1
|
Quintrell E, Russell DJ, Rahmannia S, Wyrwoll CS, Larcombe A, Kelty E. The Safety of Alcohol Pharmacotherapies in Pregnancy: A Scoping Review of Human and Animal Research. CNS Drugs 2024:10.1007/s40263-024-01126-8. [PMID: 39388037 DOI: 10.1007/s40263-024-01126-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND OBJECTIVE Alcohol pharmacotherapies pose unknown teratogenic risks in pregnancy and are therefore recommended to be avoided. This limits treatment options for pregnant individuals with alcohol use disorders (AUD). The information on the safety of these medications during pregnancy is uncertain, prompting a scoping review. The objective of this review was to investigate available information on the safety of alcohol pharmacotherapies in pregnancy. METHODS Studies published between January 1990 and July 2023 were identified through searches in BIOSIS, Embase, PsycINFO and MEDLINE databases, using terms related to pregnancy and alcohol pharmacotherapies. The alcohol pharmacotherapies investigated were naltrexone, acamprosate, disulfiram, nalmefene, baclofen, gabapentin and topiramate. Studies were screened by two independent reviewers. Covidence software facilitated the management, screening and extraction of studies. RESULTS A total of 105 studies were included in the review (naltrexone: 21, acamprosate: 4, disulfiram: 3, baclofen: 3, nalmefene: 0, topiramate: 55, gabapentin: 32) with some studies investigating multiple medications. Studies investigating naltrexone's safety in pregnancy focussed on opioid use disorders, with limited evidence regarding its safety in the context of AUD. Despite concerns about higher rates of some pregnancy complications, studies generally indicate naltrexone as a safer option compared with opioid agonists or alcohol during pregnancy. Acamprosate was not clearly associated with adverse effects of exposure in pregnancy, with two pre-clinical studies suggesting potential neuroprotective properties. Disulfiram has a high risk of congenital anomalies when used in pregnancy, believed to be due to its mechanism of action. Prenatal topiramate has also been associated with an increased risk of congenital anomalies, particularly oral clefts. There were mixed results concerning the safety of prenatal gabapentin and little to no literature investigating the safety of baclofen or nalmefene during pregnancy. CONCLUSIONS There is insufficient research on the safety of alcohol pharmacotherapies in pregnancy. Despite this, given alcohol's teratogenic effects, naltrexone could be considered to help maintain abstinence in pregnant individuals with AUD, particularly when psychosocial treatments have failed.
Collapse
Affiliation(s)
- Ebony Quintrell
- School of Population and Global Health, University of Western Australia, Nedlands, WA, 6009, Australia.
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia.
| | - Danielle J Russell
- School of Population and Global Health, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Sofa Rahmannia
- School of Population and Global Health, University of Western Australia, Nedlands, WA, 6009, Australia
| | - Caitlin S Wyrwoll
- The Kids Research Institute Australia, Nedlands, WA, Australia
- School of Human Sciences, University of Western Australia, Crawley, WA, Australia
| | - Alexander Larcombe
- Respiratory Environmental Health, Wal-yan Respiratory Research Centre, The Kids Research Institute Australia, Nedlands, WA, Australia
- Occupation, Environment and Safety, School of Population Health, Curtin University, Bentley, WA, Australia
| | - Erin Kelty
- School of Population and Global Health, University of Western Australia, Nedlands, WA, 6009, Australia
| |
Collapse
|
2
|
Boschen KE, Dragicevich CJ, Fish EW, Hepperla AJ, Simon JM, Parnell SE. Gastrulation-stage alcohol exposure induces similar rates of craniofacial malformations in male and female C57BL/6J mice. Birth Defects Res 2024; 116:e2292. [PMID: 38116840 PMCID: PMC10872400 DOI: 10.1002/bdr2.2292] [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: 07/31/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Prenatal alcohol exposure during gastrulation (embryonic day [E] 7 in mice, ~3rd week of human pregnancy) impairs eye, facial, and cortical development, recapitulating birth defects characteristic of Fetal Alcohol Syndrome (FAS). However, it is not known whether the prevalence or severity of craniofacial features associated with FAS is affected by biological sex. METHODS The current study administered either alcohol (2.9 g/kg, two i.p. doses, 4 hr apart) or vehicle to pregnant C57BL/6J females on E7, prior to gonadal sex differentiation, and assessed fetal morphology at E17. RESULTS Whereas sex did not affect fetal size in controls, alcohol-exposed females were smaller than both control females and alcohol-treated males. Alcohol exposure increased the incidence of eye defects to a similar degree in males and females. Together, these data suggest that females might be more sensitive to the general developmental effects of alcohol, but not effects specific to the craniofacies. Whole transcriptomic analysis of untreated E7 embryos found 214 differentially expressed genes in females vs. males, including those in pathways related to cilia and mitochondria, histone demethylase activity, and pluripotency. CONCLUSION Gastrulation-stage alcohol induces craniofacial malformations in male and female mouse fetuses at similar rates and severity, though growth deficits are more prevalent females. These findings support the investigation of biological sex as a contributing factor in prenatal alcohol studies.
Collapse
Affiliation(s)
- Karen E. Boschen
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Constance J. Dragicevich
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric W. Fish
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Austin J. Hepperla
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeremy M. Simon
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott E. Parnell
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
3
|
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
|
4
|
Saito M, Subbanna S, Zhang X, Canals-Baker S, Smiley JF, Wilson DA, Das BC. Effects of retinoic acid receptor α modulators on developmental ethanol-induced neurodegeneration and neuroinflammation. Front Neurosci 2023; 17:1170259. [PMID: 37205047 PMCID: PMC10187544 DOI: 10.3389/fnins.2023.1170259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/07/2023] [Indexed: 05/21/2023] Open
Abstract
Ethanol exposure in neonatal mice induces acute neurodegeneration followed by long-lasting glial activation and GABAergic cell deficits along with behavioral abnormalities, providing a third trimester model of fetal alcohol spectrum disorders (FASD). Retinoic acid (RA), the active form of vitamin A, regulates transcription of RA-responsive genes and plays essential roles in the development of embryos and their CNS. Ethanol has been shown to disturb RA metabolism and signaling in the developing brain, which may be a cause of ethanol toxicity leading to FASD. Using an agonist and an antagonist specific to RA receptor α (RARα), we studied how RA/RARα signaling affects acute and long-lasting neurodegeneration and activation of phagocytic cells and astrocytes caused by ethanol administered to neonatal mice. We found that an RARα antagonist (BT382) administered 30 min before ethanol injection into postnatal day 7 (P7) mice partially blocked acute neurodegeneration as well as elevation of CD68-positive phagocytic cells in the same brain area. While an RARα agonist (BT75) did not affect acute neurodegeneration, BT75 given either before or after ethanol administration ameliorated long-lasting astrocyte activation and GABAergic cell deficits in certain brain regions. Our studies using Nkx2.1-Cre;Ai9 mice, in which major GABAergic neurons and their progenitors in the cortex and the hippocampus are labeled with constitutively expressed tdTomato fluorescent protein, indicate that the long-lasting GABAergic cell deficits are mainly caused by P7 ethanol-induced initial neurodegeneration. However, the partial reduction of prolonged GABAergic cell deficits and glial activation by post-ethanol BT75 treatment suggests that, in addition to the initial cell death, there may be delayed cell death or disturbed development of GABAergic cells, which is partially rescued by BT75. Since RARα agonists including BT75 have been shown to exert anti-inflammatory effects, BT75 may rescue GABAergic cell deficits by reducing glial activation/neuroinflammation.
