1
|
Yang H, Tanvir MF, Jiang H. Investigating the Impact of Chronical Prenatal Alcohol Exposure on Fetal Vascular Development Across Pregnancy Stages Using Photoacoustic Tomography. JOURNAL OF BIOPHOTONICS 2025; 18:e202400410. [PMID: 39627151 PMCID: PMC11794007 DOI: 10.1002/jbio.202400410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 02/06/2025]
Abstract
Prenatal alcohol exposure (PAE) is a major contributor to fetal alcohol spectrum disorder (FASD), resulting in neurodevelopmental abnormalities. This study utilizes photoacoustic tomography (PAT) to investigate the effects of PAE on fetal brain vasculature in mice. PAT imaging was conducted from embryonic Day 10 (E10) to Day 20 (E20), aimed to compare two alcohol-exposed groups with a control group. Key vascular parameters, including blood vessel diameter and density, and oxygen saturation (sO2), were analyzed. Results show significant reductions in vessel size and density, as well as reduced sO2 levels, in alcohol-exposed groups, especially from E14 onward, compared to controls. These findings underscore the vulnerability of the fetal brain to alcohol exposure during early development and highlight the potential of PAT as a valuable tool for investigating FASD-related vascular changes.
Collapse
Affiliation(s)
- Hao Yang
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| | - Md Farhan Tanvir
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| | - Huabei Jiang
- Department of Medical Engineering, University of South Florida, Tampa, Florida
| |
Collapse
|
2
|
Shaabani Ghahremanlo M, Hojati V, Vaezi G, Sharafi S. Transgenerational of Oxidative Damage Induced by Prenatal Ethanol Exposure on Spatial Learning/Memory and BDNF in the of Male Rats. IBRO Neurosci Rep 2024; 17:398-406. [PMID: 39583589 PMCID: PMC11583777 DOI: 10.1016/j.ibneur.2024.09.001] [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: 05/19/2024] [Accepted: 09/11/2024] [Indexed: 11/26/2024] Open
Abstract
Alcohol consumption during pregnancy harms fetal development, leading to various physical and behavioral issues. This study investigates how prenatal ethanol exposure triggers oxidative stress (OS) and affects neurotrophic factors (NTFs), particularly brain-derived growth factor (BDNF) gene expression in the hippocampus, influencing learning and memory decline across two generations of male offspring from ethanol-exposed female rats. A rat model of fetal alcohol spectrum disorder (FASD) was initially generated to reflect on the deficits in the first generation, and then those transmitted via the male germline to the unexposed male ones. The pregnant rats were thus divided into four groups, namely, the control group (CTRL) receiving only distilled water (DW), and three groups being exposed to ethanol (20 %, 4.5 g/kg) by oral gavage, during the first 10-day gestation (FG), the second 10-day gestation (SG), and the entire gestation (EG) periods. Subsequent Morris water maze (MWM) tests on male offspring revealed spatial learning deficits during the second and entire gestational periods in both generations. Analysis of antioxidant enzyme activity including glutathione peroxidase (GPx), superoxide dismutase (SOD), and malondialdehyde (MDA), and BDNF gene expression in the hippocampus further highlighted the impacts of prenatal ethanol exposure. The study results demonstrated that prenatal ethanol exposure caused spatial learning/memory deficits during the SG and EG, altered antioxidant enzyme activity, and reduced BDNF gene expression in both generations. The findings underscore the role of OS in developmental and behavioral issues in FASD rat models and suggest that lasting transgenerational effects in the second generation may stem from alcohol-induced changes.
Collapse
Affiliation(s)
| | | | - Gholamhassan Vaezi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Shahram Sharafi
- Department of Biology, Damghan Branch, Islamic Azad University, Damghan, Iran
| |
Collapse
|
3
|
Lane JM, Groth SW, Sörensen S. Longitudinal effects of prenatal substance use and environmental stressors on executive functioning in low-income African American adolescents: A latent growth modeling analysis. Brain Cogn 2024; 180:106203. [PMID: 39013291 DOI: 10.1016/j.bandc.2024.106203] [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: 10/26/2023] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Adverse prenatal substance use and environmental stressors have been linked to prefrontal cortex (PFC) impairments, the brain region that regulates executive functioning. Executive functions (e.g., inhibitory control, working memory, and cognitive flexibility) are crucial for sophisticated cognitive activities throughout child and adolescent development. There is little research on how prenatal substance use and environmental stressors longitudinally program executive functioning in children over time. We investigated the associations between prenatal/environmental stressors (i.e., maternal prenatal substance use, maternal-fetal bonding, and neighborhood disorganization) and executive function performance among low-income African American youth from age 6 until age 18. Analyses were based on four waves of data collected between 1994 and 2014 in the Memphis New Mothers Study, a longitudinal randomized controlled trial that was an intervention during pregnancy and the first two years of the child's life in low-SES women and their first-born children. Mothers and their children were followed longitudinally through 18 years post-childbirth. Prenatal substance use (e.g., prenatal smoke, alcohol, and drug use) and environmental stressor (e.g., food environment, maternal-fetal bonding and neighborhood disorganizations) evaluations were gathered from mothers and children prenatally and postnatally before the age of 4.5 years. Executive function was assessed using the Child Behavior Checklist for impulsivity and inattention, while the coding subscale of the Wechsler Intelligence Scale for Children-Third Edition, the reading recognition subtest of the Peabody Individual Achievement Test, and the digit span subtest of the Wechsler Adult Intelligence Scale were employed to assess working memory at three time periods (6, 12, and 18 years). Covariate-adjusted latent growth models estimated the associations between prenatal substance use and environmental stressors and changes in executive functioning over three time points. Prenatal smoking and alcohol use were associated with changes in impulsivity scores over 12 years. Prenatal alcohol use predicted higher inattention at baseline and a slower rate of change from ages 6 to 18. Neighborhood disorganization at ages 6 and 18 predicted higher inattention and lower working memory in youth at age 18, respectively. Our findings underscore the long-term impact of prenatal substance use exposures and neighborhood environments on cognitive development and highlight the importance of early interventions to mitigate these effects.
Collapse
Affiliation(s)
- Jamil M Lane
- Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Susan W Groth
- School of Nursing, University of Rochester, Rochester, NY, USA
| | - Silvia Sörensen
- Department of Counseling and Human Development, University of Rochester, Rochester, NY, USA
| |
Collapse
|
4
|
Burmistrov DE, Gudkov SV, Franceschi C, Vedunova MV. Sex as a Determinant of Age-Related Changes in the Brain. Int J Mol Sci 2024; 25:7122. [PMID: 39000227 PMCID: PMC11241365 DOI: 10.3390/ijms25137122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.
Collapse
Affiliation(s)
- Dmitriy E. Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
5
|
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
|
6
|
Darbinian N, Merabova N, Tatevosian G, Morrison M, Darbinyan A, Zhao H, Goetzl L, Selzer ME. Biomarkers of Affective Dysregulation Associated with In Utero Exposure to EtOH. Cells 2023; 13:2. [PMID: 38201206 PMCID: PMC10778368 DOI: 10.3390/cells13010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 12/13/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
INTRODUCTION Children with fetal alcohol spectrum disorders (FASD) exhibit behavioral and affective dysregulation, including hyperactivity and depression. The mechanisms are not known, but they could conceivably be due to postnatal social or environmental factors. However, we postulate that, more likely, the affective dysregulation is associated with the effects of EtOH exposure on the development of fetal serotonergic (5-HT) and/or dopaminergic (DA) pathways, i.e., pathways that in postnatal life are believed to regulate mood. Many women who use alcohol (ethanol, EtOH) during pregnancy suffer from depression and take selective serotonin reuptake inhibitors (SSRIs), which might influence these monoaminergic pathways in the fetus. Alternatively, monoaminergic pathway abnormalities might reflect a direct effect of EtOH on the fetal brain. To distinguish between these possibilities, we measured their expressions in fetal brains and in fetal brain-derived exosomes (FB-Es) isolated from the mothers' blood. We hypothesized that maternal use of EtOH and/or SSRIs during pregnancy would be associated with impaired fetal neural development, detectable as abnormal levels of monoaminergic and apoptotic biomarkers in FB-Es. METHODS Fetal brain tissues and maternal blood were collected at 9-23 weeks of pregnancy. EtOH groups were compared with unexposed controls matched for gestational age (GA). The expression of 84 genes associated with the DA and 5-HT pathways was analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) on microarrays. FB-Es also were assayed for serotonin transporter protein (SERT) and brain-derived neurotrophic factor (BDNF) by enzyme-linked immunosorbent assay (ELISA). RESULTS Six EtOH-exposed human fetal brain samples were compared to SSRI- or polydrug-exposed samples and to unexposed controls. EtOH exposure was associated with significant upregulation of DA receptor D3 and 5-HT receptor HTR2C, while HTR3A was downregulated. Monoamine oxidase A (MAOA), MAOB, the serine/threonine kinase AKT3, and caspase-3 were upregulated, while mitogen-activated protein kinase 1 (MAPK1) and AKT2 were downregulated. ETOH was associated with significant upregulation of the DA transporter gene, while SERT was downregulated. There were significant correlations between EtOH exposure and (a) caspase-3 activation, (b) reduced SERT protein levels, and (c) reduced BDNF levels. SSRI exposure independently increased caspase-3 activity and downregulated SERT and BDNF. Early exposure to EtOH and SSRI together was associated synergistically with a significant upregulation of caspase-3 and a significant downregulation of SERT and BDNF. Reduced SERT and BDNF levels were strongly correlated with a reduction in eye diameter, a somatic manifestation of FASD. CONCLUSIONS Maternal use of EtOH and SSRI during pregnancy each was associated with changes in fetal brain monoamine pathways, consistent with potential mechanisms for the affective dysregulation associated with FASD.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
- Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
| | - Mary Morrison
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
- Department of Psychiatry, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Huaqing Zhao
- Center for Biostatistics and Epidemiology, Department of Biomedical Education and Data Science, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA;
| | - Michael Edgar Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (G.T.)
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
7
|
Gerace E, Curti L, Caffino L, Bigagli E, Mottarlini F, Castillo Díaz F, Ilari A, Luceri C, Dani C, Fumagalli F, Masi A, Mannaioni G. Ethanol-induced AMPA alterations are mediated by mGLU5 receptors through miRNA upregulation in hippocampal slices. Eur J Pharmacol 2023; 955:175878. [PMID: 37433363 DOI: 10.1016/j.ejphar.2023.175878] [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: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/13/2023]
Abstract
Prenatal alcohol exposure (PAE) affects neuronal networks and brain development causing a range of physical, cognitive and behavioural disorders in newborns that persist into adulthood. The array of consequences associated with PAE can be grouped under the umbrella-term 'fetal alcohol spectrum disorders' (FASD). Unfortunately, there is no cure for FASD as the molecular mechanisms underlying this pathology are still unknown. We have recently demonstrated that chronic EtOH exposure, followed by withdrawal, induces a significant decrease in AMPA receptor (AMPAR) expression and function in developing hippocampus in vitro. Here, we explored the EtOH-dependent pathways leading to hippocampal AMPAR suppression. Organotypic hippocampal slices (2 days in cultures) were exposed to EtOH (150 mM) for 7 days followed by 24 h EtOH withdrawal. Then, the slices were analysed by means of RT-PCR for miRNA content, western blotting for AMPA and NMDA related-synaptic proteins expression in postsynaptic compartment and electrophysiology to record electrical properties from CA1 pyramidal neurons. We observed that EtOH induces a significant downregulation of postsynaptic AMPA and NMDA subunits and relative scaffolding protein expression and, accordingly, a decrease of AMPA-mediated neurotransmission. Simultaneously, we found that chronic EtOH induced-upregulation of miRNA 137 and 501-3p and decreased AMPA-mediated neurotransmission are prevented by application of the selective mGlu5 antagonist MPEP during EtOH withdrawal. Our data indicate mGlu5 via miRNA137 and 501-3p expression as key factors in the regulation of AMPAergic neurotransmission that may contribute, at least in part, to the pathogenesis of FASD.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; Department of Health Sciences (DSS), University of Florence, Florence, Italy.
| | - Lorenzo Curti
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Italy
| | - Elisabetta Bigagli
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Italy
| | - Fernando Castillo Díaz
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Italy
| | - Alice Ilari
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Cristina Luceri
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Carlo Dani
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy; Division of Neonatology, Careggi University Hospital of Florence, Florence, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Italy
| | - Alessio Masi
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Guido Mannaioni
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
8
|
Mendes PFS, Baia-da-Silva DC, Melo WWP, Bittencourt LO, Souza-Rodrigues RD, Fernandes LMP, Maia CDSF, Lima RR. Neurotoxicology of alcohol: a bibliometric and science mapping analysis. Front Pharmacol 2023; 14:1209616. [PMID: 37593178 PMCID: PMC10427875 DOI: 10.3389/fphar.2023.1209616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/11/2023] [Indexed: 08/19/2023] Open
Abstract
Alcohol consumption is common in many societies and has increased considerably, resulting in many socioeconomic and public health problems. In this sense, studies have been carried out in order to understand the mechanisms involved in alcohol consumption and related harmful effects. This study aimed to identify and map the knowledge and to perform bibliometric analysis of the neurotoxicology of alcohol based on the 100 most cited articles. A search was carried out in the Web of Science Core Collection database and information was extracted regarding the journal, authors, keywords, year of publication, number of citations, country and continent of the corresponding author. For each selected manuscript, the study design, alcohol exposure model, dose, period of exposure, and effect on the central nervous system and research hotspots were mapped. The journal with the highest number of publications was Alcoholism: Clinical and Experimental Research (n = 11 papers), the author who contributed the most was Crews FT (n = 8 papers), the studies had a total of 288 keywords and 75% of the publications were from the United States of America. The experimental studies evaluated the effects of prenatal and postnatal exposure and were conducted in rats and mice using doses ranging from 2.5 to 14 g/kg/day, with administration by subcutaneous, intraperitoneal, intragastric, or inhalation route or with free access through drinking bottles. Among the studies mapped, the oldest one (1989) aimed to understand the systemic damage and mechanisms of action involved, while the most recent focused on understanding the receptors and mechanisms involved in addiction, as well as genetic factors. Our results show the panorama of the most widespread scientific production in the scientific community on the neurotoxicology of ethanol, a high prevalence was observed in studies that addressed fetal alcohol syndrome and/or the effects of ethanol on neurodevelopment.
