
50
|
Cobben JM, Krzyzewska IM, Venema A, Mul AN, Polstra A, Postma AV, Smigiel R, Pesz K, Niklinski J, Chomczyk MA, Henneman P, Mannens MMAM. DNA methylation abundantly associates with fetal alcohol spectrum disorder and its subphenotypes. Epigenomics 2019; 11:767-785. [DOI: 10.2217/epi-2018-0221] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Fetal alcohol spectrum disorder (FASD) involves prenatal growth delay, impaired facial and CNS development and causes severe clinical, social-economic burdens. Here, we aim to detect DNA-methylation aberrations associated with FASD and potential FASD diagnostic and prognostic biomarkers. Patients & methods: The FASD diagnosis was established according to golden-standard protocols in a discovery and independent replication cohort. Genome-wide differential methylation association and replication analyses were performed. Results: We identified several loci that were robustly associated with FASD or one of its sub phenotypes. Our findings were evaluated using previously reported genome-wide surveys. Conclusion: We have detected robust FASD associated differentially methylated positions and differentially methylated regions for FASD in general and for FASD subphenotypes, in other words on growth delay, impaired facial and CNS development.
Collapse
Affiliation(s)
- Jan Maarten Cobben
- Department of Pediatrics, Amsterdam University Medical Centers, Location AMC, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Izabela M Krzyzewska
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Andrea Venema
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Adri N Mul
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Abeltje Polstra
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Alex V Postma
- Department of Clinical Genetics, Genome Diagnostics Laboratory, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Robert Smigiel
- Department of Pediatrics & Rare Disorders, Medical University of Wroclaw, Poland
| | - Karolina Pesz
- Department of Genetics, Medical University of Wroclaw, Poland
| | - Jacek Niklinski
- Department of Molecular Biology, Medical University of Bialystok, Poland
| | - Monika A Chomczyk
- Department of Molecular Biology, Medical University of Bialystok, Poland
| | - Peter Henneman
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Marcel MAM Mannens
- Department of Anatomy, Embryology & Physiology, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| |
Collapse
|