Collapse
Affiliation(s)
- Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
- *Correspondence: Mariko Saito,
| | - Shivakumar Subbanna
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Xiuli Zhang
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Stefanie Canals-Baker
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - John F. Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Donald A. Wilson
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
- Department of Child and Adolescent Psychiatry, New York University Medical Center, New York, NY, United States
| | - Bhaskar C. Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, United States
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Bhaskar C. Das,
| |
Collapse
|
5
|
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
|
6
|
Chen Z, Li S, Guo L, Peng X, Liu Y. Prenatal alcohol exposure induced congenital heart diseases: From bench to bedside. Birth Defects Res 2020; 113:521-534. [PMID: 32578335 DOI: 10.1002/bdr2.1743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 12/27/2022]
Abstract
Alcohol consumption is increasing worldwide. Many child-bearing-aged women consume alcohol during pregnancy, intentionally or unintentionally, thereby increasing the potential risk for severe congenital diseases. Congenital heart disease (CHD) is the most common birth defect worldwide and can result from both hereditary and acquired factors. Prenatal alcohol exposure (PAE) is considered a key factor that leads to teratogenesis in CHD and its specific phenotypes, especially defects of the cardiac septa, cardiac valves, cardiac canals, and great arteries, adjacent to the chambers, both in animal experiments and clinical retrospective studies. The mechanisms underlying CHD and its phenotypes caused by PAE are associated with changes in retinoic acid biosynthesis and its signaling pathway, apoptosis and defective function of cardiac neural crest cells, disturbance of the Wntβ-catenin signaling pathway, suppression of bone morphogenetic protein (BMP) signaling, and other epigenetic mechanisms. Drug supplements and early diagnosis can help prevent PAE from inducing CHDs.
Collapse
Affiliation(s)
- Zhiyan Chen
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Sheng Li
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Linghong Guo
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Xu Peng
- Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China
| | - Yin Liu
- Department of Basic Medical Sciences, Sichuan Vocational College of Health and Rehabilitation, Zigong, Sichuan, China.,Department of Research, Zigong First People's Hospital, Zigong, Sichuan, China.,Department of Pharmacology, West China School of Basic Sciences & Forensic Medicine; Animal Research Institute, Sichuan University, Chengdu, Sichuan, China.,Department of Anesthesiology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Abstract
BACKGROUND After fluorochromes are incorporated into cells, tissues, and organisms, confocal microscopy can be used to observe three-dimensional structures. LysoTracker Red (LT) is a paraformaldehyde-fixable probe that concentrates into acidic compartments of cells and indicates regions of high lysosomal activity and phagocytosis, both of which correlate to apoptotic activity. Thus, LT is a good indicator of apoptosis visualized by confocal microscopy. Results of LT staining of apoptotic cell death correlate well with other whole mount apoptosis vital dyes such as Nile blue sulfate and neutral red, with the added benefit of being fixable in situ. Nile blue sulfate can also be used as a non-vital, nonspecific dye to visualize general morphology. Stains such as acridine orange can be used for surface staining of fixed embryos to yield confocal images that are similar to scanning electron micrographs. METHODS Mouse embryos were stained with LT, fixed with paraformaldehyde/glutaraldehyde, dehydrated with methanol (MEOH), and cleared with benzyl alcohol/benzyl benzoate (BABB). Following this treatment, the tissues were nearly transparent. Embryos are mounted on depression slides, and serial sections are imaged by confocal microscopy, followed by 3-D reconstruction. RESULTS Embryos or tissues as thick as 500 microns (μm) can be visualized after clearing with BABB. LysoTracker staining reveals apoptotic regions in organogenesis-stage mouse embryos. Morphological observation of tissue was facilitated by combining autofluorescence with Nile blue sulfate staining of fixed embryos or opaque surface staining with acridine orange staining. CONCLUSIONS The use of BABB for clearing LT vital-stained and fixed embryos matches the refractive index of the tissue to the suspending medium, allowing increased penetration of laser light in a confocal microscope. Nile blue sulfate used as a non-vital dye provides a nonspecific staining of fixed embryos that can then be cleared with methyl salicylate for confocal observation. Sample preparation and staining procedures described here, with optimization of confocal laser scanning microscopy, allow for the detection and visualization of morphological structure and apoptosis in embryos up to 500 μm thick, and stained specimens can be fixed and mounted on depression slides.