Collapse
Affiliation(s)
- Paulo Fernando Santos Mendes
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Daiane Claydes Baia-da-Silva
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Wallacy Watson Pereira Melo
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Renata Duarte Souza-Rodrigues
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Luanna Melo Pereira Fernandes
- Department of Morphology and Physiological Sciences, Center of Sciences Biological and Health, State University of Pará, Belém, Brazil
| | | | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
9
|
Baker JA, Mulligan MK, Hamre KM. Effects of genetics and sex on adolescent behaviors following neonatal ethanol exposure in BXD recombinant inbred strains. Front Neurosci 2023; 17:1197292. [PMID: 37564365 PMCID: PMC10410115 DOI: 10.3389/fnins.2023.1197292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction Fetal alcohol spectrum disorders (FASD) are the leading preventable neurodevelopmental disorders and two hallmark symptoms of FASD are abnormal behavior, and cognitive and learning deficits. The severity of alcohol's teratogenic effects on the developing brain is influenced by genetics and sex. We previously identified recombinant inbred BXD mouse strains that show differential vulnerability to ethanol-induced cell death in the developing hippocampus, a brain region important in learning and memory. The present study aimed to test the hypothesis that strains with increased vulnerability to ethanol-induced cell death in the hippocampus have concomitant deficits in multiple hippocampal-related behaviors during adolescence. Methods The current study evaluated the effects of developmental ethanol exposure on adolescent behavior in two BXD strains that show high cell death (BXD48a, BXD100), two that show low cell death (BXD60, BXD71), and the two parental strains (C57BL/6 J (B6), DBA/2 J (D2)). On postnatal day 7, male and female neonatal pups were treated with ethanol (5.0 g/kg) or saline given in two equal doses 2 h apart. Adolescent behavior was assessed across multiple behavioral paradigms including the elevated plus maze, open field, Y-maze, and T-maze. Results Our results demonstrate that the effects of developmental ethanol exposure on adolescent behavioral responses are highly dependent on strain. The low cell death strains, BXD60 and BXD71, showed minimal effect of ethanol exposure on all behavioral measures but did present sex differences. The parental -B6 and D2-strains and high cell death strains, BXD48a and BXD100, showed ethanol-induced effects on activity-related or anxiety-like behaviors. Interestingly, the high cell death strains were the only strains that showed a significant effect of postnatal ethanol exposure on hippocampal-dependent spatial learning and memory behaviors. Discussion Overall, we identified effects of ethanol exposure, strain, and/or sex on multiple behavioral measures. Interestingly, the strains that showed the most effects of postnatal ethanol exposure on adolescent behavior were the BXD strains that show high ethanol-induced cell death in the neonatal hippocampus, consistent with our hypothesis. Additionally, we found evidence for interactions among strain and sex, demonstrating that these factors have a complex effect on alcohol responses and that both are important considerations.
Collapse
Affiliation(s)
- Jessica A. Baker
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States
- Center for Behavioral Teratology, San Diego State University, San Diego, CA, United States
| | - Megan K. Mulligan
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Kristin M. Hamre
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
10
|
Wolfe M, Menon A, Oto M, Fullerton NE, Leach JP. Alcohol and the central nervous system. Pract Neurol 2023:pn-2023-003817. [PMID: 37328277 DOI: 10.1136/pn-2023-003817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2023] [Indexed: 06/18/2023]
Abstract
Ethanol use is common to most cultures but with varying doses and to varying extents. While research has focused on the effects on the liver, alcohol exerts a range of actions on the function and structure of the nervous system. In the central nervous system (CNS) it can provoke or exacerbate neurological and psychiatric disease; its effects on the peripheral nervous system are not included in this review. Sustained alcohol intake can predispose to acute neurochemical changes which, with continued ingestion and incomplete treatment, can lead to chronic structural changes in the CNS: these include generalised cortical and cerebellar atrophy, amnesic syndromes such as Korsakoff's syndrome, and specific white matter disorders such as central pontine myelinolysis and Marchiafava-Bignami syndrome. Alcohol in pregnancy commonly and significantly affects fetal health, though this receives less medical and political attention than other causes of fetal harm. This review looks at the range of disorders that can follow acute or chronic alcohol use, and how these should be managed, and we provide a practical overview on how neurologists might diagnose and manage alcohol addiction.
Collapse
Affiliation(s)
- Maytal Wolfe
- University of Glasgow, Glasgow, UK
- Queen Elizabeth University Hospital, Glasgow, UK
| | - Arun Menon
- University of Glasgow, Glasgow, UK
- Gartnavel Royal Hospital, Glasgow, UK
| | - Maria Oto
- Queen Elizabeth University Hospital, Glasgow, UK
| | - Natasha E Fullerton
- University of Glasgow, Glasgow, UK
- Queen Elizabeth University Hospital, Glasgow, UK
| | | |
Collapse
|
11
|
Bariselli S, Reuveni N, Westcott N, Mateo Y, Lovinger DM. Postnatal ethanol exposure impairs social behavior and operant extinction in the adult female mouse offspring. Front Neurosci 2023; 17:1160185. [PMID: 37260840 PMCID: PMC10229070 DOI: 10.3389/fnins.2023.1160185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/26/2023] [Indexed: 06/02/2023] Open
Abstract
Fetal Alcohol Spectrum Disorder (FASD) comprises a group of neurodevelopmental deficits caused by alcohol exposure during pregnancy. Clinical studies suggest that while the male progeny experiences serious neurodevelopmental defects, female patients have more severe cognitive, social, and affective symptoms. Other than sex, dose, frequency, and timing of exposure determine the neurobehavioral outcomes in young and adult progeny. In this regard, human studies indicate that some individuals relapse during late-term gestational periods. In mice, this interval corresponds to the first 10 days after birth (postnatal, P0-P10). In our model of postnatal ethanol exposure (PEEP0-P10), we tested whether adult female and male offspring show deficits in sociability, anxiety-like, reward consumption, and action-outcome associations. We report that female PEEP0-P10 offspring have mild social impairments and altered extinction of operant responding in the absence of anxiety-like traits and reward consumption defects. None of these deficits were detected in the male PEEP0-P10 offspring. Our data provide novel information on sex-specific neurobehavioral outcomes of postnatal ethanol exposure in female adult offspring.
Collapse
|
12
|
Weinmann T, Finkeldey L, Wittmann E, Kerber K, Schlueter JA, Zillinger I, Maciejok A, Heinen F, Landgraf MN. Prognostic factors for long-term outcome in children with fetal alcohol spectrum disorders. RESEARCH IN DEVELOPMENTAL DISABILITIES 2023; 136:104481. [PMID: 36924617 DOI: 10.1016/j.ridd.2023.104481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 02/22/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Known protective factors for long-term outcome in children with fetal alcohol spectrum disorders (FASD) are early diagnosis and a stable, non-violent supportive environment. Which factors contribute to the stability of care is not yet known. Thus, the aim of our study was to evaluate whether the age at diagnosis and the complexity of brain dysfunction play a role for placement changes in children with FASD. MATERIALS AND METHODS An online survey was conducted among caregivers and professionals caring for children with FASD and seeking help at the German FASD Competence Centre Bavaria (N = 232). The survey collected information about diagnosis, brain dysfunctions, behavioural factors influencing everyday life and changes of placement. The association of timing of diagnosis, brain dysfunctions and neurobehavioral impairment with changes of placement (<2 vs. 2 or more changes) was evaluated via logistic regression models. RESULTS About 50% of the children received their diagnosis of FASD after the age of 5 years. The complexity of brain dysfunctions in children with FASD affecting everyday life was high. 15% of the children experienced four or more changes of placement. Children with more neuropsychological impairments experienced more changes of placement (OR: 2.53, 95% CI: 1.36-4.71). CONCLUSIONS Even though our results need to be interpreted with caution due to methodological limitations such as the use of a convenience sample and limited statistical power, they imply that severely affected children with FASD experience a less stable environment. These children may therefore be at high risk for a negative prognosis. To warrant a better prognosis for the affected children, professionals urgently need to pay attention to early recognition and the complexity of neuropsychological impairments in children with FASD as well as to the support that caregivers urgently need.
Collapse
Affiliation(s)
- Tobias Weinmann
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Finkeldey
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Esther Wittmann
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Katharina Kerber
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Julia A Schlueter
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Iris Zillinger
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Anja Maciejok
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Florian Heinen
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany
| | - Mirjam N Landgraf
- Department of Paediatric Neurology and Developmental Medicine, German FASD Competence Centre Bavaria, Dr. von Hauner Children's Hospital, LMU Hospital Munich, Ludwig-Maximilians-Universität, Lindwurmstr. 5, 80337 Munich, Germany.
| |
Collapse
|
13
|
Basavarajappa BS, Subbanna S. Synaptic Plasticity Abnormalities in Fetal Alcohol Spectrum Disorders. Cells 2023; 12:442. [PMID: 36766783 PMCID: PMC9913617 DOI: 10.3390/cells12030442] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The brain's ability to strengthen or weaken synaptic connections is often termed synaptic plasticity. It has been shown to function in brain remodeling following different types of brain damage (e.g., drugs of abuse, alcohol use disorders, neurodegenerative diseases, and inflammatory conditions). Although synaptic plasticity mechanisms have been extensively studied, how neural plasticity can influence neurobehavioral abnormalities in alcohol use disorders (AUDs) is far from being completely understood. Alcohol use during pregnancy and its harmful effects on the developing offspring are major public health, social, and economic challenges. The significant attribute of prenatal alcohol exposure on offspring is damage to the central nervous system (CNS), causing a range of synaptic structural, functional, and behavioral impairments, collectively called fetal alcohol spectrum disorder (FASD). Although the synaptic mechanisms in FASD are limited, emerging evidence suggests that FASD pathogenesis involves altering a set of molecules involved in neurotransmission, myelination, and neuroinflammation. These studies identify several immediate and long-lasting changes using many molecular approaches that are essential for synaptic plasticity and cognitive function. Therefore, they can offer potential synaptic targets for the many neurobehavioral abnormalities observed in FASD. In this review, we discuss the substantial research progress in different aspects of synaptic and molecular changes that can shed light on the mechanism of synaptic dysfunction in FASD. Increasing our understanding of the synaptic changes in FASD will significantly advance our knowledge and could provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
Affiliation(s)
- Balapal S. Basavarajappa
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shivakumar Subbanna
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| |
Collapse
|
14
|
Saha PS, Knecht TM, Arrick DM, Watt MJ, Scholl JL, Mayhan WG. Prenatal exposure to alcohol impairs responses of cerebral arterioles to activation of potassium channels: Role of oxidative stress. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:87-94. [PMID: 36446735 PMCID: PMC9974881 DOI: 10.1111/acer.14980] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/24/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Potassium channels play an important role in the basal tone and dilation of cerebral resistance arterioles in response to many stimuli. However, the effect of prenatal alcohol exposure (PAE) on specific potassium channel function remains unknown. The first goal of this study was to determine the influence of PAE on the reactivity of cerebral arterioles to activation of ATP-sensitive potassium (KATP ) and BK channels. Our second goal was to determine whether oxidative stress contributed to potassium channel dysfunction of cerebral arterioles following PAE. METHODS We fed Sprague-Dawley dams a liquid diet with or without alcohol (3% EtOH) for the duration of their pregnancy (21 to 23 days). We examined in vivo responses of cerebral arterioles in control and PAE male and female offspring (14 to 16 weeks after birth) to activators of potassium channels (Iloprost [BK channels] and pinacidil [KATP channels]), before and following inhibition of oxidative stress with apocynin. RESULTS We found that PAE impaired dilation of cerebral arterioles in response to activation of potassium channels with iloprost and pinacidil, and this impairment was similar in male and female rats. In addition, treatment with apocynin reversed the impaired vasodilation to iloprost and pinacidil in PAE rats to levels observed in control rats. This effect of apocynin also was similar in male and female rats. CONCLUSIONS PAE induces dysfunction in the ability of specific potassium channels to dilate cerebral arterioles which appears to be mediated by an increase in oxidative stress. We suggest that these alterations in potassium channel function may contribute to the pathogenesis of cerebral vascular abnormalities and/or behavioral/cognitive deficits observed in fetal alcohol spectrum disorders.