Collapse
|
8
|
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
|
9
|
Petrelli B, Bendelac L, Hicks GG, Fainsod A. Insights into retinoic acid deficiency and the induction of craniofacial malformations and microcephaly in fetal alcohol spectrum disorder. Genesis 2019; 57:e23278. [DOI: 10.1002/dvg.23278] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Berardino Petrelli
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Liat Bendelac
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| | - Geoffrey G. Hicks
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| |
Collapse
|
10
|
Boschen KE, Gong H, Murdaugh LB, Parnell SE. Knockdown of Mns1 Increases Susceptibility to Craniofacial Defects Following Gastrulation-Stage Alcohol Exposure in Mice. Alcohol Clin Exp Res 2018; 42:2136-2143. [PMID: 30129265 PMCID: PMC6214710 DOI: 10.1111/acer.13876] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND MNS1 (meiosis-specific nuclear structural protein 1) is necessary for motile cilia function, such as sperm flagella or those found in the embryonic primitive node. While little is known regarding the function or expression pattern of MNS1 in the embryo, co-immunoprecipitation experiments in sperm have determined that MNS1 interacts with ciliary proteins, which are also important during development. Establishment of morphogenic gradients is dependent on normal ciliary motion in the primitive node beginning during gastrulation (gestational day [GD] 7 in the mouse, second-third week of pregnancy in humans), a critical window for face, eye, and brain development and particularly susceptible to perturbations of developmental signals. The current study investigates the role of Mns1 in craniofacial defects associated with gastrulation-stage alcohol exposure. METHODS On GD7, pregnant Mns1+/- dams were administered 2 doses of ethanol (5.8 g/kg total) or vehicle 4 hours apart to target gastrulation. On GD17, fetuses were examined for ocular defects by scoring each eye on a scale from 1 to 7 (1 = normal, 2 to 7 = defects escalating in severity). Craniofacial and brain abnormalities were also assessed. RESULTS Prenatal alcohol exposure (PAE) significantly increased the rate of defects in wild-type fetuses, as PAE fetuses had an incidence rate of 41.18% compared to a 10% incidence rate in controls. Furthermore, PAE interacted with genotype to significantly increase the defect rate and severity in Mns1+/- (64.29%) and Mns1-/- mice (92.31%). PAE Mns1-/- fetuses with severe eye defects also presented with craniofacial dysmorphologies characteristic of fetal alcohol syndrome and midline tissue loss in the brain, palate, and nasal septum. CONCLUSIONS These data demonstrate that a partial or complete knockdown of Mns1 interacts with PAE to increase the susceptibility to ocular defects and correlating craniofacial and brain anomalies, likely though interaction of alcohol with motile cilia function. These results further our understanding of genetic risk factors that may underlie susceptibility to teratogenic exposures.
Collapse
Affiliation(s)
- Karen E. Boschen
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599
| | - Henry Gong
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599
| | - Laura B. Murdaugh
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599
| | - Scott E. Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC 27599
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
11
|
Muralidharan P, Sarmah S, Marrs JA. Retinal Wnt signaling defect in a zebrafish fetal alcohol spectrum disorder model. PLoS One 2018; 13:e0201659. [PMID: 30067812 PMCID: PMC6070267 DOI: 10.1371/journal.pone.0201659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/19/2018] [Indexed: 12/11/2022] Open
Abstract
Fetal alcohol spectrum disorder caused by prenatal alcohol exposure includes ocular abnormalities (microphthalmia, photoreceptor dysfunction, cataracts). Zebrafish embryos exposed to ethanol from gastrulation through somitogenesis show severe ocular defects, including microphthalmia and photoreceptor differentiation defects. Ethanol-treated zebrafish had an enlarged ciliary marginal zone (CMZ) relative to the retina size and reduced Müller glial cells (MGCs). Ethanol exposure produced immature photoreceptors with increased proliferation, indicating cell cycle exit failure. Signaling mechanisms in the CMZ were affected by embryonic ethanol exposure, including Wnt signaling in the CMZ, Notch signaling and neurod gene expression. Retinoic acid or folic acid co-supplementation with ethanol rescued Wnt signaling and retinal differentiation. Activating Wnt signaling using GSK3 inhibitor (LSN 2105786; Eli Lilly and Co.) restored retinal cell differentiation pathways. Ethanol exposed embryos were treated with Wnt agonist, which rescued Wnt-active cells in the CMZ, Notch-active cells in the retina, proliferation, and photoreceptor terminal differentiation. Our results illustrate the critical role of Wnt signaling in ethanol-induced retinal defects.
Collapse
Affiliation(s)
- Pooja Muralidharan
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| |
Collapse
|
12
|
Parnell SE, Riley EP, Warren KR, Mitchell KT, Charness ME. The contributions of Dr. Kathleen K. Sulik to fetal alcohol spectrum disorders research and prevention. Alcohol 2018; 69:15-24. [PMID: 29571046 DOI: 10.1016/j.alcohol.2017.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 10/17/2022]
Abstract
Dr. Kathleen Sulik (Kathy) has spent 35 years studying fetal alcohol syndrome (FAS) and fetal alcohol spectrum disorders (FASD). Beginning with her landmark 1981 Science paper describing the early gestational window when alcohol can cause the craniofacial malformations characteristic of FAS, Kathy has contributed a vast amount of research furthering our knowledge of FASD. After her seminal work that definitively demonstrated that alcohol is the causative factor in FAS, she and her lab went on to explore and define the stage-dependent effects of early gestational alcohol exposure on the face and brain in numerous different ways throughout her career. She explored and discovered numerous mechanisms of alcohol's effects on the embryo, as well as describing several genetic factors that can modify susceptibility to developmental alcohol exposure. She did not restrict her research to the face and brain; her lab described in intricate detail the effects of developmental alcohol exposure on many different organs, including the heart, ears, kidneys, and limbs. In addition to her research, and in conjunction with NIAAA and the National Organization on Fetal Alcohol Syndrome (NOFAS), Kathy developed several FASD prevention curricula that are still in use today. Finally, as part of her drive to eradicate FAS and FASD, Kathy labored tirelessly with public policy makers to change how FASD is viewed by the public, how FASD is identified in affected individuals, and how FASD is studied by researchers. While no article could fully cover Kathy's contributions to FASD research and prevention, or her other contributions to embryology and teratology, this review will attempt to illustrate some of the highlights of Kathy's remarkable career.
Collapse
|
13
|
Shabtai Y, Fainsod A. Competition between ethanol clearance and retinoic acid biosynthesis in the induction of fetal alcohol syndrome. Biochem Cell Biol 2018; 96:148-160. [DOI: 10.1139/bcb-2017-0132] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Several models have been proposed to explain the neurodevelopmental syndrome induced by exposure of human embryos to alcohol, which is known as fetal alcohol spectrum disorder (FASD). One of the proposed models suggests a competition for the enzymes required for the biosynthesis of retinoic acid. The outcome of such competition is development under conditions of reduced retinoic acid signaling. Retinoic acid is one of the biologically active metabolites of vitamin A (retinol), and regulates numerous embryonic and differentiation processes. The developmental malformations characteristic of FASD resemble those observed in vitamin A deficiency syndrome as well as from inhibition of retinoic acid biosynthesis or signaling in experimental models. There is extensive biochemical and enzymatic overlap between ethanol clearance and retinoic acid biosynthesis. Several lines of evidence suggest that in the embryo, the competition takes place between acetaldehyde and retinaldehyde for the aldehyde dehydrogenase activity available. In adults, this competition also extends to the alcohol dehydrogenase activity. Ethanol-induced developmental defects can be ameliorated by increasing the levels of retinol, retinaldehyde, or retinaldehyde dehydrogenase. Acetaldehyde inhibits the production of retinoic acid by retinaldehyde dehydrogenase, further supporting the competition model. All of the evidence supports the reduction of retinoic acid signaling as the etiological trigger in the induction of FASD.