Collapse
Affiliation(s)
- Partha S. Saha
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| | - Tiffany M. Knecht
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| | - Denise M. Arrick
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| | - Michael J. Watt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jamie L. Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| | - William G. Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| |
Collapse
|
15
|
Epigenetics in fetal alcohol spectrum disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:211-239. [PMID: 37019593 DOI: 10.1016/bs.pmbts.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
During pregnancy, alcohol abuse and its detrimental effects on developing offspring are major public health, economic and social challenges. The prominent characteristic attributes of alcohol (ethanol) abuse during pregnancy in humans are neurobehavioral impairments in offspring due to damage to the central nervous system (CNS), causing structural and behavioral impairments that are together named fetal alcohol spectrum disorder (FASD). Development-specific alcohol exposure paradigms were established to recapitulate the human FASD phenotypes and establish the underlying mechanisms. These animal studies have offered some critical molecular and cellular underpinnings likely to account for the neurobehavioral impairments associated with prenatal ethanol exposure. Although the pathogenesis of FASD remains unclear, emerging literature proposes that the various genomic and epigenetic components that cause the imbalance in gene expression can significantly contribute to the development of this disease. These studies acknowledged numerous immediate and enduring epigenetic modifications, such as methylation of DNA, post-translational modifications (PTMs) of histone proteins, and regulatory networks related to RNA, using many molecular approaches. Methylated DNA profiles, PTMs of histone proteins, and RNA-regulated expression of genes are essential for synaptic and cognitive behavior. Thus, offering a solution to many neuronal and behavioral impairments reported in FASD. In the current chapter, we review the recent advances in different epigenetic modifications that cause the pathogenesis of FASD. The information discussed can help better explain the pathogenesis of FASD and thereby might provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
|
16
|
Darbinian N, Darbinyan A, Sinard J, Tatevosian G, Merabova N, D’Amico F, Khader T, Bajwa A, Martirosyan D, Gawlinski AK, Pursnani R, Zhao H, Amini S, Morrison M, Goetzl L, Selzer ME. Molecular Markers in Maternal Blood Exosomes Allow Early Detection of Fetal Alcohol Spectrum Disorders. Int J Mol Sci 2022; 24:ijms24010135. [PMID: 36613580 PMCID: PMC9820501 DOI: 10.3390/ijms24010135] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Prenatal alcohol exposure can cause developmental abnormalities (fetal alcohol spectrum disorders; FASD), including small eyes, face and brain, and neurobehavioral deficits. These cannot be detected early in pregnancy with available imaging techniques. Early diagnosis could facilitate development of therapeutic interventions. Banked human fetal brains and eyes at 9−22 weeks’ gestation were paired with maternal blood samples, analyzed for morphometry, protein, and RNA expression, and apoptotic signaling. Alcohol (EtOH)-exposed (maternal self-report) fetuses were compared with unexposed controls matched for fetal age, sex, and maternal race. Fetal brain-derived exosomes (FB-E) were isolated from maternal blood and analyzed for protein, RNA, and apoptotic markers. EtOH use by mothers, assessed by self-report, was associated with reduced fetal eye diameter, brain size, and markers of synaptogenesis. Brain caspase-3 activity was increased. The reduction in eye and brain sizes were highly correlated with amount of EtOH intake and caspase-3 activity. Levels of several biomarkers in FB-E, most strikingly myelin basic protein (MBP; r > 0.9), correlated highly with morphological abnormalities. Reduction in FB-E MBP levels was highly correlated with EtOH exposure (p < 1.0 × 10−10). Although the morphological features of FAS appear long before they can be detected by live imaging, FB-E in the mother’s blood may contain markers, particularly MBP, that predict FASD.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Correspondence: (N.D.); (M.E.S.); Tel.: +1-215-926-9318 (M.E.S.)
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - John Sinard
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Medical College of Wisconsin-Prevea Health, Green Bay, WI 54304, USA
| | - Faith D’Amico
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tarek Khader
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ahsun Bajwa
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Diana Martirosyan
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Alina K. Gawlinski
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Richa Pursnani
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Huaqing Zhao
- Center for Biostatistics and Epidemiology, Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Shohreh Amini
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Mary Morrison
- Department of Psychiatry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, USA
| | - Michael E. Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- Correspondence: (N.D.); (M.E.S.); Tel.: +1-215-926-9318 (M.E.S.)
| |
Collapse
|
17
|
Saha PS, Mayhan WG. Prenatal exposure to alcohol: mechanisms of cerebral vascular damage and lifelong consequences. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10818. [PMID: 38390614 PMCID: PMC10880760 DOI: 10.3389/adar.2022.10818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/01/2022] [Indexed: 02/24/2024]
Abstract
Alcohol is a well-known teratogen, and prenatal alcohol exposure (PAE) leads to a greater incidence of many cardiovascular-related pathologies. Alcohol negatively impacts vasculogenesis and angiogenesis in the developing fetal brain, resulting in fetal alcohol spectrum disorders (FASD). Ample preclinical evidence indicates that the normal reactivity of cerebral resistance arterioles, which regulate blood flow distribution in response to metabolic demand (neurovascular coupling), is impaired by PAE. This impairment of dilation of cerebral arteries may carry implications for the susceptibility of the brain to cerebral ischemic damage well into adulthood. The focus of this review is to consolidate findings from studies examining the influence of PAE on vascular development, give insights into relevant pathological mechanisms at the vascular level, evaluate the risks of ethanol-driven alterations of cerebrovascular reactivity, and revisit different preventive interventions that may have promise in reversing vascular changes in preclinical FASD models.
Collapse
Affiliation(s)
- Partha S Saha
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
18
|
Tsang TW, Finlay-Jones A, Perry K, Grigg JR, Popova S, Cheung MMY, Bower C, Tam P, Jamieson RV, Elliott EJ. Eye Abnormalities in Children with Fetal Alcohol Spectrum Disorders: A Systematic Review. Ophthalmic Epidemiol 2022:1-12. [DOI: 10.1080/09286586.2022.2123004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Tracey W Tsang
- The University of Sydney Children’s Hospital Westmead Clinical School, Discipline of Child and Adolescent Health, Faculty of Medicine and Health, Westmead, NSW, Australia, and Sydney Children’s Hospital Network, Kids Research, Westmead, Australia
| | - Amy Finlay-Jones
- Alcohol & Pregnancy and FASD Research, Telethon Kids Institute, West Perth, Australia
- Curtin University, West Perth, Australia
| | - Kerrin Perry
- The University of Sydney Children’s Hospital Westmead Clinical School, Discipline of Child and Adolescent Health, Faculty of Medicine and Health, Westmead, NSW, Australia, and Sydney Children’s Hospital Network, Kids Research, Westmead, Australia
| | - John R Grigg
- Save Sight Institute, Faculty of Medicine and Health, the University of Sydney, Sydney, Australia
- Sydney Eye Hospital, Sydney, Australia
| | - Svetlana Popova
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Canada; Dalla Lana School of Public Health, University of Toronto, Canada; Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, ON, Canada
| | - Melissa Mei Yin Cheung
- The University of Sydney Children’s Hospital Westmead Clinical School, Discipline of Child and Adolescent Health, Faculty of Medicine and Health, Westmead, NSW, Australia, and Sydney Children’s Hospital Network, Kids Research, Westmead, Australia
- Australian Paediatric Surveillance Unit, Kids Research, Westmead, Australia
| | - Carol Bower
- Alcohol & Pregnancy and FASD Research, Telethon Kids Institute, West Perth, Australia
| | - Patrick Tam
- Embryology Research Unit, Children’s Medical Research Institute, the University of Sydney, Westmead, NSW, Australia; and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Robyn V Jamieson
- Sydney Eye Hospital, Sydney, Australia
- Department of Clinical Genetics, Western Sydney Genetics Program, Sydney Children’s Hospitals Network, Westmead, NSW, Australia, and Eye Genetics Research Unit, Children’s Medical Research Institute, Sydney Children’s Hospitals Network, Westmead, Australia
| | - Elizabeth J Elliott
- The University of Sydney Children’s Hospital Westmead Clinical School, Discipline of Child and Adolescent Health, Faculty of Medicine and Health, Westmead, NSW, Australia, and Sydney Children’s Hospital Network, Kids Research, Westmead, Australia
- Australian Paediatric Surveillance Unit, Kids Research, Westmead, Australia
| |
Collapse
|
19
|
Sex-Related Differences in Voluntary Alcohol Intake and mRNA Coding for Synucleins in the Brain of Adult Rats Prenatally Exposed to Alcohol. Biomedicines 2022; 10:biomedicines10092163. [PMID: 36140264 PMCID: PMC9496239 DOI: 10.3390/biomedicines10092163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Maternal alcohol consumption is one of the strong predictive factors of alcohol use and consequent abuse; however, investigations of sex differences in response to prenatal alcohol exposure (PAE) are limited. Here we compared the effects of PAE throughout gestation on alcohol preference, state anxiety and mRNA expression of presynaptic proteins α-, β- and γ-synucleins in the brain of adult (PND60) male and female Wistar rats. Total RNA was isolated from the hippocampus, midbrain and hypothalamus and mRNA levels were assessed with quantitative RT-PCR. Compared with naïve males, naïve female rats consumed more alcohol in “free choice” paradigm (10% ethanol vs. water). At the same time, PAE produced significant increase in alcohol consumption and preference in males but not in females compared to male and female naïve groups, correspondingly. We found significantly lower α-synuclein mRNA levels in the hippocampus and midbrain of females compared to males and significant decrease in α-synuclein mRNA in these brain areas in PAE males, but not in females compared to the same sex controls. These findings indicate that the impact of PAE on transcriptional regulation of synucleins may be sex-dependent, and in males’ disruption in α-synuclein mRNA expression may contribute to increased vulnerability to alcohol-associated behavior.
Collapse
|
20
|
Martín-Estal I, Fajardo-Ramírez ÓR, Bermúdez de León M, Zertuche-Mery C, Benavides-Guajardo R, García-Cruz MI, Rodríguez De Ita J, Castilla-Cortázar I, Castorena-Torres F. Effect of Ethanol Consumption on the Placenta and Liver of Partially IGF-1-Deficient Mice: The Role of Metabolism via CYP2E1 and the Antioxidant Enzyme System. BIOLOGY 2022; 11:biology11091264. [PMID: 36138743 PMCID: PMC9495332 DOI: 10.3390/biology11091264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Ethanol is the most consumed drug worldwide, even during pregnancy. One of its adverse outcomes is fetal growth restriction, an alteration in development due to decreased IGF-1 levels. Several studies have shown that ethanol can impair the IGF-1 signaling pathway, thus exacerbating IGF-1 adverse effects in both intrauterine and postnatal growth and development. In this manuscript, we used a partially IGF-1-deficient mouse model to demonstrate the key role of IGF-1 in fetal development, as well as ethanol’s adverse effects on CYP2E1 expression levels and the antioxidant enzyme system during pregnancy. Abstract Ethanol use during pregnancy is a risk factor for developing adverse outcomes. Its metabolism by cytochrome P450 2E1 (CYP2E1) produces radical oxygen species (ROS), promoting cellular injury and apoptosis. To date, no studies have been conducted to elucidate the teratogenic effects due to both IGF-1 deficiency and ethanol consumption in mice placentas. The aim of this study is to determine the effect of ethanol consumption on the placenta and liver of partially IGF-1-deficient mice, the role of metabolism via CYP2E1, and the antioxidant enzyme system. Heterozygous (HZ, Igf1+/−) pregnant female mice were given water or 10% ethanol. Wild-type (WT, Igf1+/+) female mice were used as controls. At gestational day 19, pregnant dams were euthanized, and maternal liver and placentas were collected. Pregnant HZ dams were smaller than controls, and this effect was higher due to ethanol consumption. Cyp2e1 gene was overexpressed in the liver of HZ pregnant dams exposed to ethanol; at the protein level, CYP2E1 is reduced in placentas from all genotypes. The antioxidant enzymatic system was altered by ethanol consumption in both the maternal liver and placenta. The results in this work hint that IGF-1 is involved in intrauterine development because its deficiency exacerbates ethanol’s effects on both metabolism and the placenta.