Collapse
Affiliation(s)
- Yehuda Shabtai
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Abraham Fainsod
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
- Department of Cellular Biochemistry and Cancer Research, Institute for Medical Research Israel–Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
14
|
Fish EW, Parnell SE, Sulik KK, Baker LK, Murdaugh LB, Lamson D, Williams KP. Preaxial polydactyly following early gestational exposure to the smoothened agonist, SAG, in C57BL/6J mice. Birth Defects Res 2018; 109:49-54. [PMID: 27801979 DOI: 10.1002/bdra.23571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/01/2016] [Accepted: 08/29/2016] [Indexed: 01/13/2023]
Abstract
BACKGROUND While pharmacological activation of the Hedgehog (HH) signaling pathway may have therapeutic benefits for developmental and adult diseases, its teratogenic potential is of concern. The membrane molecule Smoothened (SMO) transduces HH signaling and can be acutely modulated by antagonists and agonists. The objective of the current experiments was to determine how maternal treatment with the Smo agonist, SAG, affects the developing limb. METHODS Pregnant C57BL/6J mice received a single injection of SAG (15, 17, or 20 mg/kg, i.p.) or its vehicle on gestational day (GD) 9.25, the time of limb bud induction. Embryos were examined on GD 15 for gross dysmorphology and skeletal staining was performed to visualize the number and type of digits on the fore- and hindlimbs. Additionally, in situ hybridization was performed 4 hr after GD 9.25 SAG administration to determine SAG's effects on Gli1 and Gli2 mRNA expression. RESULTS The most prevalent effect of SAG was the dose-dependent induction of pre-axial polydactyly; defects ranged from a broad thumb to the duplication of two finger-like digits on the preaxial side of the thumb. The highest SAG dose was effective in ca. 80% of the embryos and increased Gli1 and Gli2 mRNA expression in the limb bud, with Gli1 mRNA being the most upregulated. CONCLUSION Preaxial polydactyly can be caused in the developing embryo by acute maternal administration of a Smo agonist that activates HH signaling. These results are consistent with the preaxial polydactyly induced in developmental disorders associated with mutations in HH signaling genes.Birth Defects Research 109:49-54, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen K Sulik
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina
| | - Lorinda K Baker
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina
| | - Laura B Murdaugh
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina
| | - David Lamson
- Department of Pharmaceutical Sciences, BRITE Institute, North Carolina Central University, Durham, North Carolina
| | - Kevin P Williams
- Department of Pharmaceutical Sciences, BRITE Institute, North Carolina Central University, Durham, North Carolina
| |
Collapse
|
15
|
Coll TA, Chaufan G, Pérez-Tito L, Ventureira MR, Sobarzo CMA, Ríos de Molina MDC, Cebral E. Oxidative stress and cellular and tissue damage in organogenic outbred mouse embryos after moderate perigestational alcohol intake. Mol Reprod Dev 2017; 84:1086-1099. [DOI: 10.1002/mrd.22865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/08/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Tamara A. Coll
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires; Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE); Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - Gabriela Chaufan
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN); Departamento de Química Biológica; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - Leticia Pérez-Tito
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires; Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE); Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - Martín R. Ventureira
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires; Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA-CONICET); Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - Cristian M. A. Sobarzo
- Universidad de Buenos Aires, Facultad de Medicina, CONICET- Universidad de Buenos Aires; Instituto de Investigaciones Biomédicas (INBIOMED); Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - María del Carmen Ríos de Molina
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN); Departamento de Química Biológica; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| | - Elisa Cebral
- Universidad de Buenos Aires; Facultad de Ciencias Exactas y Naturales; Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
- CONICET-Universidad de Buenos Aires; Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA-CONICET); Ciudad Autonoma de Buenos Aires Buenos Aires Argentina
| |
Collapse
|
16
|
Fish EW, Murdaugh LB, Sulik KK, Williams KP, Parnell SE. Genetic vulnerabilities to prenatal alcohol exposure: Limb defects in sonic hedgehog and GLI2 heterozygous mice. Birth Defects Res 2017; 109:860-865. [PMID: 28504423 DOI: 10.1002/bdr2.1026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Genetic factors influence the physical and neurobehavioral manifestations of prenatal alcohol exposure (PAE). Animal models allow the investigation of specific genes that confer vulnerability to, or protection from, birth defects associated with fetal alcohol spectrum disorders (FASDs). The objective of the present experiments was to determine if genetic alterations in the Sonic Hedgehog (Shh) signaling pathways affect the vulnerability to PAE-induced skeletal defects involving the forelimbs and/or hindlimbs. METHOD Wild-type C57BL/6J female mice were bred with males in which one copy of the Shh or Gli2 genes had been knocked out, to produce litters with both wild-type (+/+) and heterozygous (+/-) embryos. Alcohol doses (two injections of 2.9 g/kg, 4 hours apart) or vehicles were administered starting at gestational day (GD) 9.25, 9.5, or 9.75, a critical exposure time for inducing limb defects. Limb defects were examined at GD 17 using a dysmorphology scale based on abnormalities ranging from increased interdigital spacing to the deletion of multiple fingers and the ulna. RESULTS Alcohol treatment caused a high incidence of forelimb defects, particularly on the right side, that was higher in Shh+/- and Gli2+/- fetuses compared to wild-type fetuses. Dysmorphology scores were also significantly higher in the Shh+/- and Gli2+/- mice. CONCLUSIONS These results extend previous findings demonstrating enhanced sensitivity to PAE-induced craniofacial dysmorphology and support the hypothesis that genetic alterations in the Shh signaling pathway influences the vulnerability to alcohol-induced birth defects. Moreover, these results emphasize the importance of understanding the interactions between genes and prenatal exposure to alcohol or other teratogens. Birth Defects Research 109:860-865, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina
| | - Laura B Murdaugh
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen K Sulik
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina
| | - Kevin P Williams
- Department of Pharmaceutical Sciences, BRITE Institute, North Carolina Central University, Durham, North Carolina
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
17
|
Eberhart JK, Parnell SE. The Genetics of Fetal Alcohol Spectrum Disorders. Alcohol Clin Exp Res 2016; 40:1154-65. [PMID: 27122355 DOI: 10.1111/acer.13066] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/04/2016] [Indexed: 12/29/2022]
Abstract
The term "fetal alcohol spectrum disorders" (FASD) defines the full range of ethanol (EtOH)-induced birth defects. Numerous variables influence the phenotypic outcomes of embryonic EtOH exposure. Among these variables, genetics appears to play an important role, yet our understanding of the genetic predisposition to FASD is still in its infancy. We review the current literature that relates to the genetics of FASD susceptibility and gene-EtOH interactions. Where possible, we comment on potential mechanisms of reported gene-EtOH interactions. Early indications of genetic sensitivity to FASD came from human and animal studies using twins or inbred strains, respectively. These analyses prompted searches for susceptibility loci involved in EtOH metabolism and analyses of candidate loci, based on phenotypes observed in FASD. More recently, genetic screens in animal models have provided an additional insight into the genetics of FASD. Understanding FASD requires that we understand the many factors influencing phenotypic outcome following embryonic EtOH exposure. We are gaining ground on understanding some of the genetics behind FASD, yet much work remains to be carried out. Coordinated analyses using human patients and animal models are likely to be highly fruitful in uncovering the genetics behind FASD.