Collapse
Affiliation(s)
- Irene Martín-Estal
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Óscar R. Fajardo-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Mario Bermúdez de León
- Departamento de Biología Molecular, Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Monterrey 64720, N.L., Mexico
| | - Carolina Zertuche-Mery
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Rodolfo Benavides-Guajardo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - María Isabel García-Cruz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Julieta Rodríguez De Ita
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
| | - Inma Castilla-Cortázar
- Fundación de Investigación HM Hospitales, 28015 Madrid, Spain
- Correspondence: (I.C.-C.); (F.C.-T.)
| | - Fabiola Castorena-Torres
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey 64710, N.L., Mexico
- Correspondence: (I.C.-C.); (F.C.-T.)
| |
Collapse
|
21
|
Prenatal maternal alcohol exposure: Diagnosis and prevention of fetal alcohol syndrome. Obstet Gynecol Sci 2022; 65:385-394. [PMID: 35908651 PMCID: PMC9483667 DOI: 10.5468/ogs.22123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/17/2022] [Indexed: 11/30/2022] Open
Abstract
Fetal alcohol syndrome (FAS) is a developmental and congenital disorder characterized by neurocognitive impairment, structural defects, and growth restriction due to prenatal alcohol exposure. The estimated global prevalence of alcohol use during pregnancy is 9.8%, and the estimated prevalence of FAS in the general population is 14.6 per 10,000 people. In Korea, the estimated prevalence of alcohol use during pregnancy is 16%, and the prevalence of FAS is 18–51 per 10,000 women, which is higher than the global prevalence. Women’s alcohol consumption rates have increased, especially in women of childbearing age. This could increase the incidence of FAS, leading to higher medical expenses and burden on society. Alcohol is the single most important teratogen that causes FAS, and there is no safe trimester to drink alcohol and no known safe amount of alcohol consumption during pregnancy. Thus, physicians should assess women’s drinking patterns in detail and provide education on FAS to women by understanding its pathophysiology. Moreover, the prevention of FAS requires long-term care with a multidisciplinary approach.
Collapse
|
22
|
Liu X, Vigorito M, Huang W, Khan MAS, Chang SL. The Impact of Alcohol-Induced Dysbiosis on Diseases and Disorders of the Central Nervous System. J Neuroimmune Pharmacol 2022; 17:131-151. [PMID: 34843074 DOI: 10.1007/s11481-021-10033-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022]
Abstract
The human digestive tract contains a diverse and abundant microbiota that is important for health. Excessive alcohol use can disrupt the balance of these microbes (known as dysbiosis), leading to elevated blood endotoxin levels and systemic inflammation. Using QIAGEN Ingenuity Pathway Analysis (IPA) bioinformatics tool, we have confirmed that peripheral endotoxin (lipopolysaccharide) mediates various cytokines to enhance the neuroinflammation signaling pathway. The literature has identified alcohol-mediated neuroinflammation as a possible risk factor for the onset and progression of neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), and psychiatric disorders such as addiction to alcohol and other drugs. In this review, we discuss alcohol-use-induced dysbiosis in the gut and other body parts as a causal factor in the progression of Central Nervous System (CNS) diseases including neurodegenerative disease and possibly alcohol use disorder.
Collapse
Affiliation(s)
- Xiangqian Liu
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Michael Vigorito
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Psychology, Seton Hall University, South Orange, NJ, 07079, USA
| | - Wenfei Huang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA
| | - Mohammed A S Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, Boston, MA, 02114, USA.
| | - Sulie L Chang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA.
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA.
| |
Collapse
|
23
|
Carugati M, Goodlett CR, Cudd TA, Washburn SE. The effects of gestational choline supplementation on cerebellar Purkinje cell number in the sheep model of binge alcohol exposure during the first trimester-equivalent. Alcohol 2022; 100:11-21. [PMID: 35114358 PMCID: PMC8983574 DOI: 10.1016/j.alcohol.2022.01.002] [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: 09/15/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 11/01/2022]
Abstract
Individuals with fetal alcohol spectrum disorders (FASD) incur enduring brain damage and neurodevelopmental impairments from prenatal alcohol exposure (PAE). Preclinical rodent models have demonstrated that choline supplementation during development can reduce the severity of adverse neurodevelopmental consequences of PAE. This study used the sheep model to evaluate dietary choline supplementation during pregnancy as a therapeutic intervention, testing the hypothesis that choline can ameliorate alcohol-induced cerebellar Purkinje cell loss. Pregnant ewes were randomly assigned either to a normal control [NC] group (n = 8), or to groups given intravenous infusions of alcohol (or saline) from gestational days 4-41 (the first trimester-equivalent). A weekly binge-drinking pattern was modeled, with three consecutive days of infusions of saline [SAL], 1.75 g/kg/day alcohol [1.75ALC], or 2.5 g/kg/day alcohol [2.5ALC] followed by four days off. Infused ewes were randomly assigned to receive dietary supplements throughout pregnancy of choline (10 mg/kg/day) or placebo (n = 8 per group). Mean blood alcohol concentrations (BAC) were significantly higher in the 2.5ALC groups (287 mg/dL) than the 1.75ALC groups (197 mg/dL). Lamb cerebella were harvested on postnatal day 180 and processed for stereological counts of Purkinje cells. Both alcohol doses caused significant reductions in Purkinje number relative to NC and SAL-Placebo groups, confirming previous findings. Effects of choline supplementation depended on infusion group: it significantly protected against Purkinje cell loss in the 2.5ALC group, had no effect in the 1.75ALC group, and significantly reduced numbers in the SAL-Choline group (though neither the SAL-Choline nor the SAL-Placebo group differed from the NC group). The protection by choline evident only in the 2.5ALC group suggests that multiple, BAC-dependent mechanisms of cerebellar damage may be activated with alcohol exposure in the first trimester, and that choline may protect against pathogenic mechanisms that emerge at higher BACs. These outcomes extend the evidence that early choline supplementation can mitigate some neurodevelopmental defects resulting from binge-like PAE.
Collapse
Affiliation(s)
- Megan Carugati
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States
| | - Charles R Goodlett
- Department of Psychology, Indiana University-Purdue University, Indianapolis, IN, 46202, United States
| | - Timothy A Cudd
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States
| | - Shannon E Washburn
- Department of Veterinary Physiology and Pharmacology and Michael E. DeBakey Institute, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, United States.
| |
Collapse
|
24
|
Prenatal and adolescent alcohol exposure programs immunity across the lifespan: CNS-mediated regulation. Pharmacol Biochem Behav 2022; 216:173390. [PMID: 35447157 DOI: 10.1016/j.pbb.2022.173390] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/28/2022] [Accepted: 04/11/2022] [Indexed: 12/31/2022]
Abstract
For many individuals, first exposure to alcohol occurs either prenatally due to maternal drinking, or during adolescence, when alcohol consumption is most likely to be initiated. Prenatal Alcohol Exposure (PAE) and its associated Fetal Alcohol Spectrum Disorders (FASD) in humans is associated with earlier initiation of alcohol use and increased rates of Alcohol Use Disorders (AUD). Initiation of alcohol use and misuse in early adolescence correlates highly with later AUD diagnosis as well. Thus, PAE and adolescent binge drinking set the stage for long-term health consequences due to adverse effects of alcohol on subsequent immune function, effects that may persist across the lifespan. The overarching goal of this review, therefore, is to determine the extent to which early developmental exposure to alcohol produces long-lasting, and potentially life-long, changes in immunological function. Alcohol affects the whole body, yet most studies are narrowly focused on individual features of immune function, largely ignoring the systems-level interactions required for effective host defense. We therefore emphasize the crucial role of the Central Nervous System (CNS) in orchestrating host defense processes. We argue that alcohol-mediated disruption of host immunity can occur through both (a) direct action of ethanol on neuroimmune processes, that subsequently disrupt peripheral immune function (top down); and (b) indirect action of ethanol on peripheral immune organs/cells, which in turn elicit consequent changes in CNS neuroimmune function (bottom up). Recognizing that alcohol consumption across the entire body, we argue in favor of integrative, whole-organism approaches toward understanding alcohol effects on immune function, and highlight the need for more work specifically examining long-lasting effects of early developmental exposure to alcohol (prenatal and adolescent periods) on host immunity.
Collapse
|
25
|
Gur M, Bendelac-Kapon L, Shabtai Y, Pillemer G, Fainsod A. Reduced Retinoic Acid Signaling During Gastrulation Induces Developmental Microcephaly. Front Cell Dev Biol 2022; 10:844619. [PMID: 35372345 PMCID: PMC8967241 DOI: 10.3389/fcell.2022.844619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid (RA) is a central signaling molecule regulating multiple developmental decisions during embryogenesis. Excess RA induces head malformations, primarily by expansion of posterior brain structures at the expense of anterior head regions, i.e., hindbrain expansion. Despite this extensively studied RA teratogenic effect, a number of syndromes exhibiting microcephaly, such as DiGeorge, Vitamin A Deficiency, Fetal Alcohol Syndrome, and others, have been attributed to reduced RA signaling. This causative link suggests a requirement for RA signaling during normal head development in all these syndromes. To characterize this novel RA function, we studied the involvement of RA in the early events leading to head formation in Xenopus embryos. This effect was mapped to the earliest RA biosynthesis in the embryo within the gastrula Spemann-Mangold organizer. Head malformations were observed when reduced RA signaling was induced in the endogenous Spemann-Mangold organizer and in the ectopic organizer of twinned embryos. Two embryonic retinaldehyde dehydrogenases, ALDH1A2 (RALDH2) and ALDH1A3 (RALDH3) are initially expressed in the organizer and subsequently mark the trunk and the migrating leading edge mesendoderm, respectively. Gene-specific knockdowns and CRISPR/Cas9 targeting show that RALDH3 is a key enzyme involved in RA production required for head formation. These observations indicate that in addition to the teratogenic effect of excess RA on head development, RA signaling also has a positive and required regulatory role in the early formation of the head during gastrula stages. These results identify a novel RA activity that concurs with its proposed reduction in syndromes exhibiting microcephaly.
Collapse
|
26
|
Gualdoni GS, Jacobo PV, Barril C, Ventureira MR, Cebral E. Early Abnormal Placentation and Evidence of Vascular Endothelial Growth Factor System Dysregulation at the Feto-Maternal Interface After Periconceptional Alcohol Consumption. Front Physiol 2022; 12:815760. [PMID: 35185604 PMCID: PMC8847216 DOI: 10.3389/fphys.2021.815760] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/28/2021] [Indexed: 01/16/2023] Open
Abstract
Adequate placentation, placental tissue remodeling and vascularization is essential for the success of gestation and optimal fetal growth. Recently, it was suggested that abnormal placenta induced by maternal alcohol consumption may participate in fetal growth restriction and relevant clinical manifestations of the Fetal Alcohol Spectrum Disorders (FASD). Particularly, periconceptional alcohol consumption up to early gestation can alter placentation and angiogenesis that persists in pregnancy beyond the exposure period. Experimental evidence suggests that abnormal placenta following maternal alcohol intake is associated with insufficient vascularization and defective trophoblast development, growth and function in early gestation. Accumulated data indicate that impaired vascular endothelial growth factor (VEGF) system, including their downstream effectors, the nitric oxide (NO) and metalloproteinases (MMPs), is a pivotal spatio-temporal altered mechanism underlying the early placental vascular alterations induced by maternal alcohol consumption. In this review we propose that the periconceptional alcohol intake up to early organogenesis (first trimester) alters the VEGF-NO-MMPs system in trophoblastic-decidual tissues, generating imbalances in the trophoblastic proliferation/apoptosis, insufficient trophoblastic development, differentiation and migration, deficient labyrinthine vascularization, and uncompleted remodelation and transformation of decidual spiral arterioles. Consequently, abnormal placenta with insufficiency blood perfusion, vasoconstriction and reduced labyrinthine blood exchange can be generated. Herein, we review emerging knowledge of abnormal placenta linked to pregnancy complications and FASD produced by gestational alcohol ingestion and provide evidence of the early abnormal placental angiogenesis-vascularization and growth associated to decidual-trophoblastic dysregulation of VEGF system after periconceptional alcohol consumption up to mid-gestation, in a mouse model.