Collapse
Affiliation(s)
- Johann K Eberhart
- Department of Molecular Biosciences, Institute for Cell and Molecular Biology, Institute for Neuroscience, Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, Texas
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
18
|
Muralidharan P, Sarmah S, Marrs JA. Zebrafish retinal defects induced by ethanol exposure are rescued by retinoic acid and folic acid supplement. Alcohol 2015; 49:149-63. [PMID: 25541501 DOI: 10.1016/j.alcohol.2014.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 11/01/2014] [Accepted: 11/12/2014] [Indexed: 12/13/2022]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is caused by prenatal alcohol exposure, producing craniofacial, sensory, motor, and cognitive defects. FASD is highly prevalent in low socioeconomic populations, which are frequently accompanied by malnutrition. FASD-associated ocular pathologies include microphthalmia, optic nerve hypoplasia, and cataracts. The present study characterizes specific retinal tissue defects, identifies ethanol-sensitive stages during retinal development, and dissects the effect of nutrient supplements, such as retinoic acid (RA) and folic acid (FA) on ethanol-induced retinal defects. Exposure to pathophysiological concentrations of ethanol (during midblastula transition through somitogenesis; 2-24 h post fertilization [hpf]) altered critical transcription factor expression involved in retinal cell differentiation, and produced severe retinal ganglion cell, photoreceptor, and Müller glial differentiation defects. Ethanol exposure did not alter retinal cell differentiation induction, but increased retinal cell death and proliferation. RA and FA nutrient co-supplementation rescued retinal photoreceptor and ganglion cell differentiation defects. Ethanol exposure during retinal morphogenesis stages (16-24 hpf) produced retinal defects like those seen with ethanol exposure between 2 and 24 hpf. Significantly, during an ethanol-sensitive time window (16-24 hpf), RA co-supplementation moderately rescued these defects, whereas FA co-supplementation showed significant rescue of optic nerve and photoreceptor differentiation defects. Interestingly, RA, but not FA, supplementation after ethanol exposure could reverse ethanol-induced optic nerve and photoreceptor differentiation defects. Our results indicate that various ethanol-sensitive events underlie FASD-associated retinal defects. Nutrient supplements like retinoids and folate were effective in alleviating ethanol-induced retinal defects.
Collapse
Affiliation(s)
- Pooja Muralidharan
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Swapnalee Sarmah
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - James A Marrs
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
19
|
Caspers Conway KM, Romitti PA, Holmes L, Olney RS, Richardson SD. Maternal periconceptional alcohol consumption and congenital limb deficiencies. BIRTH DEFECTS RESEARCH. PART A, CLINICAL AND MOLECULAR TERATOLOGY 2014; 100:863-76. [PMID: 25132072 PMCID: PMC4427046 DOI: 10.1002/bdra.23292] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/07/2014] [Accepted: 07/10/2014] [Indexed: 11/08/2022]
Abstract
BACKGROUND Women of childbearing age report high rates of alcohol consumption, which may result in alcohol exposure during early pregnancy. Epidemiological research on congenital limb deficiencies (LDs) and periconceptional exposure to alcohol is inconclusive. METHODS Data from the National Birth Defects Prevention Study (NBDPS) were examined for associations between LDs and patterns of maternal periconceptional (1 month before conception through the first trimester) alcohol consumption among LD case (n = 906) and unaffected control (n = 8352) pregnancies with expected delivery dates from 10/1997 through 12/2007. Adjusted odds ratios (aORs) and 95% confidence intervals were estimated from unconditional logistic regression analysis for all LDs combined, specific LD subtypes (preaxial/terminal transverse), and LD anatomic groups (upper/lower limbs); interactions with folic acid (FA) supplementation were tested. RESULTS When compared with nondrinkers, inverse associations were found between all LDs combined, preaxial, and upper LDs and any reported periconceptional alcohol consumption (aORs ranged from 0.56-0.83), drinking without binging (aORs: 0.53-0.75), and binge drinking (≥4 drinks/occasion) (aORs: 0.64-0.94); however, none of the binge drinking aORs were statistically significant. Stratification by alcohol type showed inverse associations between all LDs combined, preaxial, transverse, and upper and lower LDs for drinking without binging of wine only (aORs: 0.39-0.67) and between all LDs combined and upper LDs for drinking without binging of combinations of alcohol (aORs: 0.63-0.87). FA did not modify observed associations. CONCLUSION Maternal periconceptional alcohol consumption did not emerge as a teratogen for selected LDs in the NBDPS. Future studies should evaluate additional rare LDs among more highly exposed populations.
Collapse
Affiliation(s)
| | - Paul A. Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, Iowa
| | - Lewis Holmes
- Genetics and Teratology Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard S. Olney
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sandra D. Richardson
- Congenital Malformations Registry, Bureau of Environmental and Occupational Epidemiology, New York State Department of Health, Albany, New York
| | | |
Collapse
|
20
|
Sulik KK. Fetal alcohol spectrum disorder: pathogenesis and mechanisms. HANDBOOK OF CLINICAL NEUROLOGY 2014; 125:463-75. [PMID: 25307590 DOI: 10.1016/b978-0-444-62619-6.00026-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
This chapter provides an overview of animal model-based studies that have generated information critical to our understanding of the pathogenesis and mechanisms underlying alcohol-induced birth defects, in particular those involving the brain. Focus is placed on the developing organism itself, rather than the mother, placenta, or other extraembryonic tissues. Components of the cascades of alcohol-induced damage that are considered herein are excessive cell death, changes in the cell cycle and proliferation, cell migration, cell morphogenesis, and gene expression as well as free radical damage and interference with cell signaling. The roles played by one or more of these various factors in the genesis of structural and functional birth defects are dependent upon alcohol exposure patterns and dosage, the involved tissue, and the prenatal stage(s) at the time of exposure. Technologic advances and rapidly increasing knowledge in the fields of genetics, cell, developmental, and neurobiology are critical to accurately piecing together experimental evidence in refining our understanding of the genesis of alcohol-induced birth defects, to the planning and execution of future studies, and to applying the knowledge gained to diminish the severity or occurrence of fetal alcohol spectrum disorder.
Collapse
Affiliation(s)
- Kathleen K Sulik
- Department of Cell Biology and Physiology and Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
21
|
Sarmah S, Marrs JA. Complex cardiac defects after ethanol exposure during discrete cardiogenic events in zebrafish: prevention with folic acid. Dev Dyn 2013; 242:1184-201. [PMID: 23832875 DOI: 10.1002/dvdy.24015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/13/2013] [Accepted: 07/01/2013] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) describes a range of birth defects including various congenital heart defects (CHDs). Mechanisms of FASD-associated CHDs are not understood. Whether alcohol interferes with a single critical event or with multiple events in heart formation is not known. RESULTS Our zebrafish embryo experiments showed that ethanol interrupts different cardiac regulatory networks and perturbs multiple steps of cardiogenesis (specification, myocardial migration, looping, chamber morphogenesis, and endocardial cushion formation). Ethanol exposure during gastrulation until cardiac specification or during myocardial midline migration did not produce severe or persistent heart development defects. However, exposure comprising gastrulation until myocardial precursor midline fusion or during heart patterning stages produced aberrant heart looping and defective endocardial cushions. Continuous exposure during entire cardiogenesis produced complex cardiac defects leading to severely defective myocardium, endocardium, and endocardial cushions. Supplementation of retinoic acid with ethanol partially rescued early heart developmental defects, but the endocardial cushions did not form correctly. In contrast, supplementation of folic acid rescued normal heart development, including the endocardial cushions. CONCLUSIONS Our results indicate that ethanol exposure interrupted divergent cardiac morphogenetic events causing heart defects. Folic acid supplementation was effective in preventing a wide spectrum of ethanol-induced heart developmental defects.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | | |
Collapse
|
22
|
Biomarkers of teratogenesis: Suggestions from animal studies. Reprod Toxicol 2012; 34:180-5. [DOI: 10.1016/j.reprotox.2012.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/03/2012] [Accepted: 05/10/2012] [Indexed: 12/19/2022]
|
23
|
Zuniga A, Zeller R, Probst S. The molecular basis of human congenital limb malformations. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 1:803-22. [PMID: 23799625 DOI: 10.1002/wdev.59] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review focuses predominantly on the human congenital malformations caused by alterations affecting the morphoregulatory gene networks that control early limb bud patterning and outgrowth. Limb defects are among the most frequent congenital malformations in humans that are caused by genetic mutations or teratogenic effects resulting either in abnormal, loss of, or additional skeletal elements. Spontaneous and engineered mouse models have been used to identify and study the molecular alterations and disrupted gene networks that underlie human congenital limb malformations. More recently, mouse genetics has begun to reveal the alterations that affect the often-large cis-regulatory landscapes that control gene expression in limb buds and cause devastating effects on limb bud development. These findings have paved the way to identifying mutations in cis-regulatory regions as causal to an increasing number of congenital limb malformations in humans. In these cases, no mutations in the coding region of a presumed candidate were previously detected. This review highlights how the current understanding of the molecular gene networks and interactions that control mouse limb bud development provides insight into the etiology of human congenital limb malformations.
Collapse
Affiliation(s)
- Aimée Zuniga
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
24
|
Feasibility of Medaka (Oryzias latipes) as an Animal Model to Study Fetal Alcohol Spectrum Disorder. ADVANCES IN MOLECULAR TOXICOLOGY VOLUME 6 2012. [DOI: 10.1016/b978-0-444-59389-4.00003-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Gilliam D, Valdez N, Branson S, Dixon A, Downing C. Maternal effects on ethanol teratogenesis in a cross between A/J and C57BL/6J mice. Alcohol 2011; 45:441-9. [PMID: 21641750 DOI: 10.1016/j.alcohol.2011.02.308] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 02/04/2011] [Accepted: 02/16/2011] [Indexed: 10/18/2022]
Abstract
Genetic factors influence adverse pregnancy outcome in both humans and animal models. Animal research reveals that both the maternal and fetal genetic profiles are important for determining the risk of physical birth defects and prenatal mortality. Using a reciprocal-cross breeding design, we investigated whether the mother's genes may be more important than fetal genes in determining risk for ethanol teratogenesis. Examination of possible synergistic genetic effects on ethanol teratogenesis was made possible by using two mouse strains known to be susceptible to specific malformations. Inbred A/J (A) and C57BL/6J (B6) mice were mated to produce four fetal genotype groups: the true-bred AċA and B6ċB6 genotypes and the genetically identical AċB6 and B6ċA genotypes (the F(1) genotype). Dams were administered either 5.8 g/kg ethanol or an isocaloric amount of maltose-dextrin on day 9 of pregnancy. Fetuses were removed by laparotomy on gestation day 18, weighed, and assessed for digit, vertebral, and kidney malformations. Digit malformations in the genetically identical F(1) ethanol-exposed litters showed a pattern consistent with a maternal genetic effect (AċB6 [2%] and B6ċA [30%]). In contrast, vertebral malformations were similar in all ethanol-exposed litters (AċA [26%], AċB6 [18%], B6ċA [22%], and B6ċB6 [33%]). The percentage of malformations did not differ between male and female fetuses, indicating sex-linked factors are not responsible for the maternal effect. Ethanol exposure decreased litter weights but did not affect litter mortality compared with maltose-exposed controls. This study supports the idea that genes influence malformation risk following in utero alcohol exposure. Specifically, maternal genes influence risk more than fetal genes for some teratogenic outcomes. No evidence supported synergistic genetic effects on ethanol teratogenesis. This research supports the conclusion that uterine environment contributes to determining risk of Fetal Alcohol Spectrum Disorder.