Collapse
|
27
|
Brown J, Jonason A, Asp E, McGinn V, Carter MN, Spiller V, Jozan A. Fetal alcohol spectrum disorder and confabulation in psycholegal settings: A beginner's guide for criminal justice, forensic mental health, and legal interviewers. BEHAVIORAL SCIENCES & THE LAW 2022; 40:46-86. [PMID: 34689366 DOI: 10.1002/bsl.2540] [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: 04/08/2021] [Revised: 09/30/2021] [Accepted: 10/03/2021] [Indexed: 06/13/2023]
Abstract
Fetal alcohol spectrum disorders (FASD) are neurodevelopmental/neurobehavioral conditions caused by prenatal alcohol exposure (PAE). Impairments caused by PAE contribute to the over-representation of individuals with FASD in the United States juvenile and adult criminal justice systems. These same impairments can equally impact on individuals with FASD who are witnesses to or victims of crime who also have to navigate the complexities of the criminal justice system. Difficulties include increased susceptibility to confabulation throughout the legal process that, in turn, can contribute to increased rates of poor outcomes including false confessions and wrongful convictions. Individuals with FASD are particularity at risk of confabulation when they are subjected to tactics, such as stressful and anxiety-provoking situations, threats, and leading, suggestive, or coercive questioning. Many professionals in the forensic context are unfamiliar with FASD or related confabulation risk and may unintentionally utilize tactics that intensify impacts of pre-existing impairment. This article serves as a beginner's guide for professionals working in criminal justice settings by (a) providing research-based overviews of FASD and confabulation, (b) describing how FASD may lead to confabulation, and (c) suggesting ways that professionals can modify protocols when interacting with individuals with FASD. Suggestions in this article hold the potential to decrease the risk of confabulation in the criminal justice system and decrease problematic outcomes, such as false confessions and wrongful convictions among individuals with FASD.
Collapse
Affiliation(s)
- Jerrod Brown
- Pathways Counseling Center, Inc., St. Paul, Minnesota, USA
- Concordia University, St. Paul, Minnesota, USA
- American Institute for the Advancement of Forensic Studies, St. Paul, Minnesota, USA
| | - Alec Jonason
- Department of Psychology, Hamline University, St. Paul, Minnesota, USA
- Wesley & Lorene Artz Cognitive Neuroscience Research Center, Hamline University, St. Paul, Minnesota, USA
| | - Erik Asp
- Department of Psychology, Hamline University, St. Paul, Minnesota, USA
- Wesley & Lorene Artz Cognitive Neuroscience Research Center, Hamline University, St. Paul, Minnesota, USA
- Department of Neurology, University of Iowa, Iowa City, Iowa, USA
| | - Valerie McGinn
- The FASD Centre, Auckland, New Zealand
- School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Megan N Carter
- University of Washington, Seattle, Washington, USA
- Department of Social and Health Services, Special Commitment Center, Steilacoom, Washington, USA
| | | | - Amy Jozan
- American Institute for the Advancement of Forensic Studies, St. Paul, Minnesota, USA
| |
Collapse
|
28
|
Licheri V, Brigman JL. Altering Cell-Cell Interaction in Prenatal Alcohol Exposure Models: Insight on Cell-Adhesion Molecules During Brain Development. Front Mol Neurosci 2022; 14:753537. [PMID: 34975396 PMCID: PMC8715949 DOI: 10.3389/fnmol.2021.753537] [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: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Alcohol exposure during pregnancy disrupts the development of the brain and produces long lasting behavioral and cognitive impairments collectively known as Fetal Alcohol Spectrum Disorders (FASDs). FASDs are characterized by alterations in learning, working memory, social behavior and executive function. A large body of literature using preclinical prenatal alcohol exposure models reports alcohol-induced changes in architecture and activity in specific brain regions affecting cognition. While multiple putative mechanisms of alcohol’s long-lasting effects on morphology and behavior have been investigated, an area that has received less attention is the effect of alcohol on cell adhesion molecules (CAMs). The embryo/fetal development represents a crucial period for Central Nervous System (CNS) development during which the cell-cell interaction plays an important role. CAMs play a critical role in neuronal migration and differentiation, synaptic organization and function which may be disrupted by alcohol. In this review, we summarize the physiological structure and role of CAMs involved in brain development, review the current literature on prenatal alcohol exposure effects on CAM function in different experimental models and pinpoint areas needed for future study to better understand how CAMs may mediate the morphological, sensory and behavioral outcomes in FASDs.
Collapse
Affiliation(s)
- Valentina Licheri
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, United States.,New Mexico Alcohol Research Center, UNM Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
29
|
Abstract
The pathology of fetal alcohol syndrome and the less severe fetal alcohol spectrum disorders includes brain dysmyelination. Recent studies have shed light on the molecular mechanisms underlying these white matter abnormalities. Rodent models of fetal alcohol syndrome and human studies have shown suppressed oligodendrocyte differentiation and apoptosis of oligodendrocyte precursor cells. Ethanol exposure led to reduced expression of myelin basic protein and delayed myelin basic protein expression in rat and mouse models of fetal alcohol syndrome and in human histopathological specimens. Several studies have reported increased expression of many chemokines in dysmyelinating disorders in central nervous system, including multiple sclerosis and fetal alcohol syndrome. Acute ethanol exposure reduced levels of the neuroprotective insulin-like growth factor-1 in fetal and maternal sheep and in human fetal brain tissues, while ethanol increased the expression of tumor necrosis factor α in mouse and human neurons. White matter lesions have been induced in the developing sheep brain by alcohol exposure in early gestation. Rat fetal alcohol syndrome models have shown reduced axon diameters, with thinner myelin sheaths, as well as reduced numbers of oligodendrocytes, which were also morphologically aberrant oligodendrocytes. Expressions of markers for mature myelination, including myelin basic protein, also were reduced. The accumulating knowledge concerning the mechanisms of ethanol-induced dysmyelination could lead to the development of strategies to prevent dysmyelination in children exposed to ethanol during fetal development. Future studies using fetal oligodendrocyte- and oligodendrocyte precursor cell-derived exosomes isolated from the mother’s blood may identify biomarkers for fetal alcohol syndrome and even implicate epigenetic changes in early development that affect oligodendrocyte precursor cell and oligodendrocyte function in adulthood. By combining various imaging modalities with molecular studies, it may be possible to determine which fetuses are at risk and to intervene therapeutically early in the pregnancy.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
30
|
Chandran S, Sreeraj VS, Venkatasubramanian G, Sathyaprabha TN, Murthy P. Corpus callosum morphometry in children with prenatal alcohol exposure. Psychiatry Res Neuroimaging 2021; 318:111405. [PMID: 34743066 DOI: 10.1016/j.pscychresns.2021.111405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/19/2022]
Abstract
Alcohol is known to have a neurotoxic effect on the brain of offspring of mothers consuming alcohol during pregnancy. Impact on the neurodevelopment in children who were exposed to alcohol specifically during the antenatal period without any clinically detectable features of fetal alcohol syndrome is less well studied. In this cross-sectional study, structural magnetic resonance imaging (MRI) of the brain was acquired in 28 children whose mothers had consumed alcohol during pregnancy and 30 children of mothers who did not consume alcohol during pregnancy. Areas of Corpus callosum (CC) and its parts in the mid-sagittal section were calculated using morphometric analysis of MRI through Witelson's method. Midbody of CC was found to be significantly smaller in children exposed to alcohol during the prenatal period. CC is a sensitive white matter structure to neurotoxic effects of alcohol during prenatal life. This impact could be visible in developmental age even in those without any clinically detectable features of alcohol exposure.
Collapse
Affiliation(s)
- Sajish Chandran
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), 560061 Bengaluru India
| | - Vanteemar S Sreeraj
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru India
| | - Ganesan Venkatasubramanian
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru India
| | - Talakad N Sathyaprabha
- Department of Neurophysiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), 560061 Bengaluru India
| | - Pratima Murthy
- Department of Psychiatry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru India.
| |
Collapse
|
31
|
Bariselli S, Lovinger DM. Corticostriatal Circuit Models of Cognitive Impairments Induced by Fetal Exposure to Alcohol. Biol Psychiatry 2021; 90:516-528. [PMID: 34281711 PMCID: PMC8463431 DOI: 10.1016/j.biopsych.2021.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/13/2021] [Accepted: 05/16/2021] [Indexed: 12/26/2022]
Abstract
The term fetal alcohol spectrum disorder includes a group of diseases caused by fetal alcohol exposure (FAE). Patients with fetal alcohol spectrum disorder display heterogeneous socioemotional and cognitive deficits, particularly in the domain of executive function, that share symptoms with other neuropsychiatric disorders. Despite the availability of several preclinical models, the developmental brain defects causally linked to behavioral deficits induced by FAE remain poorly understood. Here, we first review the effects of FAE on corticostriatal development and its impact on both corticostriatal pathway function and cognitive abilities. We propose three non-mutually exclusive circuit models of corticostriatal dysfunctions to account for some of the FAE-induced cognitive deficits. One model posits that associative-sensorimotor imbalance causes hyper goal-directed behavior, and a second model implies that alteration of prefrontal-striatal behavioral suppression circuits results in loss of behavioral inhibition. A third model suggests that local striatal circuit deficits affect striatal neuronal ensemble function to impair action selection and performance. Finally, we discuss how preclinical approaches applied to these circuit models could offer potential rescue strategies for executive function deficits in patients with fetal alcohol spectrum disorder.
Collapse
Affiliation(s)
- Sebastiano Bariselli
- National Institute on Alcohol Abuse and Alcoholism (NIAAA), 5625 Fishers Lane, Bethesda, MD (20892-941),Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health (NIH), Bethesda, MD, United States
| | - David M. Lovinger
- National Institute on Alcohol Abuse and Alcoholism (NIAAA), 5625 Fishers Lane, Bethesda, MD (20892-941),Corresponding author:
| |
Collapse
|
32
|
Kruithof P, Ban S. A brief overview of fetal alcohol syndrome for health professionals. ACTA ACUST UNITED AC 2021; 30:890-893. [PMID: 34379462 DOI: 10.12968/bjon.2021.30.15.890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fetal alcohol syndrome (FAS) and fetal alcohol spectrum disorders (FASDs) are caused by prenatal alcohol exposure (PAE). They cause epigenetic changes, permanent neurodevelopmental deficits, and anomalies in growth and facial structure. This article enforces the need for health and social care professionals to have a greater understanding and awareness of how FAS and FASD may impact on the individual, the family and the community, to enable them to provide the most effective preventive and supportive care possible.
Collapse
Affiliation(s)
- Peter Kruithof
- Lecturer Global Health, School of Psychology, Faculty of Health Sciences and Wellbeing, Sunderland University, Sunderland
| | - Sasha Ban
- Senior Lecturer, Department of Nursing, Midwifery and Health, Northumbria University, Newcastle upon Tyne
| |
Collapse
|
33
|
Saha PS, Kim Sawtelle KR, Bamberg BN, Arrick DM, Watt MJ, Scholl JL, Zheng H, Mayhan WG. Rosiglitazone restores nitric oxide synthase-dependent reactivity of cerebral arterioles in rats exposed to prenatal alcohol. Alcohol Clin Exp Res 2021; 45:1359-1369. [PMID: 34120346 DOI: 10.1111/acer.14634] [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: 02/11/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Prenatal exposure to alcohol leads to a greater incidence of many cardiovascular-related diseases, presumably via a mechanism that may involve increased oxidative stress. An agonist of peroxisome proliferator-activated receptor gamma (PPARγ; rosiglitazone) has been shown to suppress alcohol-induced neuroinflammation and oxidative stress. The goal of this study was to determine whether acute and chronic treatment with rosiglitazone could restore or prevent impaired nitric oxide synthase (NOS)-dependent responses of cerebral arterioles in male and female adult (14-16 weeks old) rats exposed to alcohol in utero. METHODS We fed Sprague-Dawley dams a liquid diet with or without 3% ethanol for the duration of their pregnancy (21-23 days). In the first series of studies, we examined the reactivity of cerebral arterioles to eNOS- (ADP), nNOS-dependent (NMDA), and NOS-independent agonists in male and female adult rats before and during acute (1 hour) topical application of rosiglitazone (1 µM). In a second series of studies, we examined the influence of chronic treatment with rosiglitazone (3 mg/kg/day in drinking water for 2-3 weeks) on the responses of cerebral arterioles in male and female adult rats exposed to alcohol in utero. RESULTS We found that in utero exposure to alcohol similarly reduced responses of cerebral arterioles to ADP and NMDA, but not to nitroglycerin in male and female adult rats. In addition, acute treatment of the male and female adult rats with rosiglitazone similarly restored this impairment in cerebral vascular function to that observed in controls. We also found that chronic treatment with rosiglitazone prevented impaired vascular function in male and female adult rats that were exposed to alcohol in utero. CONCLUSIONS PPARγ activation may be an effective and relevant treatment to reverse or prevent cerebral vascular abnormalities associated with prenatal exposure to alcohol.