Collapse
|
26
|
Napoli JL. Effects of ethanol on physiological retinoic acid levels. IUBMB Life 2011; 63:701-6. [PMID: 21766417 DOI: 10.1002/iub.500] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 04/20/2011] [Indexed: 12/16/2022]
Abstract
All-trans-retinoic acid (atRA) serves essential functions during embryogenesis and throughout postnatal vertebrate life. Insufficient or excess atRA causes teratogenic and/or toxic effects in the developing embryo: interference with atRA biosynthesis or signaling likely underlies some forms of cancer. Many symptoms of vitamin A (atRA precursor) deficiency and/or toxicity overlap with those of another pleiotropic agent--ethanol. These overlapping symptoms have prompted research to understand whether interference with atRA biosynthesis and/or action may explain (in part) pathology associated with excess ethanol consumption. Ethanol affects many aspects of retinoid metabolism and mechanisms of action site specifically, but no robust data support inhibition of vitamin A metabolism, resulting in decreased atRA in vivo during normal vitamin A nutriture. Actually, ethanol either has no effect on or increases atRA at select sites. Despite this realization, insight into whether interactions between ethanol and retinoids represent cause versus effect requires additional research.
Collapse
Affiliation(s)
- Joseph L Napoli
- Program in Metabolic Biology, Nutritional Science and Toxicology, University of California-Berkeley, Berkeley, CA, USA.
| |
Collapse
|
27
|
Kashyap B, Frey RA, Stenkamp DL. Ethanol-induced microphthalmia is not mediated by changes in retinoic acid or sonic hedgehog signaling during retinal neurogenesis. Alcohol Clin Exp Res 2011; 35:1644-61. [PMID: 21554333 DOI: 10.1111/j.1530-0277.2011.01511.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Microphthalmia (reduced eye size), generally accompanied by vision defects, is a hallmark of fetal alcohol spectrum disorder (FASD) in humans. In zebrafish, embryonic ethanol exposure over the time of retinal neurogenesis also results in microphthalmia. This microphthalmia is in part the consequence of reduced retinal cell differentiation, including photoreceptors. Here we pursue 2 signaling pathways implicated in other aspects of FASD pathogenesis: retinoic acid (RA) and Sonic hedgehog (Shh). METHODS We evaluated markers for RA and Shh signaling within the eyes of embryos treated with ethanol during the period of retinal neurogenesis. We also performed rescue experiments using administration of exogenous RA and microinjection of cholesterol, which augments Shh signaling. RESULTS Using sequential or co-treatments, RA did not rescue ethanol-induced microphthalmia at any concentration tested. In addition, RA itself caused microphthalmia, although the underlying mechanisms were distinct from those of ethanol. Interestingly, RA treatment appeared to recover photoreceptor differentiation in a concentration-dependent manner. This may be an independent effect of exogenous RA, as ethanol treatment alone did not alter RA signaling in the eye. Cholesterol injection also did not rescue ethanol-induced microphthalmia at any concentration tested, and ethanol treatments did not alter expression of shh, or of ptc-2, which is normally regulated by Shh signaling. CONCLUSIONS Together these findings indicate that, during the time of retinal neurogenesis, effects of ethanol on eye development are likely independent of the RA and Shh signaling pathways. These studies suggest that FASD intervention strategies based upon augmentation of RA or Shh signaling may not prevent ethanol-induced microphthalmia.
Collapse
Affiliation(s)
- Bhavani Kashyap
- Department of Biological Sciences, University of Idaho, Moscow, Idaho 83844-3015, USA
| | | | | |
Collapse
|
28
|
Kish PE, Bohnsack BL, Gallina DD, Kasprick DS, Kahana A. The eye as an organizer of craniofacial development. Genesis 2011; 49:222-30. [PMID: 21309065 PMCID: PMC3690320 DOI: 10.1002/dvg.20716] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/03/2011] [Accepted: 01/06/2011] [Indexed: 01/01/2023]
Abstract
The formation and invagination of the optic stalk coincides with the migration of cranial neural crest (CNC) cells, and a growing body of data reveals that the optic stalk and CNC cells communicate to lay the foundations for periocular and craniofacial development. Following migration, the interaction between the developing eye and surrounding periocular mesenchyme (POM) continues, leading to induction of transcriptional regulatory cascades that regulate craniofacial morphogenesis. Studies in chick, mice, and zebrafish have revealed a remarkable level of genetic and mechanistic conservation, affirming the power of each animal model to shed light on the broader morphogenic process. This review will focus on the role of the developing eye in orchestrating craniofacial morphogenesis, utilizing morphogenic gradients, paracrine signaling, and transcriptional regulatory cascades to establish an evolutionarily-conserved facial architecture. We propose that in addition to the forebrain, the eye functions during early craniofacial morphogenesis as a key organizer of facial development, independent of its role in vision.
Collapse
Affiliation(s)
- Phillip E. Kish
- University of Michigan, Ophthalmology and Visual Sciences, Ann Arbor, Michigan, United States,
| | - Brenda L Bohnsack
- University of Michigan, Ophthalmology and Visual Sciences, Ann Arbor, Michigan, United States,
| | - Donika D. Gallina
- University of Michigan, Ophthalmology and Visual Sciences, Ann Arbor, Michigan, United States,
| | - Daniel S. Kasprick
- University of Michigan, Ophthalmology and Visual Sciences, Ann Arbor, Michigan, United States,
| | - Alon Kahana
- University of Michigan, Ophthalmology and Visual Sciences,
| |
Collapse
|
29
|
Marrs JA, Clendenon SG, Ratcliffe DR, Fielding SM, Liu Q, Bosron WF. Zebrafish fetal alcohol syndrome model: effects of ethanol are rescued by retinoic acid supplement. Alcohol 2010; 44:707-15. [PMID: 20036484 DOI: 10.1016/j.alcohol.2009.03.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 03/26/2009] [Accepted: 03/31/2009] [Indexed: 11/30/2022]
Abstract
This study was designed to develop a zebrafish experimental model to examine defects in retinoic acid (RA) signaling caused by embryonic ethanol exposure. RA deficiency may be a causative factor leading to a spectrum of birth defects classified as fetal alcohol spectrum disorder (FASD). Experimental support for this hypothesis using Xenopus showed that effects of treatment with ethanol could be partially rescued by adding retinoids during ethanol treatment. Previous studies show that treating zebrafish embryos during gastrulation and somitogenesis stages with a pathophysiological concentration of ethanol (100mM) produces effects that are characteristic features of FASD. We found that treating zebrafish embryos with RA at a low concentration (10(-9)M) and 100mM ethanol during gastrulation and somitogenesis stages significantly rescued a spectrum of defects produced by treating embryos with 100mM ethanol alone. The rescued phenotype that we observed was quantitatively more similar to embryos treated with 10(-9)M RA alone (RA toxicity) than to untreated or 100mM ethanol-treated embryos. RA rescued defects caused by 100mM ethanol treatment during gastrulation and somitogenesis stages that include early gastrulation cell movements (anterior-posterior axis), craniofacial cartilage formation, and ear development. Morphological evidence also suggests that other characteristic features of FASD (e.g., neural axis patterning) are rescued by RA supplement.