Collapse
Affiliation(s)
- Partha S Saha
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Kirsten R Kim Sawtelle
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Brittany N Bamberg
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Denise M Arrick
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Michael J Watt
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jamie L Scholl
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, USA
| |
Collapse
|
34
|
Pinheiro‐da‐Silva J, Luchiari AC. Embryonic ethanol exposure on zebrafish early development. Brain Behav 2021; 11:e02062. [PMID: 33939334 PMCID: PMC8213935 DOI: 10.1002/brb3.2062] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Embryonic exposure to ethanol leads to a condition of physical, behavioral, and cognitive deficiencies named fetal alcohol spectrum disorders (FASD). The most severe variations are in fetal alcohol syndrome (FAS), which is easier to diagnose and not studied in animal models. On the other side, the pFAS (partial fetal alcohol syndrome) includes cases of alcohol-related congenital disabilities and neurodevelopmental disorder with an inconclusive diagnosis. In recent years, the zebrafish has become a valuable model to study FASD and its variations. METHODS This study characterizes the zebrafish embryonic and larval development after low and moderate ethanol concentration exposure. Fish eggs were exposed to 0.0%, 0.25%, 0.5%, and 1.0% ethanol at 24 hr postfertilization, and embryonic development was observed every 8 hr up to 120 hpf. It evaluated movements, phenotypic abnormalities, hatching, cardiac function and heartbeat frequency, larvae length at 120 hpf, and the apoptotic cells' fluorescence stained with acridine orange. RESULTS Embryonic exposure to 0.5% and 1% ethanol presented reduced body size, decreased heartbeat rate, higher numbers of apoptotic cells, and hatching time differences. CONCLUSIONS Our results suggest any ethanol exposure during embryogenesis can be harmful and reinforces zebrafish as a suitable model for fetal alcohol spectrum disorders (FASD).
Collapse
Affiliation(s)
| | - Ana Carolina Luchiari
- Physiology and Behavior DepartmentFederal University of Rio Grande do NorteNatalBrazil
| |
Collapse
|
35
|
Pinheiro-da-Silva J, Araujo-Silva H, Luchiari AC. Does early ethanol exposure increase seeking-like behavior in zebrafish? Int J Dev Neurosci 2021; 81:416-427. [PMID: 33837569 DOI: 10.1002/jdn.10112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/16/2021] [Accepted: 04/05/2021] [Indexed: 01/22/2023] Open
Abstract
Fetal alcohol spectrum disorder (FASD) is the most common cause of birth defects. The severe variations are in fetal alcohol syndrome (FAS) but the most frequent cases are alcohol-related neurodevelopmental disorder (ARND), which is of a difficult diagnosis. ARND characteristics include impaired social behavior, anxiety and depression prevalence, cognitive deficits, and an increased chance for drug addiction. Here, we aimed to test whether early alcohol exposure leads to later alcohol preference. We hypothesize that early alcohol exposure increases the reinforcing effects on later experiences, raising the chance of addiction in adult life. Lately, the zebrafish has been a valuable model on alcohol research, allowing embryonic exposure and the study of the ontogenetic effects. For this, embryos were exposed to three different alcohol treatments: 0.0%, 0.25% and 0.5%, for 2 hr, at 24-hr post-fertilization. Then we evaluated the effects of embryonic alcohol exposure on conditioned place preference in two developmental stage: fry (10 days post-fertilization (dpf)) and young (90 dpf) zebrafish. Results show that control fish presented alcohol associative learning, which means, changes in place preference due to alcohol exposure, at both ontogenetic phases. However, zebrafish exposed to 0.25 and 0.5% alcohol during embryogenesis did not show conditioning response at any evaluated stage. These results suggest perception and cognitive deficits due to embryonic alcohol exposure that can alter alcohol responsiveness throughout a lifetime. Although low alcohol doses do not provoke malformation, it has been shown to induce several neurological and behavioral changes that are termed as Alcohol-Related Neurodevelopmental Disorders. These results may contribute to future investigations on how embryonic exposure affects the neurocircuitry related to perception and associative learning processing.
Collapse
Affiliation(s)
| | - Heloysa Araujo-Silva
- Departamento de Fisiologia e Comportamento, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Ana Carolina Luchiari
- Departamento de Fisiologia e Comportamento, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
36
|
Pyman P, Collins SE, Muggli E, Testa R, Anderson PJ. Cognitive and Behavioural Attention in Children with Low-Moderate and Heavy Doses of Prenatal Alcohol Exposure: a Systematic Review and Meta-analysis. Neuropsychol Rev 2021; 31:610-627. [PMID: 33656703 DOI: 10.1007/s11065-021-09490-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/18/2021] [Indexed: 01/02/2023]
Abstract
Attention problems are thought to be a hallmark feature of prenatal alcohol exposure (PAE). Despite decades of research however, these findings have never been pooled to assess the association between PAE and the different domains of attention functioning. This systematic review and meta-analysis aimed to examine the relationships between low-moderate, binge and heavy PAE with domains of attention functioning (encode, focus, shift, sustain and behavioural) in children. Thirteen studies compared children with PAE to abstinent controls. A significant adverse effect of any PAE on shifting attention (Cohen's d = -0.61), and a trend towards an adverse effect of heavy PAE on encoding attention (Cohen's d = -0.79) were identified. Compared to controls, there were trends showing that low-moderate PAE (odds ratio = 1.21) was associated with greater odds of behavioural attention problems. Remaining analyses were limited by insufficient studies or were non-significant. In summary, a vulnerability of higher-level attention skills to PAE was found. Urgent investigation into the effects of low to moderate PAE is needed given the prevalence of this drinking pattern, trends towards behavioural attention problems, the lack of comprehensive and high-quality research and the known impacts of attention difficulties on academic and social development in children.
Collapse
Affiliation(s)
- Philippa Pyman
- Monash University, Turner Institute for Brain and Mental Health, Melbourne, VIC, Australia. .,Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| | - Simonne E Collins
- Monash University, Turner Institute for Brain and Mental Health, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Evelyne Muggli
- Murdoch Children's Research Institute, Melbourne, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Renee Testa
- Department of Mental Health, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Peter J Anderson
- Monash University, Turner Institute for Brain and Mental Health, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Coker CR, Keller BN, Arnold AC, Silberman Y. Impact of High Fat Diet and Ethanol Consumption on Neurocircuitry Regulating Emotional Processing and Metabolic Function. Front Behav Neurosci 2021; 14:601111. [PMID: 33574742 PMCID: PMC7870708 DOI: 10.3389/fnbeh.2020.601111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/28/2020] [Indexed: 01/12/2023] Open
Abstract
The prevalence of psychiatry disorders such as anxiety and depression has steadily increased in recent years in the United States. This increased risk for anxiety and depression is associated with excess weight gain, which is often due to over-consumption of western diets that are typically high in fat, as well as with binge eating disorders, which often overlap with overweight and obesity outcomes. This finding suggests that diet, particularly diets high in fat, may have important consequences on the neurocircuitry regulating emotional processing as well as metabolic functions. Depression and anxiety disorders are also often comorbid with alcohol and substance use disorders. It is well-characterized that many of the neurocircuits that become dysregulated by overconsumption of high fat foods are also involved in drug and alcohol use disorders, suggesting overlapping central dysfunction may be involved. Emerging preclinical data suggest that high fat diets may be an important contributor to increased susceptibility of binge drug and ethanol intake in animal models, suggesting diet could be an important aspect in the etiology of substance use disorders. Neuroinflammation in pivotal brain regions modulating metabolic function, food intake, and binge-like behaviors, such as the hypothalamus, mesolimbic dopamine circuits, and amygdala, may be a critical link between diet, ethanol, metabolic dysfunction, and neuropsychiatric conditions. This brief review will provide an overview of behavioral and physiological changes elicited by both diets high in fat and ethanol consumption, as well as some of their potential effects on neurocircuitry regulating emotional processing and metabolic function.
Collapse
Affiliation(s)
- Caitlin R. Coker
- Biochemistry and Molecular & Cellular Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Bailey N. Keller
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Amy C. Arnold
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Yuval Silberman
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
38
|
da Silva Chagas L, Sandre PC, de Velasco PC, Marcondes H, Ribeiro E Ribeiro NCA, Barreto AL, Alves Mauro LB, Ferreira JH, Serfaty CA. Neuroinflammation and Brain Development: Possible Risk Factors in COVID-19-Infected Children. Neuroimmunomodulation 2021; 28:22-28. [PMID: 33530091 PMCID: PMC7900470 DOI: 10.1159/000512815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/01/2020] [Indexed: 11/19/2022] Open
Abstract
COVID-19, a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) betacoronavirus, affects children in a different way than it does in adults, with milder symptoms. However, several cases of neurological symptoms with neuroinflammatory syndromes, such as the multisystem inflammatory syndrome (MIS-C), following mild cases, have been reported. As with other viral infections, such as rubella, influenza, and cytomegalovirus, SARS-CoV-2 induces a surge of proinflammatory cytokines that affect microglial function, which can be harmful to brain development. Along with the viral induction of neuroinflammation, other noninfectious conditions may interact to produce additional inflammation, such as the nutritional imbalance of fatty acids and polyunsaturated fatty acids and alcohol consumption during pregnancy. Additionally, transient thyrotoxicosis induced by SARS-CoV-2 with secondary autoimmune hypothyroidism has been reported, which could go undetected during pregnancy. Together, those factors may pose additional risk factors for SARS-CoV-2 infection impacting mechanisms of neural development such as synaptic pruning and neural circuitry formation. The present review discusses those conditions in the perspective of the understanding of risk factors that should be considered and the possible emergence of neurodevelopmental disorders in COVID-19-infected children.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Poliana Capucho Sandre
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Patricia Coelho de Velasco
- Department of Applied Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Henrique Marcondes
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | | | - Aline Loureiro Barreto
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Luiza Beatriz Alves Mauro
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Julia Huber Ferreira
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Claudio A Serfaty
- Laboratory of Neural Plasticity, Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil,
- National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil,
| |
Collapse
|
39
|
Ståhlberg T, Khanal P, Chudal R, Luntamo T, Kronström K, Sourander A. Prenatal and perinatal risk factors for anxiety disorders among children and adolescents: A systematic review. J Affect Disord 2020; 277:85-93. [PMID: 32799108 DOI: 10.1016/j.jad.2020.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Prenatal and perinatal risk factors for anxiety disorders have rarely been studied, even though they are highly prevalent in children and adolescents. It is important to identify the common risk factors, so that targeted preventive care and early interventions can be provided. METHODS A systematic review of the PubMed and PsycInfo databases was conducted to 25 October 2019, according to the Preferred Reporting Items of Systematic Reviews and Meta-analyses guidelines. The protocol was registered on the Prospective Register of Systematic Reviews and the quality assessment was carried out using the Joanna Briggs tools. RESULTS The review identified 31 studies from eight countries, including three register studies. Cohort sizes ranged from 69 to 89,404 and diagnoses cases ranged from 4 to 7867. Although various risk factors had been researched, only few of them had been repeatedly studied and the findings were highly inconsistent. The associations between the different risk factors and anxiety disorders seemed weak compared to many other psychiatric disorders, but preterm birth and maternal somatic illnesses may increase the risk for anxiety disorders in offspring. LIMITATIONS The studies varied considerably by study design, risk factors and anxiety disorders studied, sample sizes and follow up periods. CONCLUSIONS Prenatal and perinatal risk factors for anxiety disorders have been under-researched, compared to other psychiatric disorders. Our systematic review found weak links to prenatal events, but flagged up preterm birth and maternal somatic illnesses as possible avenues for future research.
Collapse
Affiliation(s)
- Tiia Ståhlberg
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland.
| | - Prakash Khanal
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland
| | - Roshan Chudal
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland
| | - Terhi Luntamo
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland
| | - Kim Kronström
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland; Department of Adolescent Psychiatry, Turku University Hospital, Turku, Finland
| | - Andre Sourander
- Research Centre for Child Psychiatry, University of Turku, Lemminkäisenkatu 3/Teutori 3. floor, 20014 Turku, Finland; Turku University Hospital, Turku, Finland; INVEST Research Flagship, University of Turku, (Principal Investigator), Turku, Finland
| |
Collapse
|
40
|
Aglawe MM, Kale MB, Rahangdale SR, Kotagale NR, Umekar MJ, Taksande BG. Agmatine improves the behavioral and cognitive impairments associated with chronic gestational ethanol exposure in rats. Brain Res Bull 2020; 167:37-47. [PMID: 33242522 DOI: 10.1016/j.brainresbull.2020.11.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/28/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022]
Abstract
Chronic maternal ethanol exposure leads to poor intelligence, impaired cognition and array of neurological symptoms in offsprings and commonly referred as fetal alcohol spectrum disorder (FASD). Despite high prevalence and severity, the neurochemical basis of FASD remains largely unexplored. The present study evaluated the pharmacological effects of agmatine in cognitive deficits associated with FAS in rat's offsprings prenatally exposed to alcohol. Pregnant rats received ethanol in liquid modified diet during the entire gestational period of 21 days. Offsprings were treated with agmatine (20-80 mg/Kg, i.p.) during early postnatal days (PND: 21-35) and subsequently evaluated for anxiety in elevated plus maze (EPM), depression in forced swim test (FST) and learning and memory in Morris's water maze (MWM) during post adolescent phase. Hippocampal agmatine, BDNF, TNF-α and IL-6 levels were also analyzed in prenatally ethanol exposed pups. Offsprings prenatally exposed to ethanol demonstrated delayed righting reflex, reduced exploratory behavior along with anxiety, depression-like behavior and impaired memory. These behavioral abnormalities were correlated with a significant reduction in hippocampal agmatine and BDNF levels and elevation in TNF-α and IL-6 immunocontent. Chronic agmatine (40 and 80 mg/Kg, i.p.) administration for 15 days (PND: 21-35), improved entries and time spent in open arm of EPM, decreased immobility time in FST. It also reduced latency to reach the platform location; increased the number of entries, time spent in platform quadrant and also number of crossing over platform quadrant when subjected to MWM test in prenatally ethanol exposed offsprings. This study provides functional evidences for the therapeutic potential of agmatine in cognitive impairment and other neurological complications associated with FASD.