Collapse
Affiliation(s)
- James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Sulik KK. Perspectives on R.E. Shenefelt's 1972 Teratology publication entitled "Morphogenesis of malformations in hamsters caused by retinoic acid: relation to dose and stage at treatment". ACTA ACUST UNITED AC 2010; 89:275-8. [PMID: 20803687 DOI: 10.1002/bdrb.20253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kathleen K Sulik
- Department of Cell and Developmental Biology and Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, North Carolina 27599-7178, USA.
| |
Collapse
|
31
|
Lee GS, Liao X, Shimizu H, Collins MD. Genetic and pathologic aspects of retinoic acid-induced limb malformations in the mouse. ACTA ACUST UNITED AC 2010; 88:863-82. [DOI: 10.1002/bdra.20712] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
32
|
Coyle P, Martin SA, Carey LC, Summers BL, Rofe AM. Ethanol-Mediated Fetal Dysmorphology and its Relationship to the Ontogeny of Maternal Liver Metallothionein. Alcohol Clin Exp Res 2009; 33:1051-8. [DOI: 10.1111/j.1530-0277.2009.00926.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
33
|
Downing C, Balderrama-Durbin C, Broncucia H, Gilliam D, Johnson TE. Ethanol teratogenesis in five inbred strains of mice. Alcohol Clin Exp Res 2009; 33:1238-45. [PMID: 19389189 DOI: 10.1111/j.1530-0277.2009.00949.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previous studies have demonstrated individual differences in susceptibility to the detrimental effects of prenatal ethanol exposure. Many factors, including genetic differences, have been shown to play a role in susceptibility and resistance, but few studies have investigated the range of genetic variation in rodent models. METHODS We examined ethanol teratogenesis in 5 inbred strains of mice: C57BL/6J (B6), Inbred Short-Sleep, C3H/Ibg, A/Ibg, and 129S6/SvEvTac (129). Pregnant dams were intubated with either 5.8 g/kg ethanol (E) or an isocaloric amount of maltose-dextrin (MD) on day 9 of pregnancy. Dams were sacrificed on day 18 and fetuses were weighed, sexed, and examined for gross morphological malformations. Every other fetus within a litter was then either placed in Bouin's fixative for subsequent soft-tissue analyses or eviscerated and placed in ethanol for subsequent skeletal analyses. RESULTS B6 mice exposed to ethanol in utero had fetal weight deficits and digit, kidney, brain ventricle, and vertebral malformations. In contrast, 129 mice showed no teratogenesis. The remaining strains showed varying degrees of teratogenesis. CONCLUSIONS Differences among inbred strains demonstrate genetic variation in the teratogenic effects of ethanol. Identifying susceptible and resistant strains allows future studies to elucidate the genetic architecture underlying prenatal alcohol phenotypes.
Collapse
Affiliation(s)
- Chris Downing
- Institute for Behavioral Genetics, University of Colorado, Colorado, USA.
| | | | | | | | | |
Collapse
|
34
|
Summers BL, Rofe AM, Coyle P. Dietary Zinc Supplementation Throughout Pregnancy Protects Against Fetal Dysmorphology and Improves Postnatal Survival After Prenatal Ethanol Exposure in Mice. Alcohol Clin Exp Res 2009; 33:591-600. [DOI: 10.1111/j.1530-0277.2008.00873.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
35
|
Wentzel P, Eriksson UJ. Genetic influence on dysmorphogenesis in embryos from different rat strains exposed to ethanol in vivo and in vitro. Alcohol Clin Exp Res 2008; 32:874-87. [PMID: 18371156 DOI: 10.1111/j.1530-0277.2008.00647.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The aim was to investigate the susceptibility of embryos from 2 rat strains (U and H) to a 48 hours ethanol exposure in early pregnancy, both in vivo and in vitro. METHODS The embryos were studied on gestational days 9 to 11. We used 1 ethanol dose in vivo (6 g/kg x 2), 3 different ethanol concentrations in vitro (88 mM, 132 mM, 176 mM) and also attempted to diminish the teratogenic effect in vitro by supplying the antioxidant N-acetylcysteine (NAC, 0.5 mM) to the culture medium. RESULTS The U embryos were more damaged by ethanol than the H embryos, both in vivo and in vitro. NAC addition diminished, but failed to completely normalize, the embryonic maldevelopment. Ethanol increased the Bax/Bcl-2 ratio in the U embryos both in vivo and in vitro, but not in the H embryos. Furthermore, ethanol caused increased Caspase-3 immunostaining in U embryos, but not in H embryos. Ethanol exposure in vivo did not alter CuZnSOD and MnSOD mRNA levels in U and H embryos. In vitro, however, the ethanol-exposed U embryos increased their CuZnSOD and MnSOD mRNA levels, whereas the CuZnSOD mRNA was unchanged and MnSOD mRNA decreased in the H embryos, in neither strain did NAC exert any effect. The U embryos increased catalase gene expression in response to ethanol in vivo, but decreased catalase mRNA levels in vitro, changes normalized by NAC. The H embryos did not alter catalase mRNA levels in vivo, but increased gene expression in vitro, with no NAC effect. Ethanol affected the gene expression of the other ROS scavenging enzymes and the developmental genes studied - Bmp-4, Ret, Shh, Pax-6 - similarly in the 2 strains. CONCLUSIONS The findings support a role for genetic predisposition, oxidative stress, and apoptosis in ethanol teratogenicity, and suggest that the teratogenic predisposition of the more susceptible U rats may reside, at least in part, in the regulation of the ROS scavenging enzymes in the U embryos.
Collapse
Affiliation(s)
- Parri Wentzel
- Department of Medical Cell Biology, Biomedical Center, Uppsala Universitet, Uppsala, Sweden.
| | | |
Collapse
|