Collapse
Affiliation(s)
- Manish M Aglawe
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra, 441 002, India
| | - Mayur B Kale
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra, 441 002, India
| | - Sandip R Rahangdale
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra, 441 002, India
| | | | - Milind J Umekar
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra, 441 002, India
| | - Brijesh G Taksande
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, Maharashtra, 441 002, India.
| |
Collapse
|
41
|
Darbinian N, Darbinyan A, Merabova N, Bajwa A, Tatevosian G, Martirosyan D, Zhao H, Selzer ME, Goetzl L. Ethanol-mediated alterations in oligodendrocyte differentiation in the developing brain. Neurobiol Dis 2020; 148:105181. [PMID: 33189883 DOI: 10.1016/j.nbd.2020.105181] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/21/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Alterations of white matter integrity and subsequent white matter structural deficits are consistent findings in Fetal Alcohol Syndrome (FAS), but knowledge regarding the molecular mechanisms underlying these abnormalities is incomplete. Experimental rodent models of FAS have shown dysregulation of cytokine expression leading to apoptosis of oligodendrocyte precursor cells (OPCs) and altered oligodendrocyte (OL) differentiation, but whether this is representative of human FAS pathogenesis has not been determined. METHODS Fetal brain tissue (12.2-21.4 weeks gestation) from subjects undergoing elective termination of pregnancy was collected according to an IRB-approved protocol. Ethanol (EtOH) exposure status was classified based on a detailed face-to-face questionnaire adapted from the National Institute on Alcohol Abuse and Alcoholism Prenatal Alcohol and Sudden Infant Death Syndrome and Stillbirth (PASS) study. Twenty EtOH-exposed fetuses were compared with 20 gestational age matched controls. Cytokine and OPC marker mRNA expression was quantified by Real-Time Polymerase chain reaction (qRT-PCR). Patterns of protein expression of OPC markers and active Capase-3 were studied by Fluorescence Activated Cell Sorting (FACS). RESULTS EtOH exposure was associated with reduced markers of cell viability, OPC differentiation, and OL maturation, while early OL differentiation markers were unchanged or increased. Expression of mRNAs for proteins specific to more mature forms of OL lineage (platelet-derived growth factor α (PDGFRα) and myelin basic protein (MBP) was lower in the EtOH group than in controls. Expression of the multifunctional growth and differentiation-promoting growth factor IGF-1, which is essential for normal development, also was reduced. Reductions were not observed for markers of early stages of OL differentiation, including Nuclear transcription factor NK-2 homeobox locus 2 (Nkx2.2). Expression of mRNAs for the proinflammatory cytokine, tumor necrosis factor-α (TNFα), and several proinflammatory chemokines was higher in the EtOH group compared to controls, including: Growth regulated protein alpha/chemokine (C-X-C motif) ligand 1 (GRO-α/CXCL1), Interleukin 8/chemokine (C-X-C motif) ligand 8 (IL8/CXCL8), Chemokine (C-X-C motif) ligand 6/Granulocyte chemotactic protein 2 (CXCL16/GCP2), epithelial-derived neutrophil-activating protein 78/chemokine (C-X-C motif) ligand 5 (ENA-78/CXCL5), monocyte chemoattractant protein-1 (MCP-1). EtOH exposure also was associated with an increase in the proportion of cells expressing markers of early stage OPCs, such as A2B5 and NG2. Finally, apoptosis (measured by caspase-3 activation) was increased substantially in the EtOH group compared to controls. CONCLUSION Prenatal EtOH exposure is associated with excessive OL apoptosis and/or delayed OL maturation in human fetal brain. This is accompanied by markedly dysregulated expression of several chemokines and cytokines, in a pattern predictive of increased OL cytotoxicity and reduced OL differentiation. These findings are consistent with findings in animal models of FAS.
Collapse
Affiliation(s)
- Nune Darbinian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Armine Darbinyan
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, United States of America.
| | - Nana Merabova
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Ahsun Bajwa
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Gabriel Tatevosian
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Diana Martirosyan
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Huaqing Zhao
- Department of Clinical Sciences (Biostatistics and Epidemiology), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Michael E Selzer
- Center for Neural Repair and Rehabilitation (Shriners Hospitals Pediatric Research Center), Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States of America.
| | - Laura Goetzl
- Department of Obstetrics & Gynecology, University of Texas, Houston, TX 77030, United States of America.
| |
Collapse
|
42
|
Raghunathan R, Liu CH, Kouka A, Singh M, Miranda RC, Larin KV. Dose-response analysis of microvasculature changes in the murine fetal brain and the maternal extremities due to prenatal ethanol exposure. JOURNAL OF BIOMEDICAL OPTICS 2020; 25:JBO-200176RR. [PMID: 33244919 PMCID: PMC7689263 DOI: 10.1117/1.jbo.25.12.126001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 11/06/2020] [Indexed: 05/29/2023]
Abstract
SIGNIFICANCE Prenatal exposure to ethanol causes several morphological and neurobehavioral deficits. While there are some studies on the effects of ethanol exposure on blood flow, research focusing on acute changes in the microvasculature is limited. AIM The first aim of this study was to assess the dose-dependent changes in murine fetal brain microvasculature of developing fetuses in response to maternal alcohol consumption. The second aim was to quantify changes in vasculature occurring concurrently in the mother's hindlimb and the fetus's brain after maternal exposure to alcohol. APPROACH Correlation mapping optical coherence angiography was used to evaluate the effects of prenatal exposure to different doses of ethanol (3, 1.5, and 0.75 g / kg) on murine fetal brain vasculature in utero. Additionally, simultaneous imaging of maternal peripheral vessels and the fetal brain vasculature was performed to assess changes of the vasculature occurring concurrently in response to ethanol consumption. RESULTS The fetal brain vessel diameters (VDs) decreased by ∼47 % , 30%, and 14% in response to ethanol doses of 3, 1.5, and 0.75 g / kg, respectively. However, the mother's hindlimb VD increased by 63% in response to ethanol at a dose of 3 g / kg. CONCLUSIONS Results showed a dose-dependent reduction in vascular blood flow in fetal brain vessels when the mother was exposed to ethanol, whereas vessels in the maternal hindlimb exhibited concurrent vasodilation.
Collapse
Affiliation(s)
- Raksha Raghunathan
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Chih-Hao Liu
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Amur Kouka
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Rajesh C. Miranda
- Texas A&M University Health Science Center College of Medicine, Department of Neuroscience and Experimental Therapeutics, Bryan, Texas, United States
| | - Kirill V. Larin
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| |
Collapse
|
43
|
Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci Biobehav Rev 2020; 118:654-668. [PMID: 32976915 DOI: 10.1016/j.neubiorev.2020.08.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/18/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Ethanol exposure during gestation is an early life stressor that profoundly dysregulates structure and functions of the embryonal nervous system, altering the cognitive and behavioral development. Such dysregulation is also achieved by epigenetic mechanisms, which, altering the chromatin structure, redraw the entire pattern of gene expression. In parallel, an oxidative stress response at the cellular level and a global upregulation of neuroendocrine stress response, regulated by the HPA axis, exist and persist in adulthood. This neurobehavioral framework matches those observed in other psychiatric diseases such as mood diseases, depression, autism; those early life stressing events, although probably triggered by specific and different epigenetic mechanisms, give rise to largely overlapping neurobehavioral phenotypes. An early diagnosis of prenatal alcohol exposure, using reliable markers of ethanol intake, together with a deeper understanding of the pathogenic mechanisms, some of them reversible by their nature, can offer a temporal "window" of intervention. Supplementing a mother's diet with protective and antioxidant substances in addition to supportive psychological therapies can protect newborns from being affected.
Collapse
|
44
|
van Adrichem DS, Huijbregts SCJ, Van der Heijden KB, van Goozen SHM, Swaab H. Prenatal risk and physical aggression during the first years of life: The gender-specific role of inhibitory control. INFANCY 2020; 24:807-826. [PMID: 32677274 DOI: 10.1111/infa.12307] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 12/14/2018] [Accepted: 05/18/2019] [Indexed: 01/20/2023]
Abstract
Prenatal risk and a lack of inhibitory control have consistently been related to the development of physical aggression in older children. This study examined whether inhibitory control mediated the relation between prenatal risk and aggression in infants and toddlers. The role of gender in this mediation model was also examined. The sample consisted of 161 mother-child dyads (83 boys). A prenatal cumulative risk score was created from a number of well-established risk factors including maternal psychopathology, substance use, and social and socioeconomic disadvantages. At 12 months, children performed an inhibitory control task. Physical aggression was assessed through maternal reports at 12 and 20 months of age. Results showed that higher prenatal risk was associated with more physical aggression. Inhibitory control mediated this association at both 12 and 20 months: higher prenatal risk was related to lower inhibitory control, which in turn led to higher aggression. At 20 months, gender moderated the mediation effect: the mediating role of inhibitory control was only found for girls. These results suggest that even before 2 years of age, inhibitory control is an important construct involved in the relation between prenatal risk and physical aggression.
Collapse
Affiliation(s)
- Dide S van Adrichem
- Clinical Neurodevelopmental Sciences, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | - Stephan C J Huijbregts
- Clinical Neurodevelopmental Sciences, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | - Kristiaan B Van der Heijden
- Clinical Neurodevelopmental Sciences, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | - Stephanie H M van Goozen
- Clinical Neurodevelopmental Sciences, Leiden University, Leiden, The Netherlands.,School of Psychology, Cardiff University, Cardiff, UK
| | - Hanna Swaab
- Clinical Neurodevelopmental Sciences, Leiden University, Leiden, The Netherlands.,Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| |
Collapse
|
45
|
Almeida L, Andreu-Fernández V, Navarro-Tapia E, Aras-López R, Serra-Delgado M, Martínez L, García-Algar O, Gómez-Roig MD. Murine Models for the Study of Fetal Alcohol Spectrum Disorders: An Overview. Front Pediatr 2020; 8:359. [PMID: 32760684 PMCID: PMC7373736 DOI: 10.3389/fped.2020.00359] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure is associated to different physical, behavioral, cognitive, and neurological impairments collectively known as fetal alcohol spectrum disorder. The underlying mechanisms of ethanol toxicity are not completely understood. Experimental studies during human pregnancy to identify new diagnostic biomarkers are difficult to carry out beyond genetic or epigenetic analyses in biological matrices. Therefore, animal models are a useful tool to study the teratogenic effects of alcohol on the central nervous system and analyze the benefits of promising therapies. Animal models of alcohol spectrum disorder allow the analysis of key variables such as amount, timing and frequency of ethanol consumption to describe the harmful effects of prenatal alcohol exposure. In this review, we aim to synthetize neurodevelopmental disabilities in rodent fetal alcohol spectrum disorder phenotypes, considering facial dysmorphology and fetal growth restriction. We examine the different neurodevelopmental stages based on the most consistently implicated epigenetic mechanisms, cell types and molecular pathways, and assess the advantages and disadvantages of murine models in the study of fetal alcohol spectrum disorder, the different routes of alcohol administration, and alcohol consumption patterns applied to rodents. Finally, we analyze a wide range of phenotypic features to identify fetal alcohol spectrum disorder phenotypes in murine models, exploring facial dysmorphology, neurodevelopmental deficits, and growth restriction, as well as the methodologies used to evaluate behavioral and anatomical alterations produced by prenatal alcohol exposure in rodents.
Collapse
Affiliation(s)
- Laura Almeida
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Fundació Sant Joan de Déu, Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| | - Vicente Andreu-Fernández
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Nutrition and Health Deparment, Valencian International University (VIU), Valencia, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elisabet Navarro-Tapia
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rosa Aras-López
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain
| | - Mariona Serra-Delgado
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| | - Leopoldo Martínez
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Congenital Malformations Lab, Institute of Medicine and Molecular Genetic (INGEMM), Institute for Health Research of La Paz Universitary Hospital (IdiPAZ), Madrid, Spain
- Department of Pediatric Surgery, Hospital Universitario La Paz, Madrid, Spain
| | - Oscar García-Algar
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Grup de Recerca Infancia i Entorn (GRIE), Institut D'investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Neonatology, Hospital Clínic-Maternitat, ICGON, IDIBAPS, BCNatal, Barcelona, Spain
| | - María Dolores Gómez-Roig
- Maternal and Child Health and Development Network II (SAMID II), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Fundació Sant Joan de Déu, Barcelona, Spain
- BCNatal Barcelona Center for Maternal Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, Barcelona, Spain
| |
Collapse
|
46
|
Doremus-Fitzwater TL, Youngentob SL, Youngentob L, Gano A, Vore AS, Deak T. Lingering Effects of Prenatal Alcohol Exposure on Basal and Ethanol-Evoked Expression of Inflammatory-Related Genes in the CNS of Adolescent and Adult Rats. Front Behav Neurosci 2020; 14:82. [PMID: 32714160 PMCID: PMC7344178 DOI: 10.3389/fnbeh.2020.00082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/30/2020] [Indexed: 12/16/2022] Open
Abstract
Emerging data suggest that alcohol's effects on central inflammatory factors are not uniform across the lifespan. In particular, prenatal alcohol exposure (PAE) significantly alters steady-state levels of neuroimmune factors, as well as subsequent reactivity to later immune challenge. Thus, the current experiment investigated developmental sensitivities to, and long-lasting consequences of, PAE on ethanol-evoked cytokine expression in male and female adolescent and adult rats. Pregnant dams received either an ad libitum ethanol liquid diet (2.2% GD 6-8; 4.5% GD 9-10; 6.7% GD11-20; 35% daily calories from ethanol) or free-choice access to a control liquid diet and water. At birth, offspring were fostered to dams given free-choice access to the control liquid diet. Pups then matured until mid-adolescence [postnatal day (PD) 35] or adulthood (PD90), at which time they were challenged with either a binge-like dose of ethanol (4 g/kg; intragastrically) or tap water. During intoxication (3 h post-ethanol challenge), brains and blood were collected for assessment of neuroimmune gene expression (reverse transcription-polymerase chain reaction; RT-PCR) in the hippocampus, amygdala, and PVN, as well as for blood ethanol concentrations (BEC) and plasma corticosterone levels. Results revealed that rats challenged with ethanol at either PD35 or PD90 generally exhibited a characteristic cytokine signature of acute intoxication that we have previously reported: increased Il-6 and IkBα expression, with decreased Il-1β and Tnfα gene expression. With a few exceptions, this pattern of gene changes was observed in all three structures examined, at both ages of postnatal ethanol challenge, and in both sexes. While few significant effects of PAE were observed for ethanol-induced alterations in cytokine expression, there was a consistent (but nonsignificant) trend for PAE to potentiate the expression of Il-6 and IkBα in all groups except adult females. Although these data suggest that later-life ethanol challenge was a far greater driver of inflammatory signaling than PAE, the current results demonstrate PAE resulted in subtle long-term alterations in the expression of many key neuroinflammatory factors associated with NF-κB signaling. Such long-lasting impacts of PAE that may engender vulnerability to later environmental events triggering neuroinflammatory processes, such as chronic ethanol exposure or stress, could contribute to heightened vulnerability for PAE-related alterations and deficits.
Collapse
Affiliation(s)
- Tamara L. Doremus-Fitzwater
- Department of Psychology, Ithaca College, Ithaca, NY, United States
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States
| | - Steven L. Youngentob
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States
- University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| | - Lisa Youngentob
- University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| | - Anny Gano
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Andrew S. Vore
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| | - Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States
- Department of Psychology, Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
47
|
Tenenbaum A, Mandel A, Dor T, Sapir A, Sapir-Bodnaro O, Hertz P, Wexler ID. Fetal alcohol spectrum disorder among pre-adopted and foster children. BMC Pediatr 2020; 20:275. [PMID: 32493264 PMCID: PMC7271511 DOI: 10.1186/s12887-020-02164-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 05/20/2020] [Indexed: 11/10/2022] Open
Abstract
Background Fetal alcohol spectrum disorder (FASD) is a leading cause of neurodevelopmental disorders. Children in foster care or domestically adopted are at greater risk for FASD. The aim of this study was to determine the prevalence or risk for FASD in a selected population of foster and adopted children. Methods Children between 2 and 12 years who were candidates for adoption in foster care were evaluated for clinical manifestations and historical features of fetal alcohol spectrum disorder based on established criteria for FASD. Results Of the 89 children evaluated, 18 had mothers with a confirmed history of alcohol consumption during pregnancy. Two children had fetal alcohol syndrome and one had partial fetal alcohol syndrome. In addition, five had alcohol-related neurodevelopmental disorder, one had alcohol-related birth defects, and a single child had manifestations of both. Of the 71 children in which fetal alcohol exposure could not be confirmed, many had manifestations that would have established a diagnosis of FASD were a history of maternal alcohol consumption obtained. Conclusions In a population of high-risk children seen in an adoption clinic, many had manifestations associated with FASD especially where prenatal alcohol exposure was established. The reported prevalence in this study is higher than that reported in our previous study of younger children. This is most likely due to the higher number of children diagnosed with alcohol-related neurodevelopmental disorders that typically manifest at an older age.
Collapse
Affiliation(s)
- Ariel Tenenbaum
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel.
| | - Asaf Mandel
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| | - Talia Dor
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| | - Alon Sapir
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| | - Orly Sapir-Bodnaro
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| | - Pnina Hertz
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| | - Isaiah D Wexler
- Medical Unit for Adoption and Foster Care, Department of Pediatrics, Hadassah University Medical Center, Mount Scopus Campus, 92140, Jerusalem, IL, Israel
| |
Collapse
|
48
|
Reid HMO, Lysenko-Martin MR, Snowden TM, Thomas JD, Christie BR. A Systematic Review of the Effects of Perinatal Alcohol Exposure and Perinatal Marijuana Exposure on Adult Neurogenesis in the Dentate Gyrus. Alcohol Clin Exp Res 2020; 44:1164-1174. [PMID: 32246781 PMCID: PMC7905844 DOI: 10.1111/acer.14332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Marijuana and alcohol are both substances that, when used during pregnancy, may have profound effects on the developing fetus. There is evidence to suggest that both drugs have the capacity to affect working memory, one function of the hippocampal formation; however, there is a paucity of data on how perinatal exposure to alcohol or cannabis impacts the process of adult neurogenesis. METHODS This systematic review examines immunohistochemical data from adult rat and mouse models that assess perinatal alcohol or perinatal marijuana exposure. A comprehensive list of search terms was designed and used to search 3 separate databases. All results were imported to Mendeley and screened by 2 authors. Consensus was reached on a set of final papers that met the inclusion criteria, and their results were summarized. RESULTS Twelve papers were identified as relevant, 10 of which pertained to the effects of perinatal alcohol on the adult hippocampus, and 2 pertained to the effects of perinatal marijuana on the adult hippocampus. Cellular proliferation in the dentate gyrus was not affected in adult rats and mice exposed to alcohol perinatally. In general, perinatal alcohol exposure did not have a significant and reliable effect on the maturation and survival of adult born granule neurons in the dentate gyrus. In contrast, interneuron numbers appear to be reduced in the dentate gyrus of adult rats and mice exposed perinatally to alcohol. Perinatal marijuana exposure was also found to reduce inhibitory interneuron numbers in the dentate gyrus. CONCLUSIONS Perinatal alcohol exposure and perinatal marijuana exposure both act on inhibitory interneurons in the hippocampal formation of adult rats. These findings suggest simultaneous perinatal alcohol and marijuana exposure (SAM) may have a dramatic impact on inhibitory processes in the dentate gyrus.
Collapse
Affiliation(s)
- Hannah M O Reid
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Melanie R Lysenko-Martin
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Taylor M Snowden
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
| | - Jennifer D Thomas
- Center for Behavioral Teratology, (JDT), San Diego State University, San Diego, California
| | - Brian R Christie
- From the, Division of Medical Sciences, (HMOR, MRL, TMS, BRC), University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, (BRC), University of British Columbia, Victoria, British Columbia
| |
Collapse
|
49
|
Saito M, Smiley JF, Hui M, Masiello K, Betz J, Ilina M, Saito M, Wilson DA. Neonatal Ethanol Disturbs the Normal Maturation of Parvalbumin Interneurons Surrounded by Subsets of Perineuronal Nets in the Cerebral Cortex: Partial Reversal by Lithium. Cereb Cortex 2020; 29:1383-1397. [PMID: 29462278 DOI: 10.1093/cercor/bhy034] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/02/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023] Open
Abstract
Reduction in parvalbumin-positive (PV+) interneurons is observed in adult mice exposed to ethanol at postnatal day 7 (P7), a late gestation fetal alcohol spectrum disorder model. To evaluate whether PV+ cells are lost, or PV expression is reduced, we quantified PV+ and associated perineuronal net (PNN)+ cell densities in barrel cortex. While PNN+ cell density was not reduced by P7 ethanol, PV cell density decreased by 25% at P90 with no decrease at P14. PNN+ cells in controls were virtually all PV+, whereas more than 20% lacked PV in ethanol-treated adult animals. P7 ethanol caused immediate apoptosis in 10% of GFP+ cells in G42 mice, which express GFP in a subset of PV+ cells, and GFP+ cell density decreased by 60% at P90 without reduction at P14. The ethanol effect on PV+ cell density was attenuated by lithium treatment at P7 or at P14-28. Thus, reduced PV+ cell density may be caused by disrupted cell maturation, in addition to acute apoptosis. This effect may be regionally specific: in the dentate gyrus, P7 ethanol reduced PV+ cell density by 70% at P14 and both PV+ and PNN+ cell densities by 50% at P90, and delayed lithium did not alleviate ethanol's effect.
Collapse
Affiliation(s)
- Mariko Saito
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - John F Smiley
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, NYU School of Medicine, New York, NY, USA
| | - Maria Hui
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Kurt Masiello
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Judith Betz
- Division of Neurochemistry, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Maria Ilina
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Mitsuo Saito
- Department of Psychiatry, NYU School of Medicine, New York, NY, USA.,Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Donald A Wilson
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
50
|
Easey KE, Timpson NJ, Munafò MR. Association of Prenatal Alcohol Exposure and Offspring Depression: A Negative Control Analysis of Maternal and Partner Consumption. Alcohol Clin Exp Res 2020; 44:1132-1140. [PMID: 32315093 PMCID: PMC7341445 DOI: 10.1111/acer.14324] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/04/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previous research has suggested that intrauterine alcohol exposure is associated with a variety of adverse outcomes in offspring. However, few studies have investigated its association with offspring internalizing disorders in late adolescence. METHODS Using data from the Avon Longitudinal Study of Parents and Children (ALSPAC), we investigated the associations of maternal drinking in pregnancy with offspring depression at age 18 and 24 (n = 13,480). We also examined partner drinking as a negative control for intrauterine exposure for comparison. RESULTS Offspring of mothers that consumed any alcohol at 18 weeks gestation were at increased risk of having a diagnosis of depression (fully adjusted model: OR 1.17, 95% CI 1.02 to 1.34), but there was no clear evidence of association between partners' alcohol consumption at 18 weeks gestation during pregnancy and increased risk of offspring depression (fully adjusted model: OR 0.87, 95% CI 0.74 to 1.01). Postestimation tests found a positive difference between the association of maternal and partner alcohol use on offspring depression, showing a stronger association for maternal compared with partner alcohol use (OR 1.41, CI 1.07 to 1.84). CONCLUSIONS Maternal drinking in pregnancy was associated with increased risk of offspring depression at age 18. Residual confounding may explain this association, but the negative control comparison of paternal drinking provides some evidence that it may be causal, and this warrants further investigation.
Collapse
Affiliation(s)
- Kayleigh E. Easey
- UK Centre for Tobacco and Alcohol StudiesSchool of Psychological ScienceUniversity of BristolBristolUK
- MRC Integrative Epidemiology UnitUniversity of BristolBristolUK
- Bristol Medical SchoolUniversity of BristolBristolUK
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology UnitUniversity of BristolBristolUK
- Bristol Medical SchoolUniversity of BristolBristolUK
| | - Marcus R. Munafò
- UK Centre for Tobacco and Alcohol StudiesSchool of Psychological ScienceUniversity of BristolBristolUK
- MRC Integrative Epidemiology UnitUniversity of BristolBristolUK
| |
Collapse